Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
ACS Synth Biol ; 13(4): 1077-1084, 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38588591

RESUMEN

Uremic toxins (UTs) are microbiota-derived metabolites that accelerate the progression of kidney damage in patients with chronic kidney disease (CKD). One of the major UTs involved in CKD progression is p-cresol-sulfate (PCS), derived from dietary l-tyrosine (l-Tyr). Here, we engineered a probiotic strain of Escherichia coli Nissle 1917, to convert l-Tyr to the nontoxic compound p-coumaric acid via tyrosine ammonia lyase (TAL). First, a small metagenomic library was assessed to identify the TAL with the greatest whole-cell activity. Second, accessory genes implicated in the import of l-Tyr and export of PCA were overexpressed to enhance l-Tyr degradation by 106% and 56%, respectively. Last, random mutagenesis coupled to a novel selection and screening strategy was developed that identified a TAL variant with a 25% increase in whole-cell activity. Taken together, the final strain exhibits a 183% improvement over initial whole-cell activity and provides a promising candidate to degrade l-Tyr mediated PCS accumulation.


Asunto(s)
Escherichia coli , Insuficiencia Renal Crónica , Humanos , Escherichia coli/genética , Escherichia coli/metabolismo , Tóxinas Urémicas , Mutagénesis , Insuficiencia Renal Crónica/tratamiento farmacológico , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/metabolismo
2.
Cell Host Microbe ; 32(3): 382-395.e10, 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38309259

RESUMEN

Methionine is an essential proteinogenic amino acid, but its excess can lead to deleterious effects. Inborn errors of methionine metabolism resulting from loss of function in cystathionine ß-synthase (CBS) cause classic homocystinuria (HCU), which is managed by a methionine-restricted diet. Synthetic biotics are gastrointestinal tract-targeted live biotherapeutics that can be engineered to replicate the benefits of dietary restriction. In this study, we assess whether SYNB1353, an E. coli Nissle 1917 derivative, impacts circulating methionine and homocysteine levels in animals and healthy volunteers. In both mice and nonhuman primates (NHPs), SYNB1353 blunts the appearance of plasma methionine and plasma homocysteine in response to an oral methionine load. A phase 1 clinical study conducted in healthy volunteers subjected to an oral methionine challenge demonstrates that SYNB1353 is well tolerated and blunts plasma methionine by 26%. Overall, SYNB1353 represents a promising approach for methionine reduction with potential utility for the treatment of HCU.


Asunto(s)
Homocistinuria , Metionina , Humanos , Ratones , Animales , Metionina/metabolismo , Metionina/uso terapéutico , Voluntarios Sanos , Escherichia coli/genética , Escherichia coli/metabolismo , Modelos Animales de Enfermedad , Homocistinuria/tratamiento farmacológico , Homocistinuria/metabolismo , Racemetionina , Homocisteína/uso terapéutico
3.
Cell Syst ; 14(6): 512-524.e12, 2023 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-37348465

RESUMEN

To build therapeutic strains, Escherichia coli Nissle (EcN) have been engineered to express antibiotics, toxin-degrading enzymes, immunoregulators, and anti-cancer chemotherapies. For efficacy, the recombinant genes need to be highly expressed, but this imposes a burden on the cell, and plasmids are difficult to maintain in the body. To address these problems, we have developed landing pads in the EcN genome and genetic circuits to control therapeutic gene expression. These tools were applied to EcN SYNB1618, undergoing clinical trials as a phenylketonuria treatment. The pathway for converting phenylalanine to trans-cinnamic acid was moved to a landing pad under the control of a circuit that keeps the pathway off during storage. The resulting strain (EcN SYN8784) achieved higher activity than EcN SYNB1618, reaching levels near when the pathway is carried on a plasmid. This work demonstrates a simple system for engineering EcN that aids quantitative strain design for therapeutics.


Asunto(s)
Escherichia coli , Fenilcetonurias , Humanos , Escherichia coli/genética , Escherichia coli/metabolismo , Antibacterianos/metabolismo , Plásmidos/genética , Genómica , Fenilcetonurias/genética , Fenilcetonurias/terapia
4.
PLoS One ; 18(2): e0280499, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36730255

RESUMEN

E. coli Nissle (EcN) is a non-pathogenic probiotic bacterium of the Enterobacteriaceae family that has been used for over a century to promote general gut health. Despite the history of safe usage of EcN, concerns have been raised regarding the presence of the pks gene cluster, encoding the genotoxin colibactin, due to its association with colorectal cancer. Here, we sought to determine the effect of pks island removal on the in vitro and in vivo robustness and activity of EcN and EcN-derived strains. A deletion of the pks island (Δpks) was constructed in wild type and engineered strains of EcN using lambda red recombineering. Mass spectrometric measurement of N-myristoyl-D-asparagine, released during colibactin maturation, confirmed that the pks deletion abrogated colibactin production. Growth curves were comparable between Δpks strains and their isogenic parents, and wild type EcN displayed no competitive advantage to the Δpks strain in mixed culture. Deletion of pks also had no effect on the activity of strains engineered to degrade phenylalanine (SYNB1618 and SYNB1934) or oxalate (SYNB8802). Furthermore, 1:1 mixed dosing of wild type and Δpks EcN in preclinical mouse and nonhuman primate models demonstrated no competitive disadvantage for the Δpks strain with regards to transit time or colonization. Importantly, there was no significant difference on in vivo strain performance between the clinical-stage strain SYNB1934 and its isogenic Δpks variant with regards to recovery of the quantitative strain-specific biomarkers d5- trans-cinnamic acid, and d5-hippuric acid. Taken together, these data support that the pks island is dispensable for Synthetic Biotic fitness and activity in vivo and that its removal from engineered strains of EcN will not have a deleterious effect on strain efficacy.


Asunto(s)
Proteínas de Escherichia coli , Policétidos , Ratones , Animales , Escherichia coli/metabolismo , Péptidos/genética , Péptidos/metabolismo , Proteínas de Escherichia coli/genética , Policétidos/metabolismo , Familia de Multigenes
6.
Mol Syst Biol ; 18(3): e10539, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35253995

RESUMEN

Enteric hyperoxaluria (EH) is a metabolic disease caused by excessive absorption of dietary oxalate leading to the formation of chronic kidney stones and kidney failure. There are no approved pharmaceutical treatments for EH. SYNB8802 is an engineered bacterial therapeutic designed to consume oxalate in the gut and lower urinary oxalate as a potential treatment for EH. Oral administration of SYNB8802 leads to significantly decreased urinary oxalate excretion in healthy mice and non-human primates, demonstrating the strain's ability to consume oxalate in vivo. A mathematical modeling framework was constructed that combines in vitro and in vivo preclinical data to predict the effects of SYNB8802 administration on urinary oxalate excretion in humans. Simulations of SYNB8802 administration predict a clinically meaningful lowering of urinary oxalate excretion in healthy volunteers and EH patients. Together, these findings suggest that SYNB8802 is a promising treatment for EH.


Asunto(s)
Hiperoxaluria , Animales , Simulación por Computador , Femenino , Humanos , Hiperoxaluria/etiología , Hiperoxaluria/orina , Masculino , Ratones , Oxalatos/metabolismo , Oxalatos/orina
7.
Nat Commun ; 12(1): 6215, 2021 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-34711827

RESUMEN

In phenylketonuria (PKU) patients, a genetic defect in the enzyme phenylalanine hydroxylase (PAH) leads to elevated systemic phenylalanine (Phe), which can result in severe neurological impairment. As a treatment for PKU, Escherichia coli Nissle (EcN) strain SYNB1618 was developed under Synlogic's Synthetic Biotic™ platform to degrade Phe from within the gastrointestinal (GI) tract. This clinical-stage engineered strain expresses the Phe-metabolizing enzyme phenylalanine ammonia lyase (PAL), catalyzing the deamination of Phe to the non-toxic product trans-cinnamate (TCA). In the present work, we generate a more potent EcN-based PKU strain through optimization of whole cell PAL activity, using biosensor-based high-throughput screening of mutant PAL libraries. A lead enzyme candidate from this screen is used in the construction of SYNB1934, a chromosomally integrated strain containing the additional Phe-metabolizing and biosafety features found in SYNB1618. Head-to-head, SYNB1934 demonstrates an approximate two-fold increase in in vivo PAL activity compared to SYNB1618.


Asunto(s)
Terapia Biológica , Proteínas de Escherichia coli/genética , Escherichia coli/enzimología , Fenilanina Amoníaco-Liasa/genética , Fenilalanina/metabolismo , Fenilcetonurias/metabolismo , Fenilcetonurias/terapia , Técnicas Biosensibles , Cinamatos , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Humanos , Fenilanina Amoníaco-Liasa/metabolismo , Ingeniería de Proteínas
8.
Nat Metab ; 3(8): 1125-1132, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34294923

RESUMEN

Phenylketonuria (PKU) is a rare disease caused by biallelic mutations in the PAH gene that result in an inability to convert phenylalanine (Phe) to tyrosine, elevated blood Phe levels and severe neurological complications if untreated. Most patients are unable to adhere to the protein-restricted diet, and thus do not achieve target blood Phe levels. We engineered a strain of E. coli Nissle 1917, designated SYNB1618, through insertion of the genes encoding phenylalanine ammonia lyase and L-amino acid deaminase into the genome, which allow for bacterial consumption of Phe within the gastrointestinal tract. SYNB1618 was studied in a phase 1/2a randomized, placebo-controlled, double-blind, multi-centre, in-patient study ( NCT03516487 ) in adult healthy volunteers (n = 56) and patients with PKU and blood Phe level ≥600 mmol l-1 (n = 14). Participants were randomized to receive a single dose of SYNB1618 or placebo (part 1) or up to three times per day for up to 7 days (part 2). The primary outcome of this study was safety and tolerability, and the secondary outcome was microbial kinetics. A D5-Phe tracer (15 mg kg-1) was used to study exploratory pharmacodynamic effects. SYNB1618 was safe and well tolerated with a maximum tolerated dose of 2 × 1011 colony-forming units. Adverse events were mostly gastrointestinal and of mild to moderate severity. All participants cleared the bacteria within 4 days of the last dose. Dose-responsive increases in strain-specific Phe metabolites in plasma (trans-cinnamic acid) and urine (hippuric acid) were observed, providing a proof of mechanism for the potential to use engineered bacteria in the treatment of rare metabolic disorders.


Asunto(s)
Terapia Biológica/métodos , Escherichia coli , Fenilcetonurias/terapia , Amidohidrolasas/genética , Amidohidrolasas/metabolismo , Terapia Biológica/efectos adversos , Escherichia coli/enzimología , Escherichia coli/genética , Ingeniería Genética , Humanos , Fenilanina Amoníaco-Liasa/genética , Fenilanina Amoníaco-Liasa/metabolismo , Fenilcetonurias/sangre , Fenilcetonurias/genética , Resultado del Tratamiento
9.
Nat Commun ; 11(1): 1738, 2020 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-32269218

RESUMEN

A complex interplay of metabolic and immunological mechanisms underlies many diseases that represent a substantial unmet medical need. There is an increasing appreciation of the role microbes play in human health and disease, and evidence is accumulating that a new class of live biotherapeutics comprised of engineered microbes could address specific mechanisms of disease. Using the tools of synthetic biology, nonpathogenic bacteria can be designed to sense and respond to environmental signals in order to consume harmful compounds and deliver therapeutic effectors. In this perspective, we describe considerations for the design and development of engineered live biotherapeutics to achieve regulatory and patient acceptance.


Asunto(s)
Bacterias/genética , Enfermedad , Ingeniería Genética , Biomarcadores/metabolismo , Tracto Gastrointestinal/microbiología , Humanos , Neoplasias/terapia
10.
Sci Transl Med ; 11(475)2019 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-30651324

RESUMEN

The intestine is a major source of systemic ammonia (NH3); thus, capturing part of gut NH3 may mitigate disease symptoms in conditions of hyperammonemia such as urea cycle disorders and hepatic encephalopathy. As an approach to the lowering of blood ammonia arising from the intestine, we engineered the orally delivered probiotic Escherichia coli Nissle 1917 to create strain SYNB1020 that converts NH3 to l-arginine (l-arg). We up-regulated arginine biosynthesis in SYNB1020 by deleting a negative regulator of l-arg biosynthesis and inserting a feedback-resistant l-arg biosynthetic enzyme. SYNB1020 produced l-arg and consumed NH3 in an in vitro system. SYNB1020 reduced systemic hyperammonemia, improved survival in ornithine transcarbamylase-deficient spfash mice, and decreased hyperammonemia in the thioacetamide-induced liver injury mouse model. A phase 1 clinical study was conducted including 52 male and female healthy adult volunteers. SYNB1020 was well tolerated at daily doses of up to 1.5 × 1012 colony-forming units administered for up to 14 days. A statistically significant dose-dependent increase in urinary nitrate, plasma 15N-nitrate (highest dose versus placebo, P = 0.0015), and urinary 15N-nitrate was demonstrated, indicating in vivo SYNB1020 activity. SYNB1020 concentrations reached steady state by the second day of dosing, and excreted cells were alive and metabolically active as evidenced by fecal arginine production in response to added ammonium chloride. SYNB1020 was no longer detectable in feces 2 weeks after the last dose. These results support further clinical development of SYNB1020 for hyperammonemia disorders including urea cycle disorders and hepatic encephalopathy.


Asunto(s)
Escherichia coli/genética , Ingeniería Genética , Voluntarios Sanos , Hiperamonemia/terapia , Amoníaco/sangre , Amoníaco/metabolismo , Animales , Arginina/metabolismo , Vías Biosintéticas , Modelos Animales de Enfermedad , Heces/química , Femenino , Humanos , Hiperamonemia/sangre , Hiperamonemia/orina , Macaca fascicularis , Masculino , Ratones , Nitratos/sangre , Nitratos/orina , Estrés Fisiológico/genética , Análisis de Supervivencia
11.
Nat Biotechnol ; 36(9): 857-864, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30102294

RESUMEN

Phenylketonuria (PKU) is a genetic disease that is characterized by an inability to metabolize phenylalanine (Phe), which can result in neurotoxicity. To provide a potential alternative to a protein-restricted diet, we engineered Escherichia coli Nissle to express genes encoding Phe-metabolizing enzymes in response to anoxic conditions in the mammalian gut. Administration of our synthetic strain, SYNB1618, to the Pahenu2/enu2 PKU mouse model reduced blood Phe concentration by 38% compared with the control, independent of dietary protein intake. In healthy Cynomolgus monkeys, we found that SYNB1618 inhibited increases in serum Phe after an oral Phe dietary challenge. In mice and primates, Phe was converted to trans-cinnamate by SYNB1618, quantitatively metabolized by the host to hippurate and excreted in the urine, acting as a predictive biomarker for strain activity. SYNB1618 was detectable in murine or primate feces after a single oral dose, permitting the evaluation of pharmacodynamic properties. Our results define a strategy for translation of live bacterial therapeutics to treat metabolic disorders.


Asunto(s)
Terapia Genética , Fenilcetonurias/terapia , Biomarcadores/metabolismo , Escherichia coli/genética , Humanos , Fenilcetonurias/metabolismo
12.
Chem Biol ; 22(4): 535-547, 2015 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-25910245

RESUMEN

Understanding how compound penetration occurs across the complex cell walls of Gram-negative bacteria is one of the greatest challenges in discovering new drugs to treat the infections they cause. A combination of next-generation transposon sequencing, computational metadynamics simulations (CMDS), and medicinal chemistry was used to define genetic and structural elements involved in facilitated carbapenem entry into Pseudomonas aeruginosa. Here we show for the first time that these compounds are taken up not only by the major outer membrane channel OccD1 (also called OprD or PA0958) but also by a closely related channel OccD3 (OpdP or PA4501). Transport-mediating molecular interactions predicted by CMDS for these channels were first confirmed genetically, then used to guide the design of carbapenem analogs with altered uptake properties. These results bring us closer to the rational design of channel transmissibility and may ultimately lead to improved permeability of compounds across bacterial outer membranes.


Asunto(s)
Carbapenémicos/metabolismo , Pseudomonas aeruginosa/metabolismo , Sustitución de Aminoácidos , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Sitios de Unión , Carbapenémicos/química , Carbapenémicos/farmacología , Pruebas de Sensibilidad Microbiana , Simulación del Acoplamiento Molecular , Estructura Terciaria de Proteína , Pseudomonas aeruginosa/efectos de los fármacos , Especificidad por Sustrato
13.
Antiviral Res ; 115: 71-4, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25542974

RESUMEN

Respiratory Syncytial Virus (RSV) is a major cause of lower respiratory tract infections with no effective treatment available. Finding novel inhibitors of RSV is an important first step towards developing an efficacious RSV therapy. Here we report the characterization of three novel classes of RSV replication inhibitors identified through a high throughput RSV replicon screen of ∼1million compounds in the AstraZeneca compound collection. These inhibitors, cpd 1, 2, and 3, specifically targeted RSV and were not active against other viruses tested. Resistance selection in RSV A2 with cpd 1 identified escape viruses with mutations mapped to the RSV L protein, an RNA-dependent RNA polymerase (Y1631C and I1413T). Recombinant RSV containing the L Y1631C substitution conferred resistance towards cpd 1, suggesting that the RSV polymerase is the target of this inhibitor. Interestingly, cpd 3, a nucleoside analog, induced a single resistant mutation in the P protein (D231V), indicating a novel mode of action not previously reported. cpd 2 affected host cell cycle and no frequent mutation was isolated following resistance selection, suggesting its possible involvement of a host-targeted mechanism. Taken together, we have identified three novel RSV inhibitors with different modes of action, providing new chemistry starting points for the discovery and development of future RSV therapeutic treatment.


Asunto(s)
Antivirales/química , Antivirales/farmacología , Ensayos Analíticos de Alto Rendimiento , Replicón/efectos de los fármacos , Virus Sincitial Respiratorio Humano/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Benzotiazoles/química , Benzotiazoles/farmacología , Farmacorresistencia Viral/genética , Indoles/química , Indoles/farmacología , Pruebas de Sensibilidad Microbiana , Mutación , Oxadiazoles/química , Oxadiazoles/farmacología , Nucleósidos de Purina/química , Nucleósidos de Purina/farmacología , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , ARN Polimerasa Dependiente del ARN/metabolismo , Virus Sincitial Respiratorio Humano/genética , Virus Sincitial Respiratorio Humano/fisiología , Timina/análogos & derivados , Timina/química , Timina/farmacología , Proteínas Virales/genética
14.
Microb Ecol ; 68(1): 1-12, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24096885

RESUMEN

Pseudomonas aeruginosa is a ubiquitous organism that is the focus of intense research because of its prominent role in disease. Due to its relatively large genome and flexible metabolic capabilities, this organism exploits numerous environmental niches. It is an opportunistic pathogen that sets upon the human host when the normal immune defenses are disabled. Its deadliness is most apparent in cystic fibrosis patients, but it also is a major problem in burn wounds, chronic wounds, chronic obstructive pulmonary disorder, surface growth on implanted biomaterials, and within hospital surface and water supplies, where it poses a host of threats to vulnerable patients (Peleg and Hooper, N Engl J Med 362:1804-1813, 2010; Breathnach et al., J Hosp Infect 82:19-24, 2012). Once established in the patient, P. aeruginosa can be especially difficult to treat. The genome encodes a host of resistance genes, including multidrug efflux pumps (Poole, J Mol Microbiol Biotechnol 3:255-264, 2001) and enzymes conferring resistance to beta-lactam and aminoglycoside antibotics (Vahdani et al., Annal Burns Fire Disast 25:78-81, 2012), making therapy against this gram-negative pathogen particularly challenging due to the lack of novel antimicrobial therapeutics (Lewis, Nature 485: 439-440, 2012). This challenge is compounded by the ability of P. aeruginosa to grow in a biofilm, which may enhance its ability to cause infections by protecting bacteria from host defenses and chemotherapy. Here, we review recent studies of P. aeruginosa biofilms with a focus on how this unique mode of growth contributes to its ability to cause recalcitrant infections.


Asunto(s)
Biopelículas , Pseudomonas aeruginosa/crecimiento & desarrollo , Pseudomonas aeruginosa/patogenicidad , Animales , Antibacterianos/farmacología , Biopelículas/efectos de los fármacos , Fibrosis Quística/microbiología , Modelos Animales de Enfermedad , Farmacorresistencia Bacteriana , Humanos , Sistema Inmunológico , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/genética , Heridas y Lesiones/microbiología
15.
Microbiology (Reading) ; 158(Pt 2): 560-570, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22117004

RESUMEN

We have reported that Neisseria gonorrhoeae is extremely resistant to reactive nitrogen species (RNS) including peroxynitrite (PN). Recent literature suggests that catalase can provide protection against commercial preparations of PN. Though wild-type gonococci were shown to be highly resistant to 2 mM PN, Neisseria meningitidis and a gonococcal katA mutant were both shown to be extremely sensitive to 2 mM PN. Analysis of translational fusions to lacZ of the catalase promoters from N. gonorrhoeae and N. meningitidis demonstrated that basal katA expression from gonococci is 80-fold higher than in meningococci, though meningococcal katA retains a greater capacity to be activated by OxyR. This activation capacity was shown to be due to a single base pair difference in the -10 transcription element between the two kat promoters. PN resistance was initially shown to be associated with increasing catalase expression; however, commercial preparations of PN were later revealed to contain higher levels of contaminating hydrogen peroxide (H2O2) than expected. Removal of H2O2 from PN preparations with manganese dioxide markedly reduced PN toxicity in a gonococcal katA mutant. Simultaneous treatment with non-lethal concentrations of PN and H2O2 was highly lethal, indicating that these agents act synergistically. When treatment was separated by 5 min, high levels of bacterial killing occurred only when PN was added first. Our results suggest that killing of N. gonorrhoeae ΔkatA by commercial PN preparations is likely due to H2O2, that H2O2 is more toxic in the presence of PN, and that PN, on its own, may not be as toxic as previously believed.


Asunto(s)
Proteínas Bacterianas/metabolismo , Catalasa/metabolismo , Farmacorresistencia Bacteriana , Gonorrea/microbiología , Neisseria gonorrhoeae/efectos de los fármacos , Neisseria gonorrhoeae/enzimología , Ácido Peroxinitroso/farmacología , Antibacterianos/farmacología , Proteínas Bacterianas/genética , Catalasa/genética , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Humanos , Neisseria gonorrhoeae/genética
16.
Mol Microbiol ; 82(2): 489-501, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21895795

RESUMEN

Transcriptome analysis of the facultative anaerobe, Neisseria gonorrhoeae, revealed that many genes of unknown function were induced under anaerobic conditions. Mutation of one such gene, NGO1024, encoding a protein belonging to the 2-nitropropane dioxygenase-like superfamily of proteins, was found to result in an inability of gonococci to grow anaerobically. Anaerobic growth of an NG1024 mutant was restored upon supplementation with unsaturated fatty acids (UFA), but not with the saturated fatty acid palmitate. Gonococcal fatty acid profiles confirmed that NGO1024 was involved in UFA synthesis anaerobically, but not aerobically, demonstrating that gonococci contain two distinct pathways for the production of UFAs, with a yet unidentified aerobic mechanism, and an anaerobic mechanism involving NGO1024. Expression of genes involved in classical anaerobic UFA synthesis, fabA, fabM and fabB, was toxic in gonococci and unable to complement a NGO1024 mutation, suggesting that the chemistry involved in gonococcal anaerobic UFA synthesis is distinct from that of the classical pathway. NGO1024 homologues, which we suggest naming UfaA, form a distinct lineage within the 2-nitropropane dioxygenase-like superfamily, and are found in many facultative and obligate anaerobes that produce UFAs but lack fabA, suggesting that UfaA is part of a widespread pathway involved in UFA synthesis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Dioxigenasas/metabolismo , Ácidos Grasos Insaturados/biosíntesis , Hidroliasas/metabolismo , Neisseria gonorrhoeae/enzimología , Anaerobiosis , Proteínas Bacterianas/genética , Dioxigenasas/genética , Evolución Molecular , Hidroliasas/genética , Mutación , Neisseria gonorrhoeae/clasificación , Neisseria gonorrhoeae/genética , Neisseria gonorrhoeae/metabolismo , Filogenia
17.
BMC Genomics ; 12: 51, 2011 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-21251255

RESUMEN

BACKGROUND: Maintenance of an anaerobic denitrification system in the obligate human pathogen, Neisseria gonorrhoeae, suggests that an anaerobic lifestyle may be important during the course of infection. Furthermore, mounting evidence suggests that reduction of host-produced nitric oxide has several immunomodulary effects on the host. However, at this point there have been no studies analyzing the complete gonococcal transcriptome response to anaerobiosis. Here we performed deep sequencing to compare the gonococcal transcriptomes of aerobically and anaerobically grown cells. Using the information derived from this sequencing, we discuss the implications of the robust transcriptional response to anaerobic growth. RESULTS: We determined that 198 chromosomal genes were differentially expressed (~10% of the genome) in response to anaerobic conditions. We also observed a large induction of genes encoded within the cryptic plasmid, pJD1. Validation of RNA-seq data using translational-lacZ fusions or RT-PCR demonstrated the RNA-seq results to be very reproducible. Surprisingly, many genes of prophage origin were induced anaerobically, as well as several transcriptional regulators previously unknown to be involved in anaerobic growth. We also confirmed expression and regulation of a small RNA, likely a functional equivalent of fnrS in the Enterobacteriaceae family. We also determined that many genes found to be responsive to anaerobiosis have also been shown to be responsive to iron and/or oxidative stress. CONCLUSIONS: Gonococci will be subject to many forms of environmental stress, including oxygen-limitation, during the course of infection. Here we determined that the anaerobic stimulon in gonococci was larger than previous studies would suggest. Many new targets for future research have been uncovered, and the results derived from this study may have helped to elucidate factors or mechanisms of virulence that may have otherwise been overlooked.


Asunto(s)
Proteínas Bacterianas/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Neisseria gonorrhoeae/genética , Anaerobiosis/genética , Regulación Bacteriana de la Expresión Génica/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Microbiology (Reading) ; 155(Pt 12): 4093-4103, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19762442

RESUMEN

Since Neisseria gonorrhoeae and Neisseria meningitidis are obligate human pathogens, a comparison with commensal species of the same genus could reveal differences important in pathogenesis. The recent completion of commensal Neisseria genome draft assemblies allowed us to perform a comparison of the genes involved in the catalysis, assembly and regulation of the denitrification pathway, which has been implicated in the virulence of several bacteria. All species contained a highly conserved nitric oxide reductase (NorB) and a nitrite reductase (AniA or NirK) that was highly conserved in the catalytic but divergent in the N-terminal lipid modification and C-terminal glycosylation domains. Only Neisseria mucosa contained a nitrate reductase (Nar), and only Neisseria lactamica, Neisseria cinerea, Neisseria subflava, Neisseria flavescens and Neisseria sicca contained a nitrous oxide reductase (Nos) complex. The regulators of the denitrification genes, FNR, NarQP and NsrR, were highly conserved, except for the GAF domain of NarQ. Biochemical examination of laboratory strains revealed that all of the neisserial species tested except N. mucosa had a two- to fourfold lower nitrite reductase activity than N. gonorrhoeae, while N. meningitidis and most of the commensal Neisseria species had a two- to fourfold higher nitric oxide (NO) reductase activity. For N. meningitidis and most of the commensal Neisseria, there was a greater than fourfold reduction in the NO steady-state level in the presence of nitrite as compared with N. gonorrhoeae. All of the species tested generated an NO steady-state level in the presence of an NO donor that was similar to that of N. gonorrhoeae. The greatest difference between the Neisseria species was the lack of a functional Nos system in the pathogenic species N. gonorrhoeae and N. meningitidis.


Asunto(s)
Neisseria/genética , Neisseria/metabolismo , Nitritos/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Genes Bacterianos , Genómica , Humanos , Neisseria/crecimiento & desarrollo , Neisseria/patogenicidad , Nitrito Reductasas/química , Nitrito Reductasas/genética , Nitrito Reductasas/metabolismo , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Estructura Terciaria de Proteína , Especificidad de la Especie , Virulencia
19.
Microbiology (Reading) ; 155(Pt 8): 2532-2545, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19406894

RESUMEN

Neisseria gonorrhoeae encodes a number of important genes that aid in survival during times of oxidative stress. The same immune cells capable of oxygen-dependent killing mechanisms also have the capacity to generate reactive nitrogen species (RNS) that may function antimicrobially. F62 and eight additional gonococcal strains displayed a high level of resistance to peroxynitrite, while Neisseria meningitidis and Escherichia coli showed a four- to seven-log and a four-log decrease in viability, respectively. Mutation of gonococcal orthologues that are known or suspected to be involved in RNS defence in other bacteria (ahpC, dnrN and msrA) resulted in no loss of viability, suggesting that N. gonorrhoeae has a novel mechanism of resistance to peroxynitrite. Whole-cell extracts of F62 prevented the oxidation of dihydrorhodamine, and decomposition of peroxynitrite was not dependent on ahpC, dnrN or msrA. F62 grown in co-culture with E. coli strain DH10B was shown to protect E. coli viability 10-fold. Also, peroxynitrite treatment of F62 did not result in accumulation of nitrated proteins, suggesting that an active peroxynitrite reductase is responsible for peroxynitrite decomposition rather than a protein sink for amino acid modification.


Asunto(s)
Neisseria gonorrhoeae/metabolismo , Ácido Peroxinitroso/metabolismo , Relación Dosis-Respuesta a Droga , Farmacorresistencia Bacteriana/genética , Escherichia coli/efectos de los fármacos , Escherichia coli/metabolismo , Eliminación de Gen , Genes Bacterianos , Neisseria gonorrhoeae/efectos de los fármacos , Neisseria gonorrhoeae/patogenicidad , Neisseria meningitidis/efectos de los fármacos , Neisseria meningitidis/metabolismo , Óxido Nítrico/metabolismo , Oxidorreductasas/metabolismo , Ácido Peroxinitroso/farmacología , Especies Reactivas de Oxígeno/metabolismo , Virulencia
20.
Mol Microbiol ; 71(1): 227-39, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19007408

RESUMEN

Nitric oxide (NO) has been shown to be an important component of the human immune response, and as such, it is important to understand how pathogenic organisms respond to its presence. In Neisseria gonorrhoeae, recent work has revealed that NsrR, an Rrf2-type transcriptional repressor, can sense NO and control the expression of genes responsible for NO metabolism. A highly pure extract of epitope-tagged NsrR was isolated and mass spectroscopic analysis suggested that the protein contained a [2Fe-2S] cluster. NsrR/DNA interactions were thoroughly analysed in vitro. Using EMSA analysis, NsrR::FLAG was shown to interact with predicted operators in the norB, aniA and nsrR upstream regions with a K(d) of 7, 19 and 35 nM respectively. DNase I footprint analysis was performed on the upstream regions of norB and nsrR, where NsrR was shown to protect the predicted 29 bp binding sites. The presence of exogenously added NO inhibited DNA binding by NsrR. Alanine substitution of C90, C97 or C103 in NsrR abrogated repression of norB::lacZ and inhibited DNA binding, consistent with their presumed role in co-ordination of a NO-sensitive Fe-S centre required for DNA binding.


Asunto(s)
Proteínas Bacterianas/metabolismo , Neisseria gonorrhoeae/genética , Óxido Nítrico/metabolismo , Proteínas Represoras/metabolismo , Proteínas Bacterianas/genética , Huella de ADN , ADN Bacteriano/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Regulación Bacteriana de la Expresión Génica , Genes Bacterianos , Proteínas Hierro-Azufre/genética , Proteínas Hierro-Azufre/metabolismo , Mutagénesis Sitio-Dirigida , Neisseria gonorrhoeae/metabolismo , Regiones Operadoras Genéticas , Unión Proteica , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Represoras/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...