Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 113
Filtrar
1.
J Nat Prod ; 87(2): 424-438, 2024 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-38289177

RESUMEN

Ever since the isolation of Amycolatopsis mediterranei in 1957, this strain has been the focus of research worldwide. In the last 60 years or more, our understanding of the taxonomy, development of cloning vectors and conjugation system, physiology, genetics, genomics, and biosynthetic pathway of rifamycin B production in A. mediterranei has substantially increased. In particular, the development of cloning vectors, transformation system, characterization of the rifamycin biosynthetic gene cluster, and the regulation of rifamycin B production by the pioneering work of Heinz Floss have made the rifamycin polyketide biosynthetic gene cluster (PKS) an attractive target for extensive genetic manipulations to produce rifamycin B analogues which could be effective against multi-drug-resistant tuberculosis. Additionally, a better understanding of the regulation of rifamycin B production and the application of newer genomics tools, including CRISPR-assisted genome editing systems, might prove useful to overcome the limitations associated with low production of rifamycin analogues.


Asunto(s)
Actinomycetales , Rifamicinas , Amycolatopsis , Vías Biosintéticas/genética , Rifamicinas/metabolismo
2.
mBio ; 15(2): e0205123, 2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-38126769

RESUMEN

The emergence of multidrug-resistant Gram-negative bacteria underscores the need to define genetic vulnerabilities that can be therapeutically exploited. The Gram-negative pathogen, Acinetobacter baumannii, is considered an urgent threat due to its propensity to evade antibiotic treatments. Essential cellular processes are the target of existing antibiotics and a likely source of new vulnerabilities. Although A. baumannii essential genes have been identified by transposon sequencing, they have not been prioritized by sensitivity to knockdown or antibiotics. Here, we take a systems biology approach to comprehensively characterize A. baumannii essential genes using CRISPR interference (CRISPRi). We show that certain essential genes and pathways are acutely sensitive to knockdown, providing a set of vulnerable targets for future therapeutic investigation. Screening our CRISPRi library against last-resort antibiotics uncovered genes and pathways that modulate beta-lactam sensitivity, an unexpected link between NADH dehydrogenase activity and growth inhibition by polymyxins, and anticorrelated phenotypes that may explain synergy between polymyxins and rifamycins. Our study demonstrates the power of systematic genetic approaches to identify vulnerabilities in Gram-negative pathogens and uncovers antibiotic-essential gene interactions that better inform combination therapies.IMPORTANCEAcinetobacter baumannii is a hospital-acquired pathogen that is resistant to many common antibiotic treatments. To combat resistant A. baumannii infections, we need to identify promising therapeutic targets and effective antibiotic combinations. In this study, we comprehensively characterize the genes and pathways that are critical for A. baumannii viability. We show that genes involved in aerobic metabolism are central to A. baumannii physiology and may represent appealing drug targets. We also find antibiotic-gene interactions that may impact the efficacy of carbapenems, rifamycins, and polymyxins, providing a new window into how these antibiotics function in mono- and combination therapies. Our studies offer a useful approach for characterizing interactions between drugs and essential genes in pathogens to inform future therapies.


Asunto(s)
Acinetobacter baumannii , Rifamicinas , Antibacterianos/farmacología , Antibacterianos/metabolismo , Genes Esenciales , Polimixinas/farmacología , Farmacorresistencia Bacteriana Múltiple/genética , Rifamicinas/metabolismo , Rifamicinas/farmacología , Pruebas de Sensibilidad Microbiana
3.
Acc Chem Res ; 54(9): 2065-2075, 2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-33877820

RESUMEN

Rifamycin antibiotics include the WHO essential medicines rifampin, rifabutin, and rifapentine. These are semisynthetic derivatives of the natural product rifamycins, originally isolated from the soil bacterium Amycolatopsis rifamycinica. These antibiotics are primarily used to treat mycobacterial infections, including tuberculosis. Rifamycins act by binding to the ß-subunit of bacterial RNA polymerase, inhibiting transcription, which results in cell death. These antibiotics consist of a naphthalene core spanned by a polyketide ansa bridge. This structure presents a unique 3D configuration that engages RNA polymerase through a series of hydrogen bonds between hydroxyl groups linked to the naphthalene core and C21 and C23 of the ansa bridge. This binding occurs not in the enzyme active site where template-directed RNA synthesis occurs but instead in the RNA exit tunnel, thereby blocking productive formation of full-length RNA. In their clinical use to treat tuberculosis, resistance to rifamycin antibiotics arises principally from point mutations in RNA polymerase that decrease the antibiotic's affinity for the binding site in the RNA exit tunnel. In contrast, the rifamycin resistome of environmental mycobacteria and actinomycetes is much richer and diverse. In these organisms, rifamycin resistance includes many different enzymatic mechanisms that modify and alter the antibiotic directly, thereby inactivating it. These enzymes include ADP ribosyltransferases, glycosyltransferases, phosphotransferases, and monooxygenases.ADP ribosyltransferases catalyze group transfer of ADP ribose from the cofactor NAD+, which is more commonly deployed for metabolic redox reactions. ADP ribose is transferred to the hydroxyl linked to C23 of the antibiotic, thereby sterically blocking productive interaction with RNA polymerase. Like ADP ribosyltransferases, rifamycin glycosyl transferases also modify the hydroxyl of position C23 of rifamycins, transferring a glucose moiety from the donor molecule UDP-glucose. Unlike other antibiotic resistance kinases that transfer the γ-phosphate of ATP to inactivate antibiotics such as aminoglycosides or macrolides, rifamycin phosphotransferases are ATP-dependent dikinases. These enzymes transfer the ß-phosphate of ATP to the C21 hydroxyl of the rifamycin ansa bridge. The result is modification of a critical RNA polymerase binding group that blocks productive complex formation. On the other hand, rifamycin monooxygenases are FAD-dependent enzymes that hydroxylate the naphthoquinone core. The result of this modification is untethering of the ansa chain from the naphthyl moiety, disrupting the essential 3D shape necessary for productive RNA polymerase binding and inhibition that leads to cell death.All of these enzymes have homologues in bacterial metabolism that either are their direct precursors or share common ancestors to the resistance enzyme. The diversity of these resistance mechanisms, often redundant in individual bacterial isolates, speaks to the importance of protecting RNA polymerase from these compounds and validates this enzyme as a critical antibiotic target.


Asunto(s)
Antibacterianos/metabolismo , ARN Polimerasa Dependiente del ARN/metabolismo , Rifamicinas/metabolismo , Amycolatopsis/química , Antibacterianos/química , Antibacterianos/farmacología , Farmacorresistencia Bacteriana/efectos de los fármacos , Rifamicinas/química , Rifamicinas/farmacología
4.
Org Lett ; 23(6): 2342-2346, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33683897

RESUMEN

Rifamycins have been clinically utilized against mycobacterial infections for more than 50 years; however, their biosynthesis has not been fully elucidated. Here, on the basis of in vivo gene deletions, in vitro enzyme assays, isotope labeling, and site-directed mutations, we found that a flavin-dependent monooxygenase encoded by a rifamycin biosynthetic gene cluster, Rif-Orf17, not only converted the naphthoquinone chromophore of rifamycin S into benzo-γ-pyrone but also linearized rifamycin SV through phenolic hydroxylation. Both oxidation routes lead to inactivation of rifamycins.


Asunto(s)
Flavinas/química , Oxigenasas de Función Mixta/química , Rifamicinas/química , Antibacterianos/biosíntesis , Antibacterianos/química , Flavinas/metabolismo , Estructura Molecular , Familia de Multigenes , Oxidación-Reducción , Rifamicinas/metabolismo
5.
Biomolecules ; 10(10)2020 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-32998374

RESUMEN

Matrix metalloproteinases 1 (MMP-1) energetically triggers the enzymatic proteolysis of extracellular matrix collagenase (ECM), resulting in progressive skin aging. Natural flavonoids are well known for their antioxidant properties and have been evaluated for inhibition of matrix metalloproteins in human. Recently, (-)-epicatechin and proanthocyanidin B2 were reported as essential flavanols from various natural reservoirs as potential anti-inflammatory and free radical scavengers. However, their molecular interactions and inhibitory potential against MMP-1 are not yet well studied. In this study, sequential absorption, distribution, metabolism, and excretion (ADME) profiling, quantum mechanics calculations, and molecular docking simulations by extra precision Glide protocol predicted the drug-likeness of (-)-epicatechin (-7.862 kcal/mol) and proanthocyanidin B2 (-8.145 kcal/mol) with the least reactivity and substantial binding affinity in the catalytic pocket of human MMP-1 by comparison to reference bioactive compound epigallocatechin gallate (-6.488 kcal/mol). These flavanols in docked complexes with MMP-1 were further studied by 500 ns molecular dynamics simulations that revealed substantial stability and intermolecular interactions, viz. hydrogen and ionic interactions, with essential residues, i.e., His218, Glu219, His222, and His228, in the active pocket of MMP-1. In addition, binding free energy calculations using the Molecular Mechanics Generalized Born Surface Area (MM/GBSA) method suggested the significant role of Coulomb interactions and van der Waals forces in the stability of respective docked MMP-1-flavonol complexes by comparison to MMP-1-epigallocatechin gallate; these observations were further supported by MMP-1 inhibition assay using zymography. Altogether with computational and MMP-1-zymography results, our findings support (-)-epicatechin as a comparatively strong inhibitor of human MMP-1 with considerable drug-likeness against proanthocyanidin B2 in reference to epigallocatechin gallate.


Asunto(s)
Catequina/química , Metaloproteinasa 1 de la Matriz/química , Inhibidores de la Metaloproteinasa de la Matriz/química , Rifamicinas/química , Sitios de Unión , Catequina/análogos & derivados , Catequina/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Metaloproteinasa 1 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz/metabolismo , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Teoría Cuántica , Rifamicinas/metabolismo , Solubilidad , Estereoisomerismo , Termodinámica
6.
Eur J Med Chem ; 167: 96-104, 2019 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-30769243

RESUMEN

New rifamycin congeners (1-33) with incorporated amine and hydrazone substituents leading to lipophilic and/or basic nature and altered rigidity of modified C(3) arm were synthesized and structurally characterized in detail. NMR spectroscopic studies at different temperatures indicate two types of structures of rifamycin congeners that are realized in solution: zwitterionic and non-ionic forms in dependence of the basicity of modified C(3) arm. The presence of rifamycin congeners in these two possible forms has a significant impact on the physico-chemical parameters such as lipophilicity (clogP) and water solubility and different binding mode of the C(3) arm of antibiotic at RNAP binding pocket (molecular target) leading to different antibacterial potency. The highest antibacterial potency against S. aureus (including MRSA and MLSB strains) and S. epidermidis strains, even higher than reference rifampicin (Rif) and rifaximin (Rifx) antibiotics, was found for rifamycin congeners bearing at the C(3) arm relatively rigid and basic substituents (bipiperidine and guanidine groups). These modifications provide favorable docking mode and excellent water solubility resulting in high potency (MICs 0.0078 µg/mL what gives ∼ 8.5 nM), irrespective whether rifamycin congener is a tertiary amine (15) or hydrazone (29). In turn, for a higher antibacterial potency of rifamycin congeners against E. faecalis strain (MICs 0.5 µg/mL that is 0.6 µM) as compared to Rif and Rifx, the most crucial factors are: bulkiness and the lipophilic character of the end of the C(3) rebuilt arm.


Asunto(s)
Aminas/química , Antibacterianos/síntesis química , Hidrazonas/química , Simulación del Acoplamiento Molecular , Rifamicinas/química , Rifamicinas/farmacología , Antibacterianos/química , Antibacterianos/farmacología , Sitios de Unión , Interacciones Hidrofóbicas e Hidrofílicas , Espectroscopía de Resonancia Magnética , Rifampin/farmacología , Rifamicinas/síntesis química , Rifamicinas/metabolismo , Solubilidad , Relación Estructura-Actividad
7.
Sci Rep ; 8(1): 11897, 2018 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-30093677

RESUMEN

Multiple mutations in the ß subunit of the RNA polymerase (rpoß) of Mycobacterium tuberculosis (Mtb) are the primary cause of resistance to rifamycin (RIF). In the present study, bifidobacterial rpoß sequences were analyzed to characterize the mutations that contribute to the development of intrinsic resistance to RIF, isoniazid, streptomycin and pyrazinamide. Sequence variations, which mapped to cassettes 1 and 2 of the rpoß pocket, are also found in multidrug-resistant Mtb (MDR Mtb). Growth curves in the presence of osmolytes and different concentrations of RIF showed that the bacteria adapted rapidly by shortening the growth curve lag time. Insight into the adapted rpoß DNA sequences revealed that B. adolescentis harbored mutations both in the RIF pocket and in regions outside the pocket. The minimum inhibitory concentrations (MICs) and mutant prevention concentrations (MPCs) indicated that B. longum, B. adolescentis and B. animalis are resistant to antitubercular drugs. 3D-homology modeling and binding interaction studies using computational docking suggested that mutants had reduced binding affinity towards RIF. RIF-exposed/resistant bacteria exhibited variant protein profiles along with morphological differences, such as elongated and branched cells, surface conversion from rough to smooth, and formation of a concentrating ring.


Asunto(s)
Antituberculosos/farmacología , Proteínas Bacterianas/metabolismo , Bifidobacterium adolescentis/efectos de los fármacos , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , ARN Polimerasa II/metabolismo , Antituberculosos/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Bifidobacterium adolescentis/genética , Bifidobacterium adolescentis/crecimiento & desarrollo , Sitios de Unión/genética , Farmacorresistencia Bacteriana Múltiple/genética , Isoniazida/metabolismo , Isoniazida/farmacología , Pruebas de Sensibilidad Microbiana , Simulación del Acoplamiento Molecular , Mutación , Unión Proteica , Dominios Proteicos , Pirazinamida/metabolismo , Pirazinamida/farmacología , ARN Polimerasa II/química , ARN Polimerasa II/genética , Rifamicinas/metabolismo , Rifamicinas/farmacología
8.
Mol Pharm ; 15(10): 4677-4688, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29996058

RESUMEN

The use of in vitro data for the quantitative prediction of transporter-mediated clearance is critical. Central to this evaluation is the use of hepatocytes, since they contain the full complement of transporters and metabolic enzymes. In general, uptake clearance (CLuptake) is evaluated by measuring the appearance of compound in the cell. Passive clearance (CLpd) is often determined by conducting parallel studies at 4 °C or by attempting to saturate uptake pathways. Both approaches have their limitations. Recent studies have proposed the use of Rifamycin-SV (RFV) as a pan-inhibitor of hepatic uptake pathways. In our studies, we confirm that transport activity of all major hepatic uptake transporters is inhibited significantly by RFV at 1 mM (OATP1B1, 1B3, and 2B1 = NTCP (80%), OCT1 (65%), OAT2 (60%)). Under these incubation conditions, we found that the free intracellular concentration of RFV is ∼175 µM and that several major CYPs and UGTs can be reversibly inhibited. Using this approach, we also determined CLuptake and CLpd of nine known OATP substrates across three different lots of human hepatocytes. The scaling factors generated for these compounds at 37 °C with RFV and 4 °C were found to be similar. The CLpd of passively permeable compounds like metoprolol and semagacestat were found to be higher at 37 °C compared to 4 °C, indicating a temperature effect on these compounds. In addition, our data also suggests that incorporation of medium concentrations into CLuptake and CLpd calculations may be critical for highly protein bound and highly lipophilic drugs. Overall, our data indicate that RFV, instead of 4 °C, can be reliably used to measure CLuptake and CLpd of drugs.


Asunto(s)
Hepatocitos/metabolismo , Hígado/metabolismo , Rifamicinas/metabolismo , Alanina/análogos & derivados , Alanina/metabolismo , Azepinas/metabolismo , Transporte Biológico , Humanos , Cinética , Metoprolol/metabolismo
9.
Cell Chem Biol ; 25(4): 403-412.e5, 2018 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-29398560

RESUMEN

Rifamycin monooxygenases (Rox) are present in a variety of environmental bacteria and are associated with decomposition of the clinically utilized antibiotic rifampin. Here we report the structure and function of a drug-inducible rox gene from Streptomyces venezuelae, which encodes a class A flavoprotein monooxygenase that inactivates a broad range of rifamycin antibiotics. Our findings describe a mechanism of rifamycin inactivation initiated by monooxygenation of the 2-position of the naphthyl group, which subsequently results in ring opening and linearization of the antibiotic. The result is an antibiotic that no longer adopts the basket-like structure essential for binding to the RNA exit tunnel of the target RpoB, thereby providing the molecular logic of resistance. This unique mechanism of enzymatic inactivation underpins the broad spectrum of rifamycin resistance mediated by Rox enzymes and presents a new antibiotic resistance mechanism not yet seen in microbial antibiotic detoxification.


Asunto(s)
Antibacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Farmacorresistencia Bacteriana , Oxigenasas de Función Mixta/metabolismo , Rifamicinas/metabolismo , Streptomyces/enzimología , Antibacterianos/química , Antibacterianos/farmacología , Proteínas Bacterianas/química , Oxigenasas de Función Mixta/química , Simulación del Acoplamiento Molecular , Conformación Proteica , Rifamicinas/química , Rifamicinas/farmacología , Streptomyces/química , Streptomyces/efectos de los fármacos , Streptomyces/metabolismo
10.
Microb Cell Fact ; 17(1): 14, 2018 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-29375035

RESUMEN

BACKGROUND: Due to the important role of rifamycin in curing tuberculosis infection, the study on rifamycin has never been stopped. Although RifZ, which locates within the rifamycin biosynthetic cluster, has recently been characterized as a pathway-specific regulator for rifamycin biosynthesis, little is known about the regulation of rifamycin export. RESULTS: In this work, we proved that the expression of the rifamycin efflux pump (RifP) was regulated by RifQ, a TetR-family transcriptional regulator. Deletion of rifQ had little impact on bacterial growth, but resulted in improved rifamycin production, which was consistent with the reverse transcription PCR results that RifQ negatively regulated rifP's transcription. With electrophoretic mobility shift assay and DNase I Footprinting assay, RifQ was found to directly bind to the promoter region of rifP, and a typical inverted repeat was identified within the RifQ-protected sequences. The transcription initiation site of rifP was further characterized and found to be upstream of the RifQ binding sites, well explaining the RifQ-mediated repression of rifP's transcription in vivo. Moreover, rifamycin B (the end product of rifamycin biosynthesis) remarkably decreased the DNA binding affinity of RifQ, which led to derepression of rifamycin export, reducing the intracellular concentration of rifamycin B as well as its toxicity against the host. CONCLUSIONS: Here, we proved that the export of rifamycin B was repressed by RifQ in Amycolatopsis mediterranei, and the RifQ-mediated repression could be specifically relieved by rifamycin B, the end product of rifamycin biosynthesis, based on which a feedback model was proposed for regulation of rifamycin export. With the findings here, one could improve the antibiotic yield by simply inactivating the negative regulator of the antibiotic transporter.


Asunto(s)
Actinobacteria/genética , Regulación Bacteriana de la Expresión Génica , Genes Bacterianos , Rifamicinas/metabolismo , Actinobacteria/metabolismo , Antibacterianos/biosíntesis , Antibacterianos/metabolismo , Vías Biosintéticas/genética , Familia de Multigenes , Rifamicinas/biosíntesis , Transactivadores/genética
11.
Anal Chem ; 89(21): 11583-11591, 2017 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-29028328

RESUMEN

Amyloid aggregates are associated with several debilitating diseases, and there are numerous efforts to develop small molecule treatments against these diseases. One challenge associated with these efforts is determining protein binding site information for potential therapeutics because amyloid-forming proteins rapidly form oligomers and aggregates, making traditional protein structural analysis techniques challenging. Using ß-2-microglobulin (ß2m) as a model amyloid-forming protein along with two recently identified small molecule amyloid inhibitors (i.e., rifamycin SV and doxycycline), we demonstrate that covalent labeling and mass spectrometry (MS) can be used to map small-molecule binding sites for a rapidly aggregating protein. Specifically, three different covalent labeling reagents, namely diethylpyrocarbonate, 2,3-butanedione, and the reagent pair EDC/GEE, are used together to pinpoint the binding sites of rifamycin SV, doxycycline, and another molecule, suramin, which binds but does not inhibit Cu(II)-induced ß2m amyloid formation. The labeling results reveal binding sites that are consistent with the known effects of these molecules on ß2m amyloid formation and are in general agreement with molecular docking results. We expect that this combined covalent labeling approach will be applicable to other protein/small molecule systems that are difficult to study by traditional means.


Asunto(s)
Amiloide/química , Amiloide/metabolismo , Espectrometría de Masas , Simulación del Acoplamiento Molecular , Agregado de Proteínas/efectos de los fármacos , Sitios de Unión , Doxiciclina/metabolismo , Doxiciclina/farmacología , Humanos , Unión Proteica , Conformación Proteica , Proteolisis , Rifamicinas/metabolismo , Rifamicinas/farmacología , Coloración y Etiquetado , Suramina/metabolismo , Suramina/farmacología , Microglobulina beta-2/química , Microglobulina beta-2/metabolismo
12.
Artículo en Inglés | MEDLINE | ID: mdl-27270559

RESUMEN

Rifamycins inhibit RNA polymerase of most bacterial genera. Rifampicin remains part of combination therapy for treating tuberculosis (TB), and for treating Gram-positive prosthetic joint and valve infections, in which biofilms are prominent. Rifabutin has use for AIDS patients in treating mycobacterial infections TB and Mycobacterium avium complex (MAC), having fewer drug-drug interactions that interfere with AIDS medications. Rifabutin is occasionally used in combination to eradicate Helicobacter pylori (peptic ulcer disease). Rifapentine has yet to fulfill its potential in reducing time of treatment for TB. Rifaximin is a monotherapeutic agent to treat gastrointestinal (GI) disorders, such as hepatic encephalopathy, irritable bowel syndrome, and travelers' diarrhea. Rifaximin is confined to the GI tract because it is not systemically absorbed on oral dosing, achieving high local concentrations, and showing anti-inflammatory properties in addition to its antibacterial activity. Resistance issues are unavoidable with all the rifamycins when the bioburden is high, because of mutations that modify RNA polymerase.


Asunto(s)
Antibacterianos/farmacología , Antibióticos Antituberculosos/farmacología , Rifamicinas/farmacología , Antibacterianos/metabolismo , Antibacterianos/uso terapéutico , Antibióticos Antituberculosos/metabolismo , Antibióticos Antituberculosos/uso terapéutico , Farmacorresistencia Bacteriana/efectos de los fármacos , Farmacorresistencia Bacteriana/genética , Quimioterapia Combinada , Enfermedades Gastrointestinales/tratamiento farmacológico , Humanos , Rifabutina/metabolismo , Rifabutina/uso terapéutico , Rifampin/análogos & derivados , Rifampin/metabolismo , Rifampin/uso terapéutico , Rifamicinas/metabolismo , Rifamicinas/uso terapéutico , Rifaximina , Tuberculosis/tratamiento farmacológico
13.
Eur J Med Chem ; 116: 216-221, 2016 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-27061985

RESUMEN

New rifamycins (1-12) combined with different l-amino acids, containing methyl, ethyl, tert-butyl and benzyl groups at the ester part, via amine linkage, were synthesized and their structures in solution were determined by spectroscopic FT-IR and 1D and 2D NMR methods as well as visualized by DFT calculations. Two types of rifamycin structures were detected in solution: a zwitterionic one with the transferred proton from O(8)H phenol to secondary N(38) atom and a pseudocyclic structure stabilized via formation of intramolecular H-bond within the protonated basic C(3)-substituent. The presence of these rifamycins' structures influenced physico-chemical (logP, solubility) parameters and antibacterial properties. The bulkiness at the ester substituent of new rifamycins containing aromatic l-amino acids was found to be an important factor, besides the solubility, to achieve relatively high antibacterial activity against reference S. epidermidis and reference S. aureus and MRSA strains (MICs 0.016-0.063 µg/mL), comparable to that of rifampicin. SAR for the novel derivatives was discussed in view of the calculated structures of rifamycin-RNAP complexes.


Asunto(s)
Aminoácidos/química , ARN Polimerasas Dirigidas por ADN/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Ésteres/química , Rifamicinas/química , Rifamicinas/farmacología , ARN Polimerasas Dirigidas por ADN/química , ARN Polimerasas Dirigidas por ADN/metabolismo , Inhibidores Enzimáticos/metabolismo , Enlace de Hidrógeno , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/enzimología , Pruebas de Sensibilidad Microbiana , Conformación Molecular , Simulación del Acoplamiento Molecular , Rifamicinas/metabolismo , Staphylococcus epidermidis/efectos de los fármacos , Staphylococcus epidermidis/enzimología , Relación Estructura-Actividad
14.
Yakugaku Zasshi ; 135(11): 1235-44, 2015.
Artículo en Japonés | MEDLINE | ID: mdl-26521872

RESUMEN

Esterases hydrolyze compounds containing ester, amide, and thioester bonds, causing prodrug activation or detoxification. Among esterases, carboxylesterases have been studied in depth due to their ability to hydrolyze a variety of drugs. However, there are several drugs for which the involved esterase(s) is unknown. We found that flutamide, phenacetin, rifamycins (rifampicin, rifabutin, and rifapentine), and indiplon are hydrolyzed by arylacetamide deacetylase (AADAC), which is highly expressed in human liver and gastrointestinal tissues. Flutamide hydrolysis is considered associated with hepatotoxicity. Phenacetin, a prodrug of acetaminophen, was withdrawn due to side effects such as methemoglobinemia and renal failure. It was demonstrated in vitro and in vivo using mice that AADAC is responsible for phenacetin hydrolysis, which leads to methemoglobinemia. In addition, it was shown that AADAC-mediated hydrolysis attenuates the cytotoxicity of rifamycins. Thus AADAC plays critical roles in drug-induced toxicity. Another orphan esterase, α/ß hydrolase domain containing 10 (ABHD10), was found responsible for deglucuronidation of acyl-glucuronides including mycophenolic acid acyl-glucuronide and probenecid acyl-glucuronide. Because acyl-glucuronides appear associated with toxicity, ABHD10 would function as a detoxification enzyme. The roles of orphan esterases are becoming increasingly understood. Further studies will facilitate our knowledge of the pharmacologic and toxicological significance of orphan esterases in drug therapy.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/enzimología , Esterasas/metabolismo , Animales , Hidrolasas de Éster Carboxílico/genética , Hidrolasas de Éster Carboxílico/fisiología , Humanos , Hidrolasas/metabolismo , Fenacetina/metabolismo , Rifamicinas/metabolismo , Especificidad de la Especie
15.
Appl Microbiol Biotechnol ; 99(18): 7505-13, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26026938

RESUMEN

Rifamycin is a broad-spectrum antimicrobial drug produced commercially by submerged fermentation where the yields are far less in comparison to its demand in human drug therapy. Addressing the need, sequential mutational strain improvement was carried using UV and EtBr that resulted in improved strain yielding rifamycin SV up to 4.32 g/L. Further optimization of six important fermentation factors was followed which include temperature, agitation, inoculum level, period of fermentation, inorganic nitrogen source and amino acids. For the first time, we report a maximum yield of 5.32 g/L of rifamycin SV. Among the amino acids, proline known for its slowest assimilation by Amycolatopsis mediterranei produced the highest improvement in antibiotic yields. Following mutational strain improvement and process optimization, a total of 3.8-fold increase in antibiotic titre was achieved. Following a conventional procedure of mutational strain improvement, highest yield of rifamycin SV was reported by optimizing submerged fermentation process.


Asunto(s)
Actinobacteria/metabolismo , Antiinfecciosos/metabolismo , Rifamicinas/metabolismo , Actinobacteria/efectos de los fármacos , Actinobacteria/crecimiento & desarrollo , Actinobacteria/efectos de la radiación , Etidio/metabolismo , Fermentación , Ingeniería Metabólica , Mutagénesis , Selección Genética , Rayos Ultravioleta
16.
Mar Drugs ; 13(1): 249-66, 2015 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-25574739

RESUMEN

An LC-MS-based metabolomics approach was used to characterise the variation in secondary metabolite production due to changes in the salt content of the growth media as well as across different growth periods (incubation times). We used metabolomics as a tool to investigate the production of rifamycins (antibiotics) and other secondary metabolites in the obligate marine actinobacterial species Salinispora arenicola, isolated from Great Barrier Reef (GBR) sponges, at two defined salt concentrations and over three different incubation periods. The results indicated that a 14 day incubation period is optimal for the maximum production of rifamycin B, whereas rifamycin S and W achieve their maximum concentration at 29 days. A "chemical profile" link between the days of incubation and the salt concentration of the growth medium was shown to exist and reliably represents a critical point for selection of growth medium and harvest time.


Asunto(s)
Antibacterianos/aislamiento & purificación , Micromonosporaceae/química , Animales , Cromatografía Liquida , Espectrometría de Masas , Metabolómica , Micromonosporaceae/metabolismo , Poríferos/microbiología , Rifamicinas/aislamiento & purificación , Rifamicinas/metabolismo , Agua de Mar/microbiología
17.
Metab Eng ; 26: 1-16, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25149266

RESUMEN

Rifamycins are mainstay agents in treatment of many widespread diseases, but how an improved rifamycin producer can be created is still incompletely understood. Here, we describe a comparative genomic approach to investigate the mutational patterns introduced by the classical mutate-and-screen method in the genome of an improved rifamycin producer. Comparing the genome of the rifamycin B overproducer Amycolatopsis mediterranei HP-130 with those of the reference strains A. mediterranei S699 and U32, we identified 250 variations, affecting 227 coding sequences (CDS), 109 of which were HP-130-specific since they were absent in both S699 and U32. Mutational and transcriptional patterns indicated a series of genomic manipulations that not only proved the causative effect of mutB2 (coding for methylmalonyl-CoA mutase large subunit) and argS2 (coding for arginyl tRNA synthetase) mutations on the overproduction of rifamycin, but also constituted a rational strategy to genetically engineer a reference strain into an overproducer.


Asunto(s)
Actinobacteria/genética , Arginino-ARNt Ligasa/genética , Genoma Bacteriano/genética , Ingeniería Metabólica/métodos , Metilmalonil-CoA Mutasa/genética , Rifamicinas/metabolismo , Actinobacteria/clasificación , Mapeo Cromosómico/métodos , Hibridación Genómica Comparativa/métodos , Marcación de Gen/métodos , Mejoramiento Genético/métodos , Especificidad de la Especie , Regulación hacia Arriba/genética
18.
Antonie Van Leeuwenhoek ; 105(5): 849-61, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24604690

RESUMEN

A polyphasic study was carried out to establish the taxonomic status of an Atacama Desert isolate, Streptomyces strain C34(T), which synthesises novel antibiotics, the chaxalactins and chaxamycins. The organism was shown to have chemotaxonomic, cultural and morphological properties consistent with its classification in the genus Streptomyces. Analysis of 16S rRNA gene sequences showed that strain C34(T) formed a distinct phyletic line in the Streptomyces gene tree that was very loosely associated with the type strains of several Streptomyces species. Multilocus sequence analysis based on five house-keeping gene alleles underpinned the separation of strain C34(T) from all of its nearest phylogenetic neighbours, apart from Streptomyces chiangmaiensis TA-1(T) and Streptomyces hyderabadensis OU-40(T) which are not currently in the MLSA database. Strain C34(T) was distinguished readily from the S. chiangmaiensis and S. hyderabadensis strains by using a combination of cultural and phenotypic data. Consequently, strain C34(T) is considered to represent a new species of the genus Streptomyces for which the name Streptomyces leeuwenhoekii sp. nov. is proposed. The type strain is C34(T) (= DSM 42122(T) = NRRL B-24963(T)). Analysis of the whole-genome sequence of S. leeuwenhoekii, with 6,780 predicted open reading frames and a total genome size of around 7.86 Mb, revealed a high potential for natural product biosynthesis.


Asunto(s)
Antibacterianos/metabolismo , Macrólidos/metabolismo , Rifamicinas/metabolismo , Streptomyces/clasificación , Streptomyces/metabolismo , Técnicas de Tipificación Bacteriana , Análisis por Conglomerados , ADN Bacteriano/química , ADN Bacteriano/genética , ADN Ribosómico/química , ADN Ribosómico/genética , Clima Desértico , Microscopía Electrónica de Rastreo , Datos de Secuencia Molecular , Tipificación de Secuencias Multilocus , Filogenia , ARN Ribosómico 16S/genética , Microbiología del Suelo , Streptomyces/genética , Streptomyces/aislamiento & purificación
19.
Appl Microbiol Biotechnol ; 97(7): 3097-108, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23081774

RESUMEN

The developmental cycle of the obligate marine antibiotic producer actinobacterium Salinispora arenicola isolated from a Great Barrier Reef marine sponge was investigated in relation to mycelium and spore ultrastructure, synthesis of rifamycin antibiotic compounds, and expression of genes correlated with spore formation and with rifamycin precursor synthesis. The developmental cycle of S. arenicola M413 on solid agar medium was characterized by substrate mycelium growth, change of colony color, and spore formation; spore formation occurred quite early in colony growth but development of black colonies occurred only at late stages, correlated with a change in spore maturity in relation to cell wall layers. Rifamycins were detected throughout the growth cycle, but changed in relative quantity at particular phases in the cycle, with a marked increase after 32 days. Expression of the spore division gene ssgA and the rifK gene for 3-amino-5-hydroxybenzoate synthase responsible for rifamycin precursor synthesis was seen even at early stages of the growth cycle. ssgA expression significantly increased between days 26 and 31, but rifK expression effectively remained constant throughout the growth cycle, consistent with the early synthesis of rifamycin. Factors other than precursor synthesis may be responsible for an observed late increase in rifamycin production. A useful approach for measuring and exploring the regulation of antibiotic synthesis and gene expression in the marine natural product producer S. arenicola has been established.


Asunto(s)
Antibacterianos/metabolismo , Micromonosporaceae/crecimiento & desarrollo , Micromonosporaceae/metabolismo , Poríferos/microbiología , Rifamicinas/metabolismo , Animales , Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/genética , Vías Biosintéticas/genética , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/genética , Medios de Cultivo/química , Perfilación de la Expresión Génica , Micromonosporaceae/aislamiento & purificación , Micromonosporaceae/ultraestructura , Pigmentos Biológicos/metabolismo , Esporas Bacterianas/ultraestructura , Factores de Tiempo
20.
J Virol Methods ; 185(1): 156-9, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22705083

RESUMEN

Oyster mushroom spherical virus (OMSV) and oyster mushroom isometric virus (OMIV) are the causative agents of a fruiting body deformation disease in the edible mushroom Pleurotus ostreatus. The curing of these mycoviruses was facilitated by a serial transfer of infected mycelia onto a limited nutrient medium containing 1mM of cAMP and 75 µg/ml of rifamycin (cAMP-rifamycin plate). The mycelia were grown on cAMP-rifamycin plates for 5 successive passages. ELISA and RT-PCR showed that the amount of mycoviruses inside the mycelia decreased significantly with increasing numbers of passages. The mycelia became free of viruses after 5 successive passages. Cultivation of the virus-cured mycelia on a mushroom compost medium produced a normal harvest, whereas the spawn infected with viruses failed to produce any fruiting bodies.


Asunto(s)
Antivirales/metabolismo , Medios de Cultivo/química , AMP Cíclico/metabolismo , Micología/métodos , Pleurotus/virología , Rifamicinas/metabolismo , Virus/aislamiento & purificación , Ensayo de Inmunoadsorción Enzimática/métodos , Pleurotus/crecimiento & desarrollo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...