Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 320
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 19(3): e1011188, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36917600

RESUMEN

Sea louse ectoparasitosis is a major threat to fish aquaculture. Avermectins such as ivermectin and emamectin have been effectively used against sea louse infestation, but the emergence of resistance has limited their use. A better understanding of the molecular targets of avermectins is essential to the development of novel treatment strategies or new, more effective drugs. Avermectins are known to act by inhibiting neurotransmission through allosteric activation of glutamate-gated chloride channels (GluCls). We have investigated the GluCl subunit present in Caligus rogercresseyi, a sea louse affecting aquaculture in the Southern hemisphere. We identify four new subunits, CrGluCl-B to CrGluCl-E, and characterise them functionally. CrGluCl-A (previously reported as CrGluClα), CrGluCl-B and CrGluCl-C all function as glutamate channel receptors with different sensitivities to the agonist, but in contrast to subunit -A and -C, CrGluCl-B is not activated by ivermectin but is rather antagonised by the drug. CrGluCl-D channel appears active in the absence of any stimulation by glutamate or ivermectin and CrGluCl-E does not exhibit any activity. Notably, the expression of CrGluCl-B with either -A or -C subunits gives rise to receptors unresponsive to ivermectin and showing altered response to glutamate, suggesting that coexpression has led to the preferential formation of heteromers to which the presence of CrGluCl-B confers the property of ivermectin-activation refractoriness. Furthermore, there was evidence for heteromer formation with novel properties only when coexpressing pairs E/C and D/B CrGluCl subtypes. Site-directed mutagenesis shows that three transmembrane domain residues contribute to the lack of activation by ivermectin, most crucially Gln 15' in M2, with mutation Q15'T (the residue present in ivermectin-activated subunits A and C) conferring ivermectin activation to CrGluCl-B. The differential response to avermectin of these Caligus rogercresseyi GluClsubunits, which are highly conserved in the Northern hemisphere sea louse Lepeophtheirus salmonis, could have an influence on the response of these parasites to treatment with macrocyclic lactones. They could serve as molecular markers to assess susceptibility to existing treatments and might be useful molecular targets in the search for novel antiparasitic drugs.


Asunto(s)
Copépodos , Parásitos , Phthiraptera , Animales , Ivermectina/farmacología , Ivermectina/metabolismo , Phthiraptera/metabolismo , Parásitos/metabolismo , Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Ácido Glutámico/farmacología
2.
PLoS Pathog ; 18(6): e1010545, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35696434

RESUMEN

The antiparasitic drug ivermectin plays an essential role in human and animal health globally. However, ivermectin resistance is widespread in veterinary helminths and there are growing concerns of sub-optimal responses to treatment in related helminths of humans. Despite decades of research, the genetic mechanisms underlying ivermectin resistance are poorly understood in parasitic helminths. This reflects significant uncertainty regarding the mode of action of ivermectin in parasitic helminths, and the genetic complexity of these organisms; parasitic helminths have large, rapidly evolving genomes and differences in evolutionary history and genetic background can confound comparisons between resistant and susceptible populations. We undertook a controlled genetic cross of a multi-drug resistant and a susceptible reference isolate of Haemonchus contortus, an economically important gastrointestinal nematode of sheep, and ivermectin-selected the F2 population for comparison with an untreated F2 control. RNA-seq analyses of male and female adults of all populations identified high transcriptomic differentiation between parental isolates, which was significantly reduced in the F2, allowing differences associated specifically with ivermectin resistance to be identified. In all resistant populations, there was constitutive upregulation of a single gene, HCON_00155390:cky-1, a putative pharyngeal-expressed transcription factor, in a narrow locus on chromosome V previously shown to be under ivermectin selection. In addition, we detected sex-specific differences in gene expression between resistant and susceptible populations, including constitutive upregulation of a P-glycoprotein, HCON_00162780:pgp-11, in resistant males only. After ivermectin selection, we identified differential expression of genes with roles in neuronal function and chloride homeostasis, which is consistent with an adaptive response to ivermectin-induced hyperpolarisation of neuromuscular cells. Overall, we show the utility of a genetic cross to identify differences in gene expression that are specific to ivermectin selection and provide a framework to better understand ivermectin resistance and response to treatment in parasitic helminths.


Asunto(s)
Antihelmínticos , Haemonchus , Nematodos , Animales , Antihelmínticos/farmacología , Cloruros/metabolismo , Cloruros/farmacología , Resistencia a Medicamentos/genética , Femenino , Homeostasis , Ivermectina/metabolismo , Ivermectina/farmacología , Ivermectina/uso terapéutico , Masculino , Nematodos/genética , Plasticidad Neuronal , Ovinos/genética , Transcriptoma
3.
Microb Cell Fact ; 23(1): 103, 2024 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-38584273

RESUMEN

BACKGROUND: The macrolide antibiotic avermectin, a natural product derived from Streptomyces avermitilis, finds extensive applications in agriculture, animal husbandry and medicine. The mtrA (sav_5063) gene functions as a transcriptional regulator belonging to the OmpR family. As a pleiotropic regulator, mtrA not only influences the growth, development, and morphological differentiation of strains but also modulates genes associated with primary metabolism. However, the regulatory role of MtrA in avermectin biosynthesis remains to be elucidated. RESULTS: In this study, we demonstrated that MtrA, a novel OmpR-family transcriptional regulator in S. avermitilis, exerts global regulator effects by negatively regulating avermectin biosynthesis and cell growth while positively controlling morphological differentiation. The deletion of the mtrA gene resulted in an increase in avermectin production, accompanied by a reduction in biomass and a delay in the formation of aerial hyphae and spores. The Electrophoretic Mobility Shift Assay (EMSA) revealed that MtrA exhibited binding affinity towards the upstream region of aveR, the intergenic region between aveA1 and aveA2 genes, as well as the upstream region of aveBVIII in vitro. These findings suggest that MtrA exerts a negative regulatory effect on avermectin biosynthesis by modulating the expression of avermectin biosynthesis cluster genes. Transcriptome sequencing and fluorescence quantitative PCR analysis showed that mtrA deletion increased the transcript levels of the cluster genes aveR, aveA1, aveA2, aveC, aveE, aveA4 and orf-1, which explains the observed increase in avermectin production in the knockout strain. Furthermore, our findings demonstrate that MtrA positively regulates the cell division and differentiation genes bldM and ssgC, while exerting a negative regulatory effect on bldD, thereby modulating the primary metabolic processes associated with cell division, differentiation and growth in S. avermitilis, consequently impacting avermectin biosynthesis. CONCLUSIONS: In this study, we investigated the negative regulatory effect of the global regulator MtrA on avermectin biosynthesis and its effects on morphological differentiation and cell growth, and elucidated its transcriptional regulatory mechanism. Our findings indicate that MtrA plays crucial roles not only in the biosynthesis of avermectin but also in coordinating intricate physiological processes in S. avermitilis. These findings provide insights into the synthesis of avermectin and shed light on the primary and secondary metabolism of S. avermitilis mediated by OmpR-family regulators.


Asunto(s)
Ivermectina , Ivermectina/análogos & derivados , Streptomyces , Ivermectina/metabolismo , Streptomyces/metabolismo , Macrólidos/metabolismo , Regulación Bacteriana de la Expresión Génica , Proteínas Bacterianas/metabolismo
4.
World J Microbiol Biotechnol ; 40(7): 228, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38822927

RESUMEN

Doramectin, an essential animal anthelmintic, is synthesized through the fermentation process of Streptomyces avermitilis. This study delves into the transcriptomic profiles of two strains, namely the doramectin-producing wild-type S. avermitilis N72 and its highly doramectin-producing mutant counterpart, S. avermitilis XY-62. Comparative analysis revealed 860 up-regulated genes and 762 down-regulated genes in the mutant strain, notably impacting the expression of key genes pivotal in doramectin biosynthesis, including aveA1, aveA2, aveA3, aveA4, aveE, and aveBI. These findings shed light on the molecular mechanisms underpinning the heightened doramectin production in S. avermitilis XY-62, presenting promising avenues for optimizing doramectin production processes.


Asunto(s)
Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica , Ivermectina , Mutación , Streptomyces , Transcriptoma , Streptomyces/genética , Streptomyces/metabolismo , Ivermectina/análogos & derivados , Ivermectina/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Fermentación , Antihelmínticos/metabolismo
5.
Ecotoxicol Environ Saf ; 268: 115709, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37979365

RESUMEN

Ivermectin (IVM) is a dewormer commonly utilized in animal farming. Nevertheless, there is a deficiency of research on the bioecotoxicity of IVM in soil. In this study, earthworms were utilized as test animals to investigate the ecotoxicological impacts of IVM. The experiment lasted 28 days and involved adding varied doses of IVM to a culture substrate of soil mixed with cow dung and feeding it to earthworms. The experiment entailed recording earthworm weight, number of earthworm cocoons, histological damage, oxidative stress indicators, and gene expression levels. The analysis results showed that earthworm growth and reproduction were hampered by IVM. Moreover, pathological damage to the earthworms increased with increasing IVM concentration, which caused increased oxidative damage to the earthworms. These findings offer a summary of the impact of IVM on earthworms and a reference point for future research examining the ecological implications of IVM.


Asunto(s)
Oligoquetos , Contaminantes del Suelo , Animales , Bovinos , Femenino , Oligoquetos/metabolismo , Ivermectina/toxicidad , Ivermectina/metabolismo , Contaminantes del Suelo/análisis , Suelo , Ecotoxicología
6.
Appl Environ Microbiol ; 88(7): e0027822, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35323024

RESUMEN

Zinc is an essential cofactor for many metal enzymes and transcription regulators. Zn2+ availability has long been known to affect antibiotic production and morphological differentiation of Streptomyces species. However, the molecular mechanism whereby zinc regulates these processes remains unclear. We investigated the regulatory roles of the zinc-sensing regulator Zur in Streptomyces avermitilis. Our findings demonstrate that Zur plays an essential role in maintaining zinc homeostasis by repressing the expression of the zinc uptake system ZnuACB and alternative non-zinc-binding ribosomal proteins and promoting the expression of zinc exporter ZitB. Deletion of the zur gene resulted in decreased production of avermectin and oligomycin and delayed morphological differentiation, and these parameters were restored close to wild-type levels in a zur-complemented strain. Zur bound specifically to Zur box in the promoter regions of avermectin pathway-specific activator gene aveR, oligomycin polyketide synthase gene olmA1, and filipin biosynthetic pathway-specific regulatory genes pteR and pteF. Analyses by reverse transcription quantitative PCR and luciferase reporter systems indicated that Zur directly activates the transcription of these genes, i.e., that Zur directly activates biosynthesis of avermectin and oligomycin. Zur positively regulated morphological development by repressing the transcription of differentiation-related genes ssgB and minD2. Our findings, taken together, demonstrate that Zur in S. avermitilis directly controls zinc homeostasis, biosynthesis of avermectin and oligomycin, and morphological differentiation. IMPORTANCE Biosynthesis of secondary metabolites and morphological differentiation in bacteria are affected by environmental signals. The molecular mechanisms whereby zinc availability affects secondary metabolism and morphological differentiation remain poorly understood. We identified several new target genes of the zinc response regulator Zur in Streptomyces avermitilis, the industrial producer of avermectin. Zur was found to directly and positively control avermectin production, oligomycin production, and morphological differentiation in response to extracellular Zn2+ levels. Our findings clarify the regulatory functions of Zur in Streptomyces, which involve linking environmental Zn2+ status with control of antibiotic biosynthetic pathways and morphological differentiation.


Asunto(s)
Regulación Bacteriana de la Expresión Génica , Streptomyces , Antibacterianos/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Homeostasis , Ivermectina/metabolismo , Oligomicinas/metabolismo , Metabolismo Secundario , Streptomyces/metabolismo , Zinc/metabolismo
7.
PLoS Comput Biol ; 17(2): e1007856, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33571182

RESUMEN

Pentameric ligand-gated ion channels (pLGICs) are receptor proteins that are sensitive to their membrane environment, but the mechanism for how lipids modulate function under physiological conditions in a state dependent manner is not known. The glycine receptor is a pLGIC whose structure has been resolved in different functional states. Using a realistic model of a neuronal membrane coupled with coarse-grained molecular dynamics simulations, we demonstrate that some key lipid-protein interactions are dependent on the receptor state, suggesting that lipids may regulate the receptor's conformational dynamics. Comparison with existing structural data confirms known lipid binding sites, but we also predict further protein-lipid interactions including a site at the communication interface between the extracellular and transmembrane domain. Moreover, in the active state, cholesterol can bind to the binding site of the positive allosteric modulator ivermectin. These protein-lipid interaction sites could in future be exploited for the rational design of lipid-like allosteric drugs.


Asunto(s)
Canales Iónicos Activados por Ligandos/metabolismo , Modelos Neurológicos , Neuronas/metabolismo , Sitio Alostérico , Animales , Sitios de Unión , Colesterol/química , Colesterol/metabolismo , Biología Computacional , Humanos , Ivermectina/química , Ivermectina/metabolismo , Canales Iónicos Activados por Ligandos/química , Lípidos de la Membrana/química , Lípidos de la Membrana/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Simulación de Dinámica Molecular , Unión Proteica , Dominios Proteicos , Estructura Cuaternaria de Proteína , Receptores de Glicina/química , Receptores de Glicina/metabolismo
8.
Int J Mol Sci ; 23(16)2022 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-36012413

RESUMEN

The number of reported macrocyclic lactones (ML) resistance cases across all livestock hosts is steadily increasing. Different studies in the parasitic nematode Haemonchus contortus assume the participation of cytochrome P450s (Cyps) enzymes in ML resistance. Still, functional data about their individual contribution to resistance or substrate specificity is missing. Via microinjection, transgenic Caenorhabditis elegans expressing HCON_00141052 (transgene-Hco-cyp-13A11) from extrachromosomal arrays were generated. After 24 h of exposure to different concentrations of ivermectin (IVM), ivermectin aglycone (IVMa), selamectin (SEL), doramectin (DRM), eprinomectin (EPR), and moxidectin (MOX), motility assays were performed to determine the impact of the H. contortus Cyp to the susceptibility of the worms against each ML. While transgene-Hco-cyp-13A11 significantly decreased susceptibility to IVM (four-fold), IVMa (2-fold), and SEL (3-fold), a slight effect for DRM and no effect for MOX, and EPR was observed. This substrate specificity of Hco-cyp-13A11 could not be explained by molecular modeling and docking studies. Hco-Cyp-13A11 molecular models were obtained for alleles from isolates with different resistance statuses. Although 14 amino acid polymorphisms were detected, none was resistance specific. In conclusion, Hco-cyp-13A11 decreased IVM, IVMa, and SEL susceptibility to a different extent, but its potential impact on ML resistance is not driven by polymorphisms.


Asunto(s)
Antihelmínticos , Haemonchus , Animales , Animales Modificados Genéticamente , Antihelmínticos/farmacología , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Resistencia a Medicamentos/genética , Haemonchus/genética , Ivermectina/metabolismo , Ivermectina/farmacología , Lactonas/metabolismo , Lactonas/farmacología
9.
Proteins ; 89(11): 1425-1441, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34169568

RESUMEN

The novel coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) still has serious negative effects on health, social life, and economics. Recently, vaccines from various companies have been urgently approved to control SARS-CoV-2 infections. However, any specific antiviral drug has not been confirmed so far for regular treatment. An important target is the main protease (Mpro ), which plays a major role in replication of the virus. In this study, Gaussian and residue network models are employed to reveal two distinct potential allosteric sites on Mpro that can be evaluated as drug targets besides the active site. Then, Food and Drug Administration (FDA)-approved drugs are docked to three distinct sites with flexible docking using AutoDock Vina to identify potential drug candidates. Fourteen best molecule hits for the active site of Mpro are determined. Six of these also exhibit high docking scores for the potential allosteric regions. Full-atom molecular dynamics simulations with MM-GBSA method indicate that compounds docked to active and potential allosteric sites form stable interactions with high binding free energy (∆Gbind ) values. ∆Gbind values reach -52.06 kcal/mol for the active site, -51.08 kcal/mol for the potential allosteric site 1, and - 42.93 kcal/mol for the potential allosteric site 2. Energy decomposition calculations per residue elucidate key binding residues stabilizing the ligands that can further serve to design pharmacophores. This systematic and efficient computational analysis successfully determines ivermectine, diosmin, and selinexor currently subjected to clinical trials, and further proposes bromocriptine, elbasvir as Mpro inhibitor candidates to be evaluated against SARS-CoV-2 infections.


Asunto(s)
Antivirales/metabolismo , Benzofuranos/química , Proteasas 3C de Coronavirus/metabolismo , Reposicionamiento de Medicamentos/métodos , Imidazoles/química , Sitio Alostérico , Antivirales/química , Antivirales/farmacología , Benzofuranos/metabolismo , Benzofuranos/farmacología , Sitios de Unión , Bromocriptina/química , Bromocriptina/metabolismo , Bromocriptina/farmacología , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Proteasas 3C de Coronavirus/química , Diosmina/química , Diosmina/metabolismo , Hidrazinas/química , Hidrazinas/metabolismo , Hidrazinas/farmacología , Imidazoles/metabolismo , Imidazoles/farmacología , Ivermectina/química , Ivermectina/metabolismo , Ivermectina/farmacología , Ligandos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Triazoles/química , Triazoles/metabolismo , Triazoles/farmacología , Estados Unidos , United States Food and Drug Administration
10.
Mol Microbiol ; 113(1): 123-142, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31628680

RESUMEN

BldD generally functions as a repressor controlling morphological development of Streptomyces. In this work, evidences that BldD also activates antibiotic production are provided. In Streptomyces roseosporus (which produces daptomycin widely used for treatment of human infections), deletion of bldD notably reduced daptomycin production, but enhanced sporulation. BldD stimulated daptomycin production by directly activating transcription of dpt structural genes and dptR3 (which encodes an indirect activator of daptomycin production), and repressed its own gene. BldD-binding sites on promoter regions of dptE, dptR3, and bldD were all found to contain BldD box-like sequences, facilitating prediction of new BldD targets. Two Streptomyces global regulatory genes, adpA and afsR, were confirmed to be directly activated by BldD. The protein AfsR was shown to act as an activator of daptomycin production, but a repressor of development. BldD directly represses nine key developmental genes. In Streptomyces avermitilis (which produces effective anthelmintic agents avermectins), BldD homolog (BldDsav) directly activates avermectin production through ave structural genes and cluster-situated activator gene aveR. This is the first report that BldD activates antibiotic biosynthesis both directly and via a cascade mechanism. BldD homologs are widely distributed among Streptomyces, our findings suggest that BldD may activate antibiotic production in other Streptomyces species.


Asunto(s)
Antibacterianos/biosíntesis , Proteínas Bacterianas/metabolismo , Proteínas de Unión al ADN/metabolismo , Daptomicina/biosíntesis , Streptomyces/metabolismo , Factores de Transcripción/metabolismo , Regulación Bacteriana de la Expresión Génica , Ivermectina/análogos & derivados , Ivermectina/metabolismo , Streptomyces/genética , Streptomyces/crecimiento & desarrollo
11.
PLoS Pathog ; 15(2): e1007598, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30759156

RESUMEN

Resistance to the anthelmintic macrocyclic lactone ivermectin (IVM) has a great impact on the control of parasitic nematodes. The mechanisms by which nematodes adapt to IVM remain to be deciphered. We have identified NHR-8, a nuclear hormone receptor involved in the xenobiotic response in Caenorhabditis elegans, as a new regulator of tolerance to IVM. Loss-of-function nhr-8(ok186) C. elegans mutants subjected to larval development assays and electropharyngeogram measurements, displayed hypersensitivity to IVM, and silencing of nhr-8 in IVM-resistant worms increased IVM efficacy. In addition, compared to wild-type worms, nhr-8 mutants under IVM selection pressure failed to acquire tolerance to the drug. In addition, IVM-hypersensitive nhr-8(ok186) worms displayed low transcript levels of several genes from the xenobiotic detoxification network and a concomitant low Pgp-mediated drug efflux activity. Interestingly, some pgp and cyp genes known to impact IVM tolerance in many nematode species, were down regulated in nhr-8 mutants and inversely upregulated in IVM-resistant worms. Moreover, pgp-6 overexpression in nhr-8(ok186) C. elegans increased tolerance to IVM. Importantly, NHR-8 function was rescued in nhr-8(ok186) C. elegans with the homolog of the parasitic nematode Haemonchus contortus, and silencing of Hco-nhr-8 by RNAi on L2 H. contortus larvae increased IVM susceptibility in both susceptible and resistant H. contortus isolates. Thus, our data show that NHR-8 controls the tolerance and development of resistance to IVM in C. elegans and the molecular basis for this relates to the NHR-8-mediated upregulation of IVM detoxification genes. Since our results show that Hco-nhr-8 functions similarly to Cel-nhr-8, this study helps to better understand mechanisms underlying failure in drug efficacy and open perspectives in finding new compounds with NHR-8 antagonist activity to potentiate IVM efficacy.


Asunto(s)
Proteínas de Caenorhabditis elegans/efectos de los fármacos , Proteínas de Caenorhabditis elegans/metabolismo , Ivermectina/metabolismo , Receptores Citoplasmáticos y Nucleares/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Antihelmínticos , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/fisiología , Resistencia a Medicamentos , Regulación de la Expresión Génica/efectos de los fármacos , Haemonchus , Ivermectina/farmacología , Larva , Infecciones por Nematodos/virología , Receptores Citoplasmáticos y Nucleares/fisiología , Factores de Transcripción/efectos de los fármacos , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/efectos de los fármacos
12.
Appl Environ Microbiol ; 87(17): e0047321, 2021 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-34160269

RESUMEN

The heat shock response (HSR) is a universal cellular response that promotes survival following temperature increase. In filamentous Streptomyces, which accounts for ∼70% of commercial antibiotic production, HSR is regulated by transcriptional repressors; in particular, the widespread MerR-family regulator HspR has been identified as a key repressor. However, functions of HspR in other biological processes are unknown. The present study demonstrates that HspR pleiotropically controls avermectin production, morphological development, and heat shock and H2O2 stress responses in the industrially important species Streptomyces avermitilis. HspR directly activated ave structural genes (aveA1 and aveA2) and H2O2 stress-related genes (katA1, catR, katA3, oxyR, ahpC, and ahpD), whereas it directly repressed heat shock genes (HSGs) (the dnaK1-grpE1-dnaJ1-hspR operon, clpB1p, clpB2p, and lonAp) and developmental genes (wblB, ssgY, and ftsH). HspR interacted with PhoP (response regulator of the widespread PhoPR two-component system) at dnaK1p to corepress the important dnaK1-grpE1-dnaJ1-hspR operon. PhoP exclusively repressed target HSGs (htpG, hsp18_1, and hsp18_2) different from those of HspR (clpB1p, clpB2p, and lonAp). A consensus HspR-binding site, 5'-TTGANBBNNHNNNDSTSHN-3', was identified within HspR target promoter regions, allowing prediction of the HspR regulon involved in broad cellular functions. Taken together, our findings demonstrate a key role of HspR in the coordination of a variety of important biological processes in Streptomyces species. IMPORTANCE Our findings are significant to clarify the molecular mechanisms underlying HspR function in Streptomyces antibiotic production, development, and H2O2 stress responses through direct control of its target genes associated with these biological processes. HspR homologs described to date function as transcriptional repressors but not as activators. The results of the present study demonstrate that HspR acts as a dual repressor/activator. PhoP cross talks with HspR at dnaK1p to coregulate the heat shock response (HSR), but it also has its own specific target heat shock genes (HSGs). The novel role of PhoP in the HSR further demonstrates the importance of this regulator in Streptomyces. Overexpression of hspR strongly enhanced avermectin production in Streptomyces avermitilis wild-type and industrial strains. These findings provide new insights into the regulatory roles and mechanisms of HspR and PhoP and facilitate methods for antibiotic overproduction in Streptomyces species.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas de Choque Térmico/metabolismo , Peróxido de Hidrógeno/farmacología , Ivermectina/análogos & derivados , Proteínas Represoras/metabolismo , Streptomyces/crecimiento & desarrollo , Streptomyces/metabolismo , Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Proteínas de Choque Térmico/genética , Calor , Ivermectina/metabolismo , Regulón , Proteínas Represoras/genética , Streptomyces/efectos de los fármacos , Streptomyces/genética , Estrés Fisiológico
13.
Arch Biochem Biophys ; 698: 108677, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33197431

RESUMEN

We investigate the effect of the alcohol-induced increase in the content of CYP2E1 in human liver microsomes (HLM) on the function of CYP3A4. Membrane incorporation of the purified CYP2E1 into HLM considerably increases the rate of metabolism of 7-benzyloxyquinoline (BQ) and attenuates the homotropic cooperativity observed with this CYP3A4-specific substrate. It also eliminates the activating effect of α-naphthoflavone (ANF) seen in some HLM samples. To probe the physiological relevance of these effects, we compared three pooled preparations of HLM from normal donors (HLM-N) with a pooled preparation from ten heavy alcohol consumers (HLM-A). The composition of the P450 pool in all samples was characterized by the mass-spectrometric determination of 11 cytochrome P450 species. The fractional content of CYP2E1 in HLM-A was from 2.0 to 3.4 times higher than in HLM-N. In contrast, the content of CYP3A4 in HLM-A was the lowest among all samples. Despite that, HLM-A exhibited a much higher metabolism rate and a lower homotropic cooperativity with BQ, similar to CYP2E1-enriched HLM-N. To substantiate the involvement of interactions between CYP2E1 and CYP3A4 in these effects, we probed hetero-association of these proteins in CYP3A4-containing Supersomes™ with a technique employing CYP2E1 labeled with BODIPY-618 maleimide. These experiments evinced the interactions between the two enzymes and revealed an inhibitory effect of ANF on their association. Our results demonstrate that the functional properties of CYP3A4 are fundamentally dependent on the composition of the cytochrome P450 ensemble and suggest a possible impact of chronic alcohol exposure on the pharmacokinetics of drugs metabolized by CYP3A4.


Asunto(s)
Citocromo P-450 CYP2E1/metabolismo , Citocromo P-450 CYP3A/metabolismo , Etanol/toxicidad , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Secuencia de Aminoácidos , Amitriptilina/metabolismo , Benzoflavonas/farmacología , Citocromo P-450 CYP2E1/análisis , Citocromo P-450 CYP3A/análisis , Activadores de Enzimas/farmacología , Femenino , Humanos , Ivermectina/metabolismo , Masculino , Midazolam/metabolismo , Nitrofenoles/metabolismo , Quinolinas/metabolismo
14.
Phys Chem Chem Phys ; 23(40): 22957-22971, 2021 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-34636373

RESUMEN

The identification of chemical compounds able to bind specific sites of the human/viral proteins involved in the SARS-CoV-2 infection cycle is a prerequisite to design effective antiviral drugs. Here we conduct a molecular dynamics study with the aim to assess the interactions of ivermectin, an antiparasitic drug with broad-spectrum antiviral activity, with the human Angiotensin-Converting Enzyme 2 (ACE2), the viral 3CLpro and PLpro proteases, and the viral SARS Unique Domain (SUD). The drug/target interactions have been characterized in silico by describing the nature of the non-covalent interactions found and by measuring the extent of their time duration along the MD simulation. Results reveal that the ACE2 protein and the ACE2/RBD aggregates form the most persistent interactions with ivermectin, while the binding with the remaining viral proteins is more limited and unspecific.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Antivirales/metabolismo , Proteasas 3C de Coronavirus/metabolismo , Proteasas Similares a la Papaína de Coronavirus/metabolismo , Ivermectina/metabolismo , Enzima Convertidora de Angiotensina 2/química , Antivirales/química , Sitios de Unión , Proteasas 3C de Coronavirus/química , Proteasas Similares a la Papaína de Coronavirus/química , G-Cuádruplex , Humanos , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Ivermectina/química , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Unión Proteica , Dominios Proteicos , ARN/genética , ARN/metabolismo , SARS-CoV-2
15.
Nature ; 526(7572): 224-9, 2015 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-26344198

RESUMEN

The strychnine-sensitive glycine receptor (GlyR) mediates inhibitory synaptic transmission in the spinal cord and brainstem and is linked to neurological disorders, including autism and hyperekplexia. Understanding of molecular mechanisms and pharmacology of glycine receptors has been hindered by a lack of high-resolution structures. Here we report electron cryo-microscopy structures of the zebrafish α1 GlyR with strychnine, glycine, or glycine and ivermectin (glycine/ivermectin). Strychnine arrests the receptor in an antagonist-bound closed ion channel state, glycine stabilizes the receptor in an agonist-bound open channel state, and the glycine/ivermectin complex adopts a potentially desensitized or partially open state. Relative to the glycine-bound state, strychnine expands the agonist-binding pocket via outward movement of the C loop, promotes rearrangement of the extracellular and transmembrane domain 'wrist' interface, and leads to rotation of the transmembrane domain towards the pore axis, occluding the ion conduction pathway. These structures illuminate the GlyR mechanism and define a rubric to interpret structures of Cys-loop receptors.


Asunto(s)
Microscopía por Crioelectrón , Receptores de Glicina/metabolismo , Receptores de Glicina/ultraestructura , Pez Cebra , Regulación Alostérica , Animales , Sitios de Unión , Glicina/metabolismo , Glicina/farmacología , Activación del Canal Iónico/efectos de los fármacos , Ivermectina/metabolismo , Ivermectina/farmacología , Modelos Moleculares , Neurotransmisores/metabolismo , Neurotransmisores/farmacología , Conformación Proteica/efectos de los fármacos , Subunidades de Proteína/química , Subunidades de Proteína/efectos de los fármacos , Subunidades de Proteína/metabolismo , Receptores de Glicina/agonistas , Receptores de Glicina/antagonistas & inhibidores , Rotación , Transducción de Señal , Estricnina/metabolismo , Estricnina/farmacología
16.
Arch Toxicol ; 95(5): 1535-1546, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33719007

RESUMEN

The review presents metabolic properties of Ivermectin (IVM) as substrate and inhibitor of human P450 (P450, CYP) enzymes and drug transporters. IVM is metabolized, both in vivo and in vitro, by C-hydroxylation and O-demethylation reactions catalyzed by P450 3A4 as the major enzyme, with a contribution of P450 3A5 and 2C9. In samples from both in vitro and in vivo metabolism, a number of metabolites were detected and as major identified metabolites were 3″-O-demethylated, C4-methyl hydroxylated, C25 isobutyl-/isopropyl-hydroxylated, and products of oxidation reactions. Ivermectin inhibited P450 2C9, 2C19, 2D6, and CYP3A4 with IC50 values ranging from 5.3 µM to no inhibition suggesting that it is no or weak inhibitor of the enzymes. It is suggested that P-gp (MDR1) transporter participate in IVM efflux at low drug concentration with a slow transport rate. At the higher, micromolar concentration range, which saturates MDR1 (P-gp), MRP1, and to a lesser extent, MRP2 and MRP3 participate in IVM transport across physiological barriers. IVM exerts a potent inhibition of P-gp (ABCB1), MRP1 (ABCC1), MRP2 (ABCC2), and BCRP1 (ABCG2), and medium to weak inhibition of OATP1B1 (SLC21A6) and OATP1B3 (SLCOB3) transport activity. The metabolic and transport properties of IVM indicate that when IVM is co-administered with other drugs/chemicals that are potent inhibitors/inducers P4503A4 enzyme and of MDR1 (P-gp), BCRP or MRP transporters, or when polymorphisms of the drug transporters and P450 3A4 exist, drug-drug or drug-toxic chemical interactions might result in suboptimal response to the therapy or to toxic effects.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Insecticidas/toxicidad , Ivermectina/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transporte Biológico , Células Cultivadas , Citocromo P-450 CYP3A , Inhibidores Enzimáticos del Citocromo P-450 , Interacciones Farmacológicas , Humanos , Hidroxilación , Ivermectina/farmacología , Proteínas de Transporte de Membrana , Microsomas Hepáticos , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos , Proteínas de Neoplasias , Preparaciones Farmacéuticas
17.
Nature ; 512(7514): 333-7, 2014 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-25143115

RESUMEN

Cys-loop receptors are neurotransmitter-gated ion channels that are essential mediators of fast chemical neurotransmission and are associated with a large number of neurological diseases and disorders, as well as parasitic infections. Members of this ion channel superfamily mediate excitatory or inhibitory neurotransmission depending on their ligand and ion selectivity. Structural information for Cys-loop receptors comes from several sources including electron microscopic studies of the nicotinic acetylcholine receptor, high-resolution X-ray structures of extracellular domains and X-ray structures of bacterial orthologues. In 2011 our group published structures of the Caenorhabditis elegans glutamate-gated chloride channel (GluCl) in complex with the allosteric partial agonist ivermectin, which provided insights into the structure of a possibly open state of a eukaryotic Cys-loop receptor, the basis for anion selectivity and channel block, and the mechanism by which ivermectin and related molecules stabilize the open state and potentiate neurotransmitter binding. However, there remain unanswered questions about the mechanism of channel opening and closing, the location and nature of the shut ion channel gate, the transitions between the closed/resting, open/activated and closed/desensitized states, and the mechanism by which conformational changes are coupled between the extracellular, orthosteric agonist binding domain and the transmembrane, ion channel domain. Here we present two conformationally distinct structures of C. elegans GluCl in the absence of ivermectin. Structural comparisons reveal a quaternary activation mechanism arising from rigid-body movements between the extracellular and transmembrane domains and a mechanism for modulation of the receptor by phospholipids.


Asunto(s)
Apoproteínas/química , Caenorhabditis elegans/química , Canales de Cloruro/química , Canales de Cloruro/metabolismo , Receptores de Canales Iónicos con Asa de Cisteína Activados por Ligando/química , Receptores de Canales Iónicos con Asa de Cisteína Activados por Ligando/metabolismo , Regulación Alostérica/efectos de los fármacos , Animales , Apoproteínas/metabolismo , Sitios de Unión , Unión Competitiva/efectos de los fármacos , Membrana Celular/metabolismo , Cristalografía por Rayos X , Agonismo Parcial de Drogas , Ácido Glutámico/metabolismo , Activación del Canal Iónico , Ivermectina/química , Ivermectina/metabolismo , Ivermectina/farmacología , Ligandos , Modelos Moleculares , Movimiento/efectos de los fármacos , Fosfatidilcolinas/química , Fosfatidilcolinas/metabolismo , Fosfatidilcolinas/farmacología , Unión Proteica , Multimerización de Proteína/efectos de los fármacos , Estructura Terciaria de Proteína/efectos de los fármacos , Relación Estructura-Actividad
18.
Molecules ; 25(3)2020 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-31979297

RESUMEN

The experiment developed the ultra-high-performance liquid chromatography-tandem mass spectrometry (UPLC/MS/MS) method for testing emamectin benzoate, and studied the metabolism of emamectin benzoate in rice plants and rice-growing environments via application of this testing method. The dissipation curve of emamectin benzoate standard substance was good at 0.5-200 µg L-1, and its correlation coefficient was greater than 0.99. In the concentration range of 0.1-50 µg kg-1, the average recovery rate of plants, soil, and field water was 82 %-102 %, and relative standard deviation (RSD) was between 0.3 % and 15.9 %. Half-lives in rice plants and soil were 0.8-2.8 days and 1.9-3.8 days, respectively, and emamectin benzoate was not detected in rice or rice hull. The experiment showed that emamectin benzoate is harmless to human health at the concentration recommended by the manufacturer.


Asunto(s)
Cromatografía Liquida/métodos , Ivermectina/análogos & derivados , Oryza/química , Residuos de Plaguicidas/análisis , Suelo/química , Espectrometría de Masas en Tándem/métodos , Monitoreo del Ambiente/métodos , Ivermectina/química , Ivermectina/metabolismo , Tallos de la Planta/química , Sensibilidad y Especificidad
19.
Bioorg Med Chem ; 27(12): 2387-2396, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-30626556

RESUMEN

A series of new doramectin derivatives containing carbamate, ester and sulfonate were synthesized, and their structures were characterized by 1H and 13C nuclear magnetic resonance (NMR) and high-resolution mass spectrum (HRMS). Their insecticidal activities against oriental armyworm, diamondback moth, and corn borer were evaluated and compared with the parent doramectin and commercial avermectins, metolcarb, fenpropathrin. Among all compounds, three compounds (3a, 3g and 3h) showed excellent insecticidal effect. In particular, compound 3g containing cyclopropyl carbamate against oriental armyworm, diamondback moth, and corn borer, exhibited the most promising insecticidal activity with the final mortality rate of 66.67%, 36.67%, 40.00% at the concentration of 12.5 mg/L, respectively. The LC50 values of 3g were 5.8859, 22.3214, and 22.0205 mg/L, showing 6.74, 2.23, 2.21-fold higher potency than parent doramectin (LC50 values of 39.6907, 49.7736, and 48.6129 mg/L) and 6.83, 1.93, 3.36-fold higher potency than commercial avermectins (LC50 values of 40.2489, 42.9922, and 73.9508 mg/L). Additionally, molecular docking simulations revealed that 3g displayed stronger hydrogen-bonding action in binding with the GABA receptor than parent doramectin, which were crucial for keeping high insecticidal activity. The present work demonstrated that these compounds containing alkyl carbamate group could be considered as potential candidates for the development of novel pesticides in the future.


Asunto(s)
Insecticidas/toxicidad , Ivermectina/análogos & derivados , Animales , Sitios de Unión , Diseño de Fármacos , Insecticidas/síntesis química , Insecticidas/metabolismo , Ivermectina/síntesis química , Ivermectina/metabolismo , Ivermectina/toxicidad , Simulación del Acoplamiento Molecular , Estructura Molecular , Mariposas Nocturnas/efectos de los fármacos , Fenilcarbamatos/toxicidad , Piretrinas/toxicidad , Receptores de GABA/química , Receptores de GABA/metabolismo , Relación Estructura-Actividad
20.
Appl Microbiol Biotechnol ; 103(20): 8459-8472, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31422450

RESUMEN

Streptomyces avermitilis is well known as the producer of anthelmintic agent avermectins, which are widely used in agriculture, veterinary medicine, and human medicine. aveI encodes a TetR-family regulator, which is the homolog of AtrA. It was reported that deletion of aveI caused enhanced avermectin production. In this study, we investigated the regulatory function of the AveI in S. avermitilis. By binding to the 15-nt palindromic sequence in the promoter regions, AveI directly regulates at least 35 genes. AveI represses avermectin production by directly regulating the transcription of the cluster-situated regulator gene aveR and structural genes aveA1, aveA3, and aveD. AveI represses oligomycin production by repressing the CSR gene olmRII and structural genes olmC. AveI activates melanin biosynthesis by activating the expression of melC1C2 operon. AveI activates morphological differentiation by activating the expression of ssgR and ssgD genes, repressing the expression of wblI gene. Besides, AveI regulates many genes involved in primary metabolism, including substrates transport, the metabolism of amino acids, lipids, and carbohydrates. Therefore, AveI functions as a global regulator in S. avermitilis, controls not only secondary metabolism and morphological differentiation, but also primary metabolism.


Asunto(s)
Productos Biológicos/metabolismo , Regulación Bacteriana de la Expresión Génica , Ivermectina/análogos & derivados , Melaninas/metabolismo , Oligomicinas/metabolismo , Streptomyces/metabolismo , Factores de Transcripción/metabolismo , Ivermectina/metabolismo , Streptomyces/citología , Streptomyces/genética , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA