Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 315
Filtrar
1.
Microb Cell Fact ; 23(1): 103, 2024 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-38584273

RESUMO

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.


Assuntos
Ivermectina , Ivermectina/análogos & derivados , Streptomyces , Ivermectina/metabolismo , Streptomyces/metabolismo , Macrolídeos/metabolismo , Regulação Bacteriana da Expressão Gênica , Proteínas de Bactérias/metabolismo
2.
Plant Physiol Biochem ; 206: 108296, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38141401

RESUMO

The ivermectin is a potent nematocide and insecticide, which has low toxicity for humans and domestic animals, but due to low biotransformation, it can be dangerous for non-target organisms. The recent determination of ivermectin absorption and accumulation in tissues of higher plants and multiple shreds of evidence of its negative impact on plant physiology provide a basis for the search for ivermectin's molecular targets and mechanisms of action in plant cells. In this research, for the first time, the ivermectin effect on microtubules of Arabidopsis thaliana cells was studied. It was revealed that ivermectin (250 µg mL-1) disrupts the microtubule network, induces the loss of microtubule orientation, leads to microtubule curvature and shrinkage, and their longitudinal and cross-linked bundling in various cells of A. thaliana primary roots. Further, the previously proposed binding of ivermectin to the ß1-tubulin taxane site was developed and confirmed using molecular dynamics simulations of ivermectin complexes with Haemonchus contortus and A. thaliana ß1-tubulins. It was predicted that similar to other microtubule stabilizing agents ivermectin binding causes M-loop stabilization in both H. contortus and A. thaliana ß-tubulin, which leads to the enhancement of lateral contacts between subunits of adjacent protofilaments preventing microtubule depolymerization.


Assuntos
Arabidopsis , Tubulina (Proteína) , Humanos , Animais , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo , Ivermectina/farmacologia , Ivermectina/metabolismo , Arabidopsis/metabolismo , Microtúbulos/metabolismo , Sítios de Ligação
3.
Ecotoxicol Environ Saf ; 268: 115709, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37979365

RESUMO

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.


Assuntos
Oligoquetos , Poluentes do Solo , Animais , Bovinos , Feminino , Oligoquetos/metabolismo , Ivermectina/toxicidade , Ivermectina/metabolismo , Poluentes do Solo/análise , Solo , Ecotoxicologia
4.
mBio ; 14(5): e0144123, 2023 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-37702492

RESUMO

IMPORTANCE: Dengue virus (DENV) is a major human pathogen that can cause hemorrhagic fever and shock syndrome. One important factor of DENV pathogenicity is non-structural protein 1 (NS1), a glycoprotein that is secreted from infected cells. Here we study the mode of action of the widely used drug ivermectin, used to treat parasitic infections and recently shown to lower NS1 blood levels in DENV-infected patients. We found that ivermectin blocks the nuclear transport of transcription factors required for the expression of chaperones that support the folding and secretion of glycoproteins, including NS1. Impairing nuclear transport of these transcription factors by ivermectin or depleting them from infected cells dampens NS1 folding and thus its secretion. These results reveal a novel mode of action of ivermectin that might apply to other flaviviruses as well.


Assuntos
Vírus da Dengue , Dengue , Humanos , Vírus da Dengue/genética , Vírus da Dengue/metabolismo , Chaperona BiP do Retículo Endoplasmático , Ivermectina/farmacologia , Ivermectina/metabolismo , Carioferinas , Chaperonas Moleculares/metabolismo , Fatores de Transcrição/metabolismo , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo
5.
PLoS One ; 18(5): e0285262, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37141255

RESUMO

Parasitic nematodes pose a significant threat to human and animal health, as well as cause economic losses in the agricultural sector. The use of anthelmintic drugs, such as Ivermectin (IVM), to control these parasites has led to widespread drug resistance. Identifying genetic markers of resistance in parasitic nematodes can be challenging, but the free-living nematode Caenorhabditis elegans provides a suitable model. In this study, we aimed to analyze the transcriptomes of adult C. elegans worms of the N2 strain exposed to the anthelmintic drug Ivermectin (IVM), and compare them to those of the resistant strain DA1316 and the recently identified Abamectin Quantitative Trait Loci (QTL) on chromosome V. We exposed pools of 300 adult N2 worms to IVM (10-7 and 10-8 M) for 4 hours at 20°C, extracted total RNA and sequenced it on the Illumina NovaSeq6000 platform. Differentially expressed genes (DEGs) were determined using an in-house pipeline. The DEGs were compared to genes from a previous microarray study on IVM-resistant C. elegans and Abamectin-QTL. Our results revealed 615 DEGs (183 up-regulated and 432 down-regulated genes) from diverse gene families in the N2 C. elegans strain. Of these DEGs, 31 overlapped with genes from IVM-exposed adult worms of the DA1316 strain. We identified 19 genes, including the folate transporter (folt-2) and the transmembrane transporter (T22F3.11), which exhibited an opposite expression in N2 and the DA1316 strain and were deemed potential candidates. Additionally, we compiled a list of potential candidates for further research including T-type calcium channel (cca-1), potassium chloride cotransporter (kcc-2), as well as other genes such as glutamate-gated channel (glc-1) that mapped to the Abamectin-QTL.


Assuntos
Anti-Helmínticos , Ivermectina , Animais , Humanos , Ivermectina/farmacologia , Ivermectina/metabolismo , Caenorhabditis elegans/metabolismo , Transcriptoma , Locos de Características Quantitativas , Anti-Helmínticos/farmacologia , Resistência a Medicamentos/genética
6.
PLoS Pathog ; 19(3): e1011188, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36917600

RESUMO

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.


Assuntos
Copépodes , Parasitos , Ftirápteros , Animais , Ivermectina/farmacologia , Ivermectina/metabolismo , Ftirápteros/metabolismo , Parasitos/metabolismo , Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Ácido Glutâmico/farmacologia
7.
J Proteomics ; 280: 104892, 2023 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-36997062

RESUMO

Controlling Rhipicephalus microplus is among the most significant challenges for livestock production worldwide. The indiscriminate use of acaricides stimulates the selection of resistant tick populations and is therefore ineffective. Understanding the molecular foundations of resistance could help inform the search for new alternatives for tick control. Although the ovary has been suggested as a relevant target organ for tick control, there are few existing studies that focus on tick ovarian tissue. Therefore, we conducted a comparative proteomic analysis on ovaries of R. microplus strains with differential resistance to ivermectin. In resistant ticks, we observed the over-accumulation of proteins involved in several biological processes, including translation, proteolysis, transport, cellular organization, differentiation, and xenobiotic detoxification. We also observed the accumulation of many structural and extracellular proteins such as papilin-like protein, which glycosylation increase its stability-based molecular modeling. Therefore, we propose that ovaries of ivermectin-resistant ticks overcome the negative impact of ivermectin through the activation of detoxification mechanisms and structural proteins associated with the remodeling of the ovary's extracellular matrix. SIGNIFICANCE: Understanding the molecular foundation of ivermectin resistance in Rhipicephalus microplus represents an essential step in cattle farming, which could provide clues and alternatives for tick control. Excessive use of chemicals like ivermectin allows the generation of resistant tick strains in different countries. However, limited molecular information is available concerning the tick's resistance to ivermectin. Detailed proteomics scrutiny in various tick organs will provide more comprehensive molecular information. Thus, we conducted an ovary comparative proteomic-based TMT-SPS-MS3 approach. We highlight in ivermectin-resistant ticks the over-accumulation of structural proteins and enzymes connected to detoxification mechanisms.


Assuntos
Doenças dos Bovinos , Rhipicephalus , Infestações por Carrapato , Feminino , Animais , Bovinos , Ivermectina/metabolismo , Ivermectina/farmacologia , Ovário , Rhipicephalus/metabolismo , Proteômica , Xenobióticos/metabolismo , Xenobióticos/farmacologia , Infestações por Carrapato/veterinária
8.
ACS Chem Neurosci ; 14(6): 1156-1165, 2023 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-36821490

RESUMO

The α7 nicotinic acetylcholine receptor (α7nAChR) mediates signaling in the central nervous system and cholinergic anti-inflammatory pathways. Ivermectin is a positive allosteric modulator of a full-length α7nAChR and an agonist of the α7nAChR construct containing transmembrane (TMD) and intracellular (ICD) domains, but structural insights of the binding have not previously been determined. Here, combining nuclear magnetic resonance as a primary experimental tool with Rosetta comparative modeling and molecular dynamics simulations, we have revealed details of ivermectin binding to the α7nAChR TMD + ICD and corresponding structural changes in an ivermectin-induced desensitized state. Ivermectin binding was stabilized predominantly by hydrophobic interactions from interfacial residues between adjacent subunits near the extracellular end of the TMD, where the inter-subunit gap was substantially expanded in comparison to the apo structure. The ion-permeation pathway showed a profile distinctly different from the resting-state profile but similar to profiles of desensitized α7nAChR. The ICD also exhibited structural changes, including reorientation of the MX and h3 helices relative to the channel axis. The resulting structures of the α7nAChR TMD + ICD in complex with ivermectin provide opportunities for discovering new modulators of therapeutic potential and exploring the structural basis of cytoplasmic signaling under different α7nAChR functional states.


Assuntos
Ivermectina , Receptor Nicotínico de Acetilcolina alfa7 , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Ivermectina/farmacologia , Ivermectina/química , Ivermectina/metabolismo , Simulação de Dinâmica Molecular , Transdução de Sinais
9.
Artigo em Inglês | MEDLINE | ID: mdl-36470398

RESUMO

Avermectin is a widely used insecticide, and it is mainly effective against animal parasites and insects. Given its extensive use in agriculture, a large amount of avermectin is accumulated in natural waters. Avermectin is a neurotoxin that affects the autonomous behavior of zebrafish and inhibits neurological responses in invertebrates via GABA-chloride channels. In this study, we used zebrafish as a model organism to explore the lethal teratogenic effects of different avermectin concentrations. We found that 50-µg/L avermectin could cause significant malformation abnormalities during the development of zebrafish heart, changes in heart rate, and significant reduction in hatching rate and body length. Transcriptome data revealed that 499 genes were upregulated and 877 genes were downregulated at 72 h post-fertilization (hpf), whereas 1805 genes were upregulated and 836 genes were downregulated at 120 hpf. According to gene ontology (GO) enrichment analysis, avermectin affected cardiac circulation and myocardial fiber development. KEGG analysis revealed that avermectin treatment significantly altered the activity of signal pathways associated with cardiac rhythm and vascular smooth muscle contraction. The main target of avermectin was identified as the heart, as it affected heart development and function by altering cardiac-related gene expression that led to a heart defect phenotype. Our findings indicate that developing zebrafish are sensitive to avermectin, which targets the heart.


Assuntos
Cardiotoxicidade , Peixe-Zebra , Animais , Cardiotoxicidade/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Ivermectina/metabolismo , Desenvolvimento Embrionário , Embrião não Mamífero
10.
Int J Mol Sci ; 23(16)2022 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-36012413

RESUMO

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.


Assuntos
Anti-Helmínticos , Haemonchus , Animais , Animais Geneticamente Modificados , Anti-Helmínticos/farmacologia , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Resistência a Medicamentos/genética , Haemonchus/genética , Ivermectina/metabolismo , Ivermectina/farmacologia , Lactonas/metabolismo , Lactonas/farmacologia
11.
PLoS Pathog ; 18(6): e1010545, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35696434

RESUMO

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.


Assuntos
Anti-Helmínticos , Haemonchus , Nematoides , Animais , Anti-Helmínticos/farmacologia , Cloretos/metabolismo , Cloretos/farmacologia , Resistência a Medicamentos/genética , Feminino , Homeostase , Ivermectina/metabolismo , Ivermectina/farmacologia , Ivermectina/uso terapêutico , Masculino , Nematoides/genética , Plasticidade Neuronal , Ovinos/genética , Transcriptoma
12.
Biochim Biophys Acta Biomembr ; 1864(9): 183977, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35654148

RESUMO

Spin label electron paramagnetic resonance (EPR) spectroscopy was used to study the mechanisms of action of ivermectin and curcumin against Leishmania (L.) amazonensis promastigotes. EPR spectra showed that treatment of the parasites with both compounds results in plasma membrane rigidity due to oxidative processes. With the IC50 and EPR measurements for assays using different parasite concentrations, estimations could be made for the membrane-water partition coefficient (KM/W), and the concentration of the compound in the membrane (cm50) and in the aqueous phase (cw50), which inhibits cell growth by 50%. The KM/W values indicated that ivermectin has a greater affinity than curcumin for the parasite membrane. Therefore, the activity of ivermectin was higher for experiments with low cell concentrations, but for concentrations greater than 1.5 × 108 parasites/mL the compounds did not show significantly different results. The cm50 values indicated that the concentration of compound in the membrane leading to growth inhibition or membrane alteration is approximately 1 M for both ivermectin and curcumin. This high membrane concentration suggests that many ivermectin molecules per chlorine channel are needed to cause an increase in chlorine ion influx.


Assuntos
Antiprotozoários , Curcumina , Leishmania mexicana , Leishmania , Antiprotozoários/química , Antiprotozoários/farmacologia , Membrana Celular/metabolismo , Curcumina/metabolismo , Curcumina/farmacologia , Ivermectina/análise , Ivermectina/metabolismo , Ivermectina/farmacologia , Estresse Oxidativo
13.
Appl Environ Microbiol ; 88(7): e0027822, 2022 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-35323024

RESUMO

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.


Assuntos
Regulação Bacteriana da Expressão Gênica , Streptomyces , Antibacterianos/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Homeostase , Ivermectina/metabolismo , Oligomicinas/metabolismo , Metabolismo Secundário , Streptomyces/metabolismo , Zinco/metabolismo
14.
Int J Oncol ; 60(3)2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35137919

RESUMO

Glioblastoma (GBM) is one of the most widespread and lethal types of cancer. However, there are currently no drugs or therapeutic strategies that can completely cure GBM. Doramectin (DRM) has a broad range of activities against endoparasites and ectoparasites, and is extensively used in livestock. In the present study, the effect of DRM on the induction of autophagy in U87 and C6 GBM and glioma cell lines, as well as the mechanism of autophagy, were examined. First, transmission electron microscopy, plasmid transfection and western blot analysis demonstrated that DRM could induce autophagy in U87 and C6 cells in vitro. Next, MTT and colony formation assays revealed that DRM­induced autophagy prevented U87 and C6 cell viability and colony formation ratio. In addition, DRM­induced autophagy promoted U87 and C6 cell apoptosis, as indicated by DAPI analysis and flow cytometry. Furthermore, transcriptome analysis demonstrated that DRM modulated a number of genes and pathways involved in autophagy. In a nude mouse xenograft model, immunohistochemical staining and the TUNEL assay demonstrated that the effect of DRM on the tumor was consistent with that in vivo. These data indicated that DRM induced autophagy mainly by blocking the PI3K/AKT/mTOR signaling pathway in GBM cells. DRM­induced autophagy promoted the inhibition of GBM cell proliferation and apoptosis in vitro and in vivo. The present study suggested that DRM may be an effective drug for the treatment of GBM.


Assuntos
Glioblastoma/tratamento farmacológico , Ivermectina/análogos & derivados , Animais , Autofagia/efeitos dos fármacos , Autofagia/genética , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/metabolismo , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Modelos Animais de Doenças , Humanos , Ivermectina/metabolismo , Ivermectina/farmacologia , Ratos/metabolismo
15.
Phys Chem Chem Phys ; 23(40): 22957-22971, 2021 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-34636373

RESUMO

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.


Assuntos
Enzima de Conversão de Angiotensina 2/metabolismo , Antivirais/metabolismo , Proteases 3C de Coronavírus/metabolismo , Proteases Semelhantes à Papaína de Coronavírus/metabolismo , Ivermectina/metabolismo , Enzima de Conversão de Angiotensina 2/química , Antivirais/química , Sítios de Ligação , Proteases 3C de Coronavírus/química , Proteases Semelhantes à Papaína de Coronavírus/química , Quadruplex G , Humanos , Ligação de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Ivermectina/química , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Ligação Proteica , Domínios Proteicos , RNA/genética , RNA/metabolismo , SARS-CoV-2
16.
Artigo em Inglês | MEDLINE | ID: mdl-34098083

RESUMO

The pyrethroid deltamethrin and the macrocyclic lactone emamectin benzoate (EMB) are used to treat infestations of farmed salmon by parasitic salmon lice, Lepeophtheirus salmonis. While the efficacy of both compounds against Atlantic populations of the parasite has decreased as a result of the evolution of resistance, the molecular mechanisms of drug resistance in L. salmonis are currently not fully understood. The functionally diverse carboxylesterases (CaE) family includes members involved in pesticide resistance phenotypes of terrestrial arthropods. The present study had the objective to characterize the CaE family in L. salmonis and assess its role in drug resistance. L. salmonis CaE homologues were identified by homology searches in the parasite's transcriptome and genome. The transcript expression of CaEs predicted to be catalytically competent was studied using quantitative reverse-transcription PCR in drug susceptible and multi-resistant L. salmonis. The above strategy led to the identification of 21 CaEs genes/pseudogenes. Phylogenetic analyses assigned 13 CaEs to clades involved in neurodevelopmental signaling and cell adhesion, while three sequences were predicted to encode secreted enzymes. Ten CaEs were identified as being potentially catalytically competent. Transcript expression of acetylcholinesterase (ace1b) was significantly increased in multi-resistant lice compared to drug-susceptible L. salmonis, with transcript abundance further increased in preadult-II females following EMB exposure. In summary, results from the present study demonstrate that L. salmonis possesses fewer CaE gene family members than most arthropods characterized so far. Drug resistance in L. salmonis was associated with overexpression of ace1b.


Assuntos
Hidrolases de Éster Carboxílico/genética , Copépodes/enzimologia , Copépodes/genética , Regulação Enzimológica da Expressão Gênica/fisiologia , Animais , Antiparasitários/metabolismo , Antiparasitários/farmacologia , Inseticidas/metabolismo , Inseticidas/farmacologia , Ivermectina/análogos & derivados , Ivermectina/metabolismo , Ivermectina/farmacologia , Nitrilas/metabolismo , Nitrilas/farmacologia , Filogenia , Piretrinas/metabolismo , Piretrinas/farmacologia
17.
Appl Environ Microbiol ; 87(17): e0047321, 2021 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-34160269

RESUMO

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.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Choque Térmico/metabolismo , Peróxido de Hidrogênio/farmacologia , Ivermectina/análogos & derivados , Proteínas Repressoras/metabolismo , Streptomyces/crescimento & desenvolvimento , Streptomyces/metabolismo , Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Proteínas de Choque Térmico/genética , Temperatura Alta , Ivermectina/metabolismo , Regulon , Proteínas Repressoras/genética , Streptomyces/efeitos dos fármacos , Streptomyces/genética , Estresse Fisiológico
18.
Proteins ; 89(11): 1425-1441, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34169568

RESUMO

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.


Assuntos
Antivirais/metabolismo , Benzofuranos/química , Proteases 3C de Coronavírus/metabolismo , Reposicionamento de Medicamentos/métodos , Imidazóis/química , Sítio Alostérico , Antivirais/química , Antivirais/farmacologia , Benzofuranos/metabolismo , Benzofuranos/farmacologia , Sítios de Ligação , Bromocriptina/química , Bromocriptina/metabolismo , Bromocriptina/farmacologia , Proteases 3C de Coronavírus/antagonistas & inibidores , Proteases 3C de Coronavírus/química , Diosmina/química , Diosmina/metabolismo , Hidrazinas/química , Hidrazinas/metabolismo , Hidrazinas/farmacologia , Imidazóis/metabolismo , Imidazóis/farmacologia , Ivermectina/química , Ivermectina/metabolismo , Ivermectina/farmacologia , Ligantes , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Triazóis/química , Triazóis/metabolismo , Triazóis/farmacologia , Estados Unidos , United States Food and Drug Administration
19.
Arch Toxicol ; 95(5): 1535-1546, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33719007

RESUMO

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.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Inseticidas/toxicidade , Ivermectina/metabolismo , Subfamília B de Transportador de Cassetes de Ligação de ATP , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transporte Biológico , Células Cultivadas , Citocromo P-450 CYP3A , Inibidores das Enzimas do Citocromo P-450 , Interações Medicamentosas , Humanos , Hidroxilação , Ivermectina/farmacologia , Proteínas de Membrana Transportadoras , Microssomos Hepáticos , Proteína 2 Associada à Farmacorresistência Múltipla , Proteínas Associadas à Resistência a Múltiplos Medicamentos , Proteínas de Neoplasias , Preparações Farmacêuticas
20.
Carbohydr Polym ; 259: 117749, 2021 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-33673979

RESUMO

The increasing world-wide demand for food has prompted the development of efficient and environmentally friendly pesticide formulations. In this article, we have prepared CMC-g-PRSG carrier based on two compounds from natural materials carboxymethyl cellulose (CMC) and rosin (RS). The model pesticide avermectin (AVM) was encapsulated through hydrophobic interaction, and self-assembled to form nanopesticide AVM@CMC-g-PRSG with an average particle size of 167 nm. The prepared nanopesticide displays enhanced dispersibility and stability of AVM in water, and can effectively adhere to the leaves to prevent loss. The release rate of AVM encapsulated in the nanocarrier can be controlled by adjusting pH, and AVM half-life under ultraviolet radiation shows a 3-fold increase allowing control of pests for prolonged periods of time in practical applications. Biological safety tests showed that AVM@CMC-g-PRSG effectively reduces the toxicity of AVM to aquatic animals. Therefore, the cheap and degradable carrier CMC-g-PRSG can improve the effect of hydrophobic pesticides.


Assuntos
Carboximetilcelulose Sódica/química , Portadores de Fármacos/química , Resinas Vegetais/química , Animais , Liberação Controlada de Fármacos , Meia-Vida , Concentração de Íons de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Ivermectina/análogos & derivados , Ivermectina/química , Ivermectina/metabolismo , Ivermectina/farmacologia , Larva/efeitos dos fármacos , Nanopartículas/química , Tamanho da Partícula , Controle de Pragas/métodos , Praguicidas/química , Praguicidas/metabolismo , Praguicidas/farmacologia , Raios Ultravioleta , Peixe-Zebra/crescimento & desenvolvimento , Peixe-Zebra/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...