Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 10(5): e1004116, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24809504

RESUMEN

Treatment and control of schistosomiasis relies on the only available drug, praziquantel, and the search for alternative chemotherapeutic agents is therefore urgent. Egg production is required for the transmission and immunopathology of schistosomiasis and females of S. mansoni lay 300 eggs daily. A large fraction of the total mRNA in the mature female worm encodes one eggshell protein, Smp14. We report that the nuclear receptors SmRXR1 and SmNR1 regulate Smp14 transcription through the recruitment of two histone acetyltransferases (HATs), SmGCN5 and SmCBP1. The treatment of HEK293 cells with histone deacetylase (HDAC) inhibitors (NaB or TSA) produced an 8-fold activation of the SmRXR1/SmNR1-mediated Smp14 promoter activity. Incubation with synthetic HAT inhibitors, including PU139, significantly impaired the Smp14 promoter activity in these cells. Worm pairs cultivated in the presence of PU139 exhibited limited expression of Smp14 mRNA and protein. ChIP analysis demonstrated chromatin condensation at the Smp14 promoter site in worms treated with PU139. ChIP also revealed the presence of H3K27me3 and the absence of RNA Pol II at the Smp14 promoter region in the PU139-treated worms. Most significantly, the PU139-mediated inhibition of Smp14 expression resulted in a significant number of abnormal eggs as well as defective eggs within the ootype. In addition, scanning electron microscopy revealed structural defects and unformed eggshells, and vitelline cell leakage was apparent. The dsRNAi-targeting of SmGCN5 or SmCBP1 significantly decreased Smp14 transcription and protein synthesis, which compromised the reproductive system of mature female worms, egg-laying and egg morphology. Our data strongly suggest that the inhibition of Smp14 expression targeting SmGCN5 and/or SmCBP1 represents a novel and effective strategy to control S. mansoni egg development.


Asunto(s)
Antihelmínticos/uso terapéutico , Proteínas del Huevo/genética , Epigénesis Genética/fisiología , Oviparidad/genética , Schistosoma mansoni/fisiología , Esquistosomiasis/prevención & control , Esquistosomiasis/transmisión , Animales , Proteínas del Huevo/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Células HEK293 , Proteínas del Helminto/genética , Proteínas del Helminto/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Histona Desacetilasas/uso terapéutico , Humanos , Masculino , Terapia Molecular Dirigida/métodos , Schistosoma mansoni/genética , Schistosoma mansoni/crecimiento & desarrollo
2.
Proc Natl Acad Sci U S A ; 110(21): 8627-31, 2013 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-23641003

RESUMEN

Several organisms have retained methyltransferase 2 (Dnmt2) as their only candidate DNA methyltransferase gene. However, information about Dnmt2-dependent methylation patterns has been limited to a few isolated loci and the results have been discussed controversially. In addition, recent studies have shown that Dnmt2 functions as a tRNA methyltransferase, which raised the possibility that Dnmt2-only genomes might be unmethylated. We have now used whole-genome bisulfite sequencing to analyze the methylomes of Dnmt2-only organisms at single-base resolution. Our results show that the genomes of Schistosoma mansoni and Drosophila melanogaster lack detectable DNA methylation patterns. Residual unconverted cytosine residues shared many attributes with bisulfite deamination artifacts and were observed at comparable levels in Dnmt2-deficient flies. Furthermore, genetically modified Dnmt2-only mouse embryonic stem cells lost the DNA methylation patterns found in wild-type cells. Our results thus uncover fundamental differences among animal methylomes and suggest that DNA methylation is dispensable for a considerable number of eukaryotic organisms.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/fisiología , ADN Protozoario/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas Protozoarias/metabolismo , Schistosoma mansoni/enzimología , Animales , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Protozoario/genética , Proteínas de Drosophila/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/enzimología , Ratones , Ratones Noqueados , Proteínas Protozoarias/genética , Schistosoma mansoni/genética
3.
Clin Transl Sci ; 16(4): 631-646, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36631939

RESUMEN

The severe acute respiratory syndrome coronavirus 2, the agent of the ongoing coronavirus disease 2019 (COVID-19) pandemic, has spread worldwide since it was first identified in November 2019 in Wuhan, China. Since then, progress in pathogenesis linked severity of this systemic disease to the hyperactivation of network of cytokine-driven pro-inflammatory cascades. Here, we aimed to identify molecular biomarkers of disease severity by measuring the serum levels of inflammatory mediators in a Brazilian cohort of patients with COVID-19 and healthy controls (HCs). Critically ill patients in the intensive care unit were defined as such by dependence on oxygen supplementation (93% intubated and 7% face mask), and computed tomography profiles showing ground-glass opacity pneumonia associated to and high levels of D-dimer. Our panel of mediators included HMGB1, ATP, tissue factor, PGE2 , LTB4 , and cys-LTs. Follow-up studies showed increased serum levels of every inflammatory mediator in patients with COVID-19 as compared to HCs. Originally acting as a transcription factor, HMGB1 acquires pro-inflammatory functions following secretion by activated leukocytes or necrotic tissues. Serum levels of HMGB1 were positively correlated with cys-LTs, D-dimer, aspartate aminotransferase, and alanine aminotransferase. Notably, the levels of the classical alarmin HMGB1 were higher in deceased patients, allowing their discrimination from patients that had been discharged at the early pulmonary and hyperinflammatory phase of COVID-19. In particular, we verified that HMGB1 levels above 125.4 ng/ml is the cutoff that distinguishes patients that are at higher risk of death. In conclusion, we propose the use of serum levels of HMGB1 as a biomarker of severe prognosis of COVID-19.


Asunto(s)
COVID-19 , Proteína HMGB1 , Humanos , Tromboplastina , COVID-19/diagnóstico , Biomarcadores , Pronóstico , Lípidos , Adenosina Trifosfato
4.
PLoS Negl Trop Dis ; 16(6): e0010559, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35759510

RESUMEN

Epigenetic mechanisms are responsible for a wide range of biological phenomena in insects, controlling embryonic development, growth, aging and nutrition. Despite this, the role of epigenetics in shaping insect-pathogen interactions has received little attention. Gene expression in eukaryotes is regulated by histone acetylation/deacetylation, an epigenetic process mediated by histone acetyltransferases (HATs) and histone deacetylases (HDACs). In this study, we explored the role of the Aedes aegypti histone acetyltransferase CBP (AaCBP) after infection with Zika virus (ZIKV), focusing on the two main immune tissues, the midgut and fat body. We showed that the expression and activity of AaCBP could be positively modulated by blood meal and ZIKV infection. Nevertheless, Zika-infected mosquitoes that were silenced for AaCBP revealed a significant reduction in the acetylation of H3K27 (CBP target marker), followed by downmodulation of the expression of immune genes, higher titers of ZIKV and lower survival rates. Importantly, in Zika-infected mosquitoes that were treated with sodium butyrate, a histone deacetylase inhibitor, their capacity to fight virus infection was rescued. Our data point to a direct correlation among histone hyperacetylation by AaCBP, upregulation of antimicrobial peptide genes and increased survival of Zika-infected-A. aegypti.


Asunto(s)
Aedes , Infección por el Virus Zika , Virus Zika , Aedes/genética , Animales , Epigénesis Genética , Histona Acetiltransferasas/genética , Histonas/genética , Mosquitos Vectores , Virus Zika/fisiología
5.
PLoS Negl Trop Dis ; 14(7): e0008332, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32609727

RESUMEN

Treatment and control of schistosomiasis still rely on only one effective drug, praziquantel (PZQ) and, due to mass treatment, the increasing risk of selecting for schistosome strains that are resistant to PZQ has alerted investigators to the urgent need to develop novel therapeutic strategies. The histone-modifying enzymes (HMEs) represent promising targets for the development of epigenetic drugs against Schistosoma mansoni. In the present study, we targeted the S. mansoni lysine-specific demethylase 1 (SmLSD1), a transcriptional corepressor, using a novel and selective synthetic inhibitor, MC3935, which was used to treat schistosomula and adult worms in vitro. By using cell viability assays and optical and electron microscopy, we showed that treatment with MC3935 affected parasite motility, egg-laying, tegument, and cellular organelle structures, culminating in the death of schistosomula and adult worms. In silico molecular modeling and docking analysis suggested that MC3935 binds to the catalytic pocket of SmLSD1. Western blot analysis revealed that MC3935 inhibited SmLSD1 demethylation activity of H3K4me1/2. Knockdown of SmLSD1 by RNAi recapitulated MC3935 phenotypes in adult worms. RNA-Seq analysis of MC3935-treated parasites revealed significant differences in gene expression related to critical biological processes. Collectively, our findings show that SmLSD1 is a promising drug target for the treatment of schistosomiasis and strongly support the further development and in vivo testing of selective schistosome LSD1 inhibitors.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Histona Demetilasas/antagonistas & inhibidores , Schistosoma mansoni/efectos de los fármacos , Schistosoma mansoni/ultraestructura , Esquistosomiasis mansoni/tratamiento farmacológico , Animales , Antihelmínticos/farmacología , Resistencia a Medicamentos , Microscopía Electrónica de Rastreo , Oviposición/efectos de los fármacos , Praziquantel/farmacología , Esquistosomiasis mansoni/parasitología , Esquistosomiasis mansoni/patología
6.
Biochem Biophys Res Commun ; 390(4): 1245-9, 2009 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-19879244

RESUMEN

Schistosoma mansoni HMGB1 (SmHMGB1) was revealed to be a substrate for the parasite histone acetyltransferases SmGCN5 and SmCBP1. We found that full-length SmHMGB1, as well as its HMG-box B (but not HMG-box A) were acetylated in vitro by SmGCN5 and SmCBP1. However, SmCBP1 was able to acetylate both substrates more efficiently than SmGCN5. Interestingly, the removal of the C-terminal acidic tail of SmHMGB1 (SmHMGB1DeltaC) resulted in increased acetylation of the protein. We showed by mammalian cell transfection assays that SmHMGB1 and SmHMGB1DeltaC were transported from the nucleus to the cytoplasm after sodium butyrate (NaB) treatment. Importantly, after NaB treatment, SmHMGB1 was also present outside the cell. Together, our data suggest that acetylation of SmHMGB1 plays a role in cellular trafficking, culminating with its secretion to the extracellular milieu. The possible role of SmHMGB1 acetylation in the pathogenesis of schistosomiasis is discussed.


Asunto(s)
Núcleo Celular/metabolismo , Proteína HMGB1/metabolismo , Histona Acetiltransferasas/metabolismo , Schistosoma mansoni/metabolismo , Acetilación , Transporte Activo de Núcleo Celular , Animales , Células Cultivadas , Esquistosomiasis mansoni/parasitología
7.
Biochem Biophys Res Commun ; 371(4): 689-93, 2008 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-18455507

RESUMEN

The eggs produced by sexually mature female Schistosma mansoni are responsible for the pathogenesis of the disease. The eggshell precursor gene p14 is expressed only in the vitelline cells of sexually mature female worms in response to a yet unidentified male stimulus. Herein, we report the identification of a novel nuclear receptor response element in the upstream region of the p14 gene. This element contains the canonical hexameric DNA core motif, 5'-PuGGTCA, composed of an atypically spaced direct repeat (DR17). Schistosome nuclear receptors SmRXR1 and SmNR1 specifically bound to the p14-DR17 element as a heterodimer. SmRXR1, but not SmNR1, bound to the motif as a monomer. Introduction of mutations in the TCA core sequence completely abolished the binding by SmRXR1/SmNR1 heterodimer. This finding supports our hypothesis that the expression of Schistosoma mansonip14 gene is regulated through the nuclear receptor signaling pathway.


Asunto(s)
Regulación de la Expresión Génica , Genes de Helminto , Proteínas del Helminto/metabolismo , Receptores de Glutamato/metabolismo , Secuencias Repetitivas de Ácidos Nucleicos , Receptores X Retinoide/metabolismo , Schistosoma mansoni/genética , Animales , Secuencia de Bases , Dimerización , Femenino , Datos de Secuencia Molecular , Secuencias Reguladoras de Ácidos Nucleicos , Schistosoma mansoni/metabolismo , Transducción de Señal
8.
Biochem Biophys Res Commun ; 370(1): 53-6, 2008 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-18346457

RESUMEN

The transcriptional co-activator GCN5, a histone acetyltransferase (HAT), is part of large multimeric complexes that are required for chromatin remodeling and transcription activation. As in other eukaryotes, the DNA from the parasite Schistosome mansoni is organized into nucleosomes and the genome encodes components of chromatin-remodeling complexes. Using a series of synthetic peptides we determined that Lys-14 of histone H3 was acetylated by the recombinant SmGCN5-HAT domain. SmGCN5 was also able to acetylate schistosome non-histone proteins, such as the nuclear receptors SmRXR1 and SmNR1, and the co-activator SmNCoA-62. Electron microscopy revealed the presence of SmGCN5 protein in the nuclei of vitelline cells. Within the nucleus, SmGCN5 was found to be located in interchromatin granule clusters (IGCs), which are transcriptionally active structures. The data suggest that SmGCN5 is involved in transcription activation.


Asunto(s)
Proteínas del Helminto/metabolismo , Histona Acetiltransferasas/metabolismo , Schistosoma mansoni/enzimología , Schistosoma mansoni/genética , Activación Transcripcional , Acetilación , Animales , Núcleo Celular/enzimología , Eucromatina/enzimología , Genes de Helminto , Proteínas del Helminto/análisis , Histona Acetiltransferasas/análisis , Histonas/metabolismo , Ratones , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas Recombinantes/metabolismo , Vitelinas/metabolismo , Vitelinas/ultraestructura
9.
Int J Parasitol ; 38(10): 1133-47, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18359485

RESUMEN

The Schistosoma mansoni nuclear receptors (NR) SmRXR1 and SmNR1 have recently been shown to form a heterodimer and to bind to canonic hormone response DNA elements. Recruitment of co-regulatory proteins to NRs is required for their transcriptional and biological activities. Here, we cloned a novel S. mansoni NR co-activator, SmNCoA-62. SmNCoA-62 is highly homologous to the human Vitamin D receptor co-activator NCoA62/SKIP. SmNCoA-62 contains the SNW nuclear receptor interaction domain and a putative C-terminus transactivation domain. By using in vitro pull-down assays, we fully mapped the interaction domains of S. mansoni NR co-activators, SmNCoA-62, SmGCN5 and SmCBP1 with SmRXR1 and SmNR1, as well as the domains that mediate interactions amongst the co-activators themselves. By mutagenesis analysis, we showed that SmCBP1 LxxLL motif 2 and LxxLL motif 3, but not LxxLL motif 1, were essential to mediate the interactions of SmCBP1 with the EF domains of SmRXR1 and SmNR1. Histone acetyltransferases SmGCN5 and SmCBP1 specifically acetylated the C/D domains of SmRXR1 and SmNR1. In addition, two acetylation sites of SmNR1 were identified. SmGCN5 and SmCBP1 also acetylated SmNCoA-62 but with significant differences in their acetylation activities. Using gel shift analysis, we were able to demonstrate, in vitro, the assembly of the co-activators on the SmRXR1/SmNR1 heterodimer bound to DNA. LxxLL motifs 2 and 3 of SmCBP1 seemed to play a crucial role for the assembly of the co-activators to the DNA-bound SmRXR1/SmNR1 heterodimer.


Asunto(s)
Proteínas del Helminto/genética , Receptores Citoplasmáticos y Nucleares/genética , Schistosoma mansoni/genética , Acetilación , Secuencia de Aminoácidos , Animales , Mapeo Cromosómico , Clonación Molecular , ADN Complementario/genética , ADN de Helmintos/genética , Ensayo de Cambio de Movilidad Electroforética/métodos , Regulación del Desarrollo de la Expresión Génica , Genes de Helminto , Proteínas del Helminto/metabolismo , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Filogenia , Receptores Citoplasmáticos y Nucleares/metabolismo , Schistosoma mansoni/clasificación , Schistosoma mansoni/metabolismo , Análisis de Secuencia de ADN/métodos , Especificidad de la Especie
10.
Exp Parasitol ; 120(2): 200-4, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18655787

RESUMEN

Eukaryotic LIM domain proteins contain zinc finger forming motifs rich in cysteine and histidine that enable them to interact with other proteins. A cDNA clone isolated from an adult schistosome cDNA library revealed a sequence that coded for a novel class of proteins bearing 6 LIM domains and an N-terminal PET domain, SmLIMPETin. Phylogeny reconstruction of SmLIMPETin and comparison of its sequence to invertebrate homologues and to the vertebrate four-and-a-half LIM domains protein family (FHLs), uncovered a novel LIM domain protein family, the invertebrate LIM and PET domain protein family (LIMPETin). Northern blots, RT-PCR and Western blot showed that SmLIMPETin gene was less expressed in sexually mature adult females compared to sexually immature adult females and sexually mature and immature adult males, and not expressed in schistosomula.


Asunto(s)
Proteínas Portadoras/química , Proteínas del Helminto/química , Schistosoma mansoni/química , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Proteínas Portadoras/genética , Regulación hacia Abajo , Femenino , Proteínas del Helminto/genética , Masculino , Datos de Secuencia Molecular , Filogenia , Schistosoma mansoni/clasificación , Schistosoma mansoni/genética
11.
Gene ; 671: 152-160, 2018 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-29859286

RESUMEN

High-mobility group B (HMGB) proteins have highly conserved, unique DNA-binding domains, HMG boxes, that can bind non-B-type DNA structures, such as bent, kinked and unwound structures, with high affinity. HMGB proteins also promote DNA bending, looping and unwinding. In this study, we determined the role of the Aedes aegypti single HMG-box domain protein AaHMGB; characterized its structure, spatiotemporal expression levels, subcellular localization, and nucleic acid binding activities; and compared these properties with those of its double-HMG-box counterpart protein, AaHMGB1. Via qRT-PCR, we showed that AaHMGB is expressed at much higher levels than AaHMGB1 throughout mosquito development. In situ hybridization results suggested a role for AaHMGB and AaHMGB1 during embryogenesis. Immunolocalization in the midgut revealed that AaHMGB is exclusively nuclear. Circular dichroism and fluorescence spectroscopy analyses showed that AaHMGB exhibits common features of α-helical structures and is more stably folded than AaHMGB1, likely due to the presence of one or two HMG boxes. Using several DNA substrates or single-stranded RNAs as probes, we observed significant differences between AaHMGB and AaHMGB1 in terms of their binding patterns, activity and/or specificity. Importantly, we showed that the phosphorylation of AaHMGB plays a critical role in its DNA-binding activity. Our study provides additional insight into the roles of single- versus double-HMG-box-containing proteins in nucleic acid interactions for better understanding of mosquito development, physiology and homeostasis.


Asunto(s)
Aedes/crecimiento & desarrollo , Proteínas HMGB/química , Proteínas HMGB/genética , Proteínas HMGB/metabolismo , Aedes/genética , Aedes/metabolismo , Animales , Núcleo Celular/metabolismo , Dicroismo Circular , Regulación del Desarrollo de la Expresión Génica , Proteínas de Insectos/química , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Modelos Moleculares , Fosforilación , Estructura Secundaria de Proteína , Distribución Tisular
12.
Insect Biochem Mol Biol ; 80: 32-41, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27867076

RESUMEN

High Mobility Group protein 1 (HMGB1) is a non-histone, chromatin-associated nuclear protein that functions in regulating eukaryotic gene expression. We investigated the influence and mechanism of action of Aedes aegypti HMGB1 (AaHMGB1) on mosquito Rel1A-mediated transcription from target gene promoters. The DNA-binding domain (RHD) of AaRel1A was bacterially expressed and purified, and AaHMGB1 dramatically enhanced RHD binding to consensus NF-kB/Rel DNA response elements. Luciferase reporter analyses using a cecropin gene promoter showed that AaHMGB1 potentiates the transcriptional activity of AaRel1A in Aag-2 cells. Moreover, overexpression of AaHMGB1 in Aag-2 cells led to an increase in mRNA levels of antimicrobial peptide genes. In vitro GST pull-down assays revealed that the presence of DNA is a pre-requisite for assembly of a possible ternary complex containing DNA, AaHMGB1 and AaRel1A. Notably, DNA bending by AaHMGB1 enhanced the binding of AaRel1A to a DNA fragment containing a putative NF-kB/Rel response element. Importantly, AaHMGB1 was identified as a potential immune modulator in A. aegypti through AaHMGB1 overexpression or RNAi silencing in Aag-2 cells followed by bacterial challenge or through AaHMGB1 RNAi knockdown in mosquitoes followed by Dengue virus (DENV) infection. We propose a model in which AaHMGB1 bends NF-kB/Rel target DNA to recruit and allow more efficient AaRel1A binding to activate transcription of effector genes, culminating in a stronger Toll pathway-mediated response against DENV infection.


Asunto(s)
Aedes/genética , Proteína HMGB1/genética , Proteínas de Insectos/genética , Chaperonas Moleculares/genética , Aedes/metabolismo , Animales , ADN/genética , ADN/metabolismo , Proteína HMGB1/metabolismo , Proteínas de Insectos/metabolismo , Chaperonas Moleculares/metabolismo
13.
Gene ; 377: 33-45, 2006 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-16644144

RESUMEN

The parasitic helminth Schistosoma mansoni contains three HMGB proteins, HMGB1, HMGB2 and HMGB3, of primary amino acid sequences highly similar to vertebrate proteins. In this report we describe the characterization of the HMGB1 proteins and their genes from S. mansoni and Schistosoma japonicum. The deduced amino acid sequences of HMGB1 proteins from both schistosome species are identical, and comprise 176 residues. The proteins contain the two evolutionarily highly conserved HMG-box domains, A and B, exhibiting 60% similarity to mammalian HMGB1. Unlike the human HMGB1 which contains an unbroken run of 30 glutamic or aspartic residues, the SmHMGB1 or SjHMGB1 proteins possess unusually short acidic C-terminal tails (5 acidic residues interrupted by 2 serines). Southern hybridization and DNA sequencing revealed a single copy HMGB1 gene, composed of 3 exons and two introns, in S. mansoni. The exon/intron boundaries are identical to those of the human HMGB1 gene, with the exception that the second exon of the SmHMGB1 gene which is not split into two exons as in the human HMGB1 gene. RNA blot analysis revealed that the SmHMGB1 gene is constitutively expressed in similar levels both in male and female worms. The single-sized mRNA for SmHMGB1 is consistent with the size derived from the cDNA. Although DNA binding properties of SmHMGB1 (or SjHMGB1) protein seem to be similar to those previously reported with human HMGB1, i.e., preferential binding to supercoiled DNA over linear DNA, specific recognition of DNA four-way junctions, DNA-induced supercoiling in the presence of topoisomerase I, and DNA bending, we have observed two important differences relative to those observed with the human HMGB1: (i) the inability of the isolated SmHMGB1 domain A to bend DNA (as revealed by T4 ligase-mediated circularization assay), and (ii) higher DNA supercoiling and bending potential of the SmHMGB1 protein as compared to its human counterpart. The latter finding may indicate that the long acidic C-tail of human HMGB1 has much stronger repressive role on DNA bending or DNA supercoiling by topoisomerase I at physiological ionic strength than the short C-tail of the SmHMGB1 protein. Considering the important role of HMGB1 in DNA replication, transcription, recombination, and in particularly, the mediation of inflammation responses in mammalian cells, further studies on schistosome HMGB proteins may provide valuable information related to schistosomiasis, where inflammation plays a critical role in this disease.


Asunto(s)
Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Proteínas del Helminto/genética , Proteínas del Helminto/metabolismo , Schistosoma japonicum/genética , Schistosoma japonicum/metabolismo , Schistosoma mansoni/genética , Schistosoma mansoni/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Clonación Molecular , ADN Complementario/genética , ADN de Helmintos/química , ADN de Helmintos/genética , ADN de Helmintos/metabolismo , ADN Superhelicoidal/química , ADN Superhelicoidal/genética , ADN Superhelicoidal/metabolismo , Genes de Helminto , Humanos , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Schistosoma japonicum/patogenicidad , Schistosoma mansoni/patogenicidad , Homología de Secuencia de Aminoácido
14.
Int J Parasitol ; 46(4): 253-62, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26820302

RESUMEN

The Schistosoma mansoni High Mobility Group Box (HMGB) proteins SmHMGB1, SmHMGB2 and SmHMGB3 share highly conserved HMG box DNA binding domains but have significantly different C-terminal acidic tails. Here, we used three full-length and tailless forms of the S. mansoni HMGB proteins to examine the functional roles of their acidic tails. DNA binding assays revealed that the different lengths of the acidic tails among the three SmHMGB proteins significantly and distinctively influenced their DNA transactions. Spectroscopic analyses indicated that the longest acidic tail of SmHMGB3 contributes to the structural stabilisation of this protein. Using immunohistochemical analysis, we showed distinct patterns of SmHMGB1, SmHMGB2 and SmHMGB3 expression in different tissues of adult worms. RNA interference approaches indicated a role for SmHMGB2 and SmHMGB3 in the reproductive system of female worms, whereas for SmHMGB1 no clear phenotype was observed. Schistosome HMGB proteins can be phosphorylated, acetylated and methylated. Importantly, the acetylation and methylation of schistosome HMGBs were greatly enhanced upon removal of the acidic tail. These data support the notion that the C-terminal acidic tails dictate the differences in the structure, expression and function of schistosome HMGB proteins.


Asunto(s)
Proteínas HMGB/química , Schistosoma mansoni/química , Secuencia de Aminoácidos , Animales , Dicroismo Circular , ADN de Helmintos/metabolismo , Femenino , Proteínas HMGB/genética , Proteínas HMGB/fisiología , Inmunohistoquímica , Masculino , Especificidad de Órganos , Dominios y Motivos de Interacción de Proteínas/fisiología , Procesamiento Proteico-Postraduccional/fisiología , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Interferencia de ARN , Schistosoma mansoni/genética
15.
Sci Rep ; 6: 36444, 2016 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-27805064

RESUMEN

Aedes aegypti mosquitoes are important vectors of viral diseases. Mosquito host factors play key roles in virus control and it has been suggested that dengue virus replication is regulated by Dnmt2-mediated DNA methylation. However, recent studies have shown that Dnmt2 is a tRNA methyltransferase and that Dnmt2-dependent methylomes lack defined DNA methylation patterns, thus necessitating a systematic re-evaluation of the mosquito genome methylation status. We have now searched the Ae. aegypti genome for candidate DNA modification enzymes. This failed to reveal any known (cytosine-5) DNA methyltransferases, but identified homologues for the Dnmt2 tRNA methyltransferase, the Mettl4 (adenine-6) DNA methyltransferase, and the Tet DNA demethylase. All genes were expressed at variable levels throughout mosquito development. Mass spectrometry demonstrated that DNA methylation levels were several orders of magnitude below the levels that are usually detected in organisms with DNA methylation-dependent epigenetic regulation. Furthermore, whole-genome bisulfite sequencing failed to reveal any evidence of defined DNA methylation patterns. These results suggest that the Ae. aegypti genome is unmethylated. Interestingly, additional RNA bisulfite sequencing provided first evidence for Dnmt2-mediated tRNA methylation in mosquitoes. These findings have important implications for understanding the mechanism of Dnmt2-dependent virus regulation.


Asunto(s)
Aedes/genética , Metilación de ADN , Secuencia de Aminoácidos , Animales , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Bacteriano/análisis , ADN Bacteriano/química , ADN Bacteriano/metabolismo , Genoma , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Espectrometría de Masas , ARN de Transferencia/química , ARN de Transferencia/metabolismo , Alineación de Secuencia , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Secuenciación Completa del Genoma
16.
Mol Biochem Parasitol ; 135(1): 21-30, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15287583

RESUMEN

PUR-alpha is a highly conserved protein in eukaryotes belonging to the family of single-stranded DNA-binding proteins. Because PUR-alpha is a multifunctional protein that participates in several regulatory events at the level of gene transcription, it became relevant to investigate the structural features of Schistosoma mansoni PUR-alpha (SmPUR-alpha) that could be correlated to its mode of action. Using deletion constructs on a dot blot assay we mapped the domains of GST-SmPUR-alpha fusion protein involved in the interactions with DNA and RNA. Individually, the N-terminal amino acid residues 1-26 and the C-terminal residues 196-276 of GST-SmPUR-alpha which did not contain nucleic acid-binding domains, did not bind ssDNA or RNA. In contrast, domains encompassing the N-terminal and Class I and C-terminal plus Class I exhibited the highest binding affinity. Seemingly, the latter (GST-SmPUR-alpha 174-276) played a major role in nucleic acid interaction as judged by affinity alone. Other combinations of the deletion constructs displayed either intermediary or no binding affinity to the DNA or RNA probes. Gel shift competition assay showed that GST-SmPUR-alpha bound to ssDNA with higher affinity than to RNA. Because SmPUR-alpha contains two putative phosphorylation sites the protein was tested as a substrate to casein kinase II. GST-SmPUR-alpha could be phosphorylated in vitro by casein kinase II at both, the N- and C-terminus of the protein. The multifunctional nature of SmPUR-alpha was demonstrated by experiments measuring the physical interaction between SmPUR-alpha and the transcription factor SMYB1. This was determined in vivo (yeast two hybrid) and in vitro (GST-pull down). Furthermore, we showed that SmPUR-alpha and SMYB1 acted synergistically to bind preferentially to pyrimidine-rich sequences.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas del Helminto/genética , Proteínas del Helminto/metabolismo , Mapeo de Interacción de Proteínas , Schistosoma mansoni/fisiología , Factores de Transcripción/metabolismo , Animales , Sitios de Unión , Western Blotting , Quinasa de la Caseína II/metabolismo , ADN de Cadena Simple/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Genes de Helminto , Fosforilación , Unión Proteica , ARN , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Eliminación de Secuencia , Técnicas del Sistema de Dos Híbridos
17.
PLoS One ; 7(6): e39104, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22723938

RESUMEN

BACKGROUND: 3-(3-chloro-phenyl)-5-(4-pyridyl)-4,5-dihydroisoxazole (DIC) is a five-membered heterocyclic compound containing a N-O bond. The anti-inflammatory effects of this compound were studied both in vitro and in vivo. PRINCIPAL FINDINGS: DIC effectively decreased TNF-α and IL-6 release from LPS-stimulated macrophages in a dose dependent manner. DIC diminished the levels of COX-2 with subsequent inhibition of PGE(2) production. DIC also compromised HMGB1 translocation from the nucleus to the cytoplasm. Moreover, DIC prevented the nuclear translocation of NF-κB and inhibited the MAPK pathway. In vivo, DIC inhibited migration of neutrophils to the peritoneal cavity of mice. CONCLUSIONS: This study presents the potential utilization of a synthetic compound, as a lead for the development of novel anti-inflammatory drugs.


Asunto(s)
Antiinflamatorios/farmacología , Isoxazoles/farmacología , Piridinas/farmacología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/síntesis química , Movimiento Celular/efectos de los fármacos , Movimiento Celular/inmunología , Dinoprostona/biosíntesis , Activación Enzimática/efectos de los fármacos , Femenino , Proteína HMGB1/metabolismo , Interleucina-6/biosíntesis , Isoxazoles/administración & dosificación , Isoxazoles/síntesis química , Lipopolisacáridos/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Peritonitis/inducido químicamente , Peritonitis/tratamiento farmacológico , Piridinas/administración & dosificación , Piridinas/síntesis química , Transducción de Señal , Tioglicolatos/efectos adversos , Factor de Necrosis Tumoral alfa/biosíntesis
18.
PLoS One ; 7(7): e40192, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22802955

RESUMEN

The mosquito Aedes aegypti can spread the dengue, chikungunya and yellow fever viruses. Thus, the search for key molecules involved in the mosquito survival represents today a promising vector control strategy. High Mobility Group Box (HMGB) proteins are essential nuclear factors that maintain the high-order structure of chromatin, keeping eukaryotic cells viable. Outside the nucleus, secreted HMGB proteins could alert the innate immune system to foreign antigens and trigger the initiation of host defenses. In this work, we cloned and functionally characterized the HMGB1 protein from Aedes aegypti (AaHMGB1). The AaHMGB1 protein typically consists of two HMG-box DNA binding domains and an acidic C-terminus. Interestingly, AaHMGB1 contains a unique alanine/glutamine-rich (AQ-rich) C-terminal region that seems to be exclusive of dipteran HMGB proteins. AaHMGB1 is localized to the cell nucleus, mainly associated with heterochromatin. Circular dichroism analyses of AaHMGB1 or the C-terminal truncated proteins revealed α-helical structures. We showed that AaHMGB1 can effectively bind and change the topology of DNA, and that the AQ-rich and the C-terminal acidic regions can modulate its ability to promote DNA supercoiling, as well as its preference to bind supercoiled DNA. AaHMGB1 is phosphorylated by PKA and PKC, but not by CK2. Importantly, phosphorylation of AaHMGB1 by PKA or PKC completely abolishes its DNA bending activity. Thus, our study shows that a functional HMGB1 protein occurs in Aedes aegypt and we provide the first description of a HMGB1 protein containing an AQ-rich regulatory C-terminus.


Asunto(s)
Proteína HMGB1/química , Proteínas de Insectos/química , Aedes , Secuencia de Aminoácidos , Animales , Núcleo Celular/metabolismo , Clonación Molecular , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , ADN Superhelicoidal/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteína HMGB1/aislamiento & purificación , Datos de Secuencia Molecular , Fosforilación , Proteína Quinasa C/metabolismo
19.
PLoS One ; 6(8): e23572, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21887276

RESUMEN

BACKGROUND: The helminth Schistosoma mansoni parasite resides in mesenteric veins where fecundated female worms lay hundred of eggs daily. Some of the egg antigens are trapped in the liver and induce a vigorous granulomatous response. High Mobility Group Box 1 (HMGB1), a nuclear factor, can also be secreted and act as a cytokine. Schistosome HMGB1 (SmHMGB1) is secreted by the eggs and stimulate the production of key cytokines involved in the pathology of schistosomiasis. Thus, understanding the mechanism of SmHMGB1 release becomes mandatory. Here, we addressed the question of how the nuclear SmHMGB1 can reach the extracellular space. PRINCIPAL FINDINGS: We showed in vitro and in vivo that CK2 phosphorylation was involved in the nucleocytoplasmic shuttling of SmHMGB1. By site-directed mutagenesis we mapped the two serine residues of SmHMGB1 that were phosphorylated by CK2. By DNA bending and supercoiling assays we showed that CK2 phosphorylation of SmHMGB1 had no effect in the DNA binding activities of the protein. We showed by electron microscopy, as well as by cell transfection and fluorescence microscopy that SmHMGB1 was present in the nucleus and cytoplasm of adult schistosomes and mammalian cells. In addition, we showed that treatments of the cells with either a phosphatase or a CK2 inhibitor were able to enhance or block, respectively, the cellular traffic of SmHMGB1. Importantly, we showed by confocal microscopy and biochemically that SmHMGB1 is significantly secreted by S. mansoni eggs of infected animals and that SmHMGB1 that were localized in the periovular schistosomotic granuloma were phosphorylated. CONCLUSIONS: We showed that secretion of SmHMGB1 is regulated by phosphorylation. Moreover, our results suggest that egg-secreted SmHMGB1 may represent a new egg antigen. Therefore, the identification of drugs that specifically target phosphorylation of SmHMGB1 might block its secretion and interfere with the pathogenesis of schistosomiasis.


Asunto(s)
Quinasa de la Caseína II/metabolismo , ADN Protozoario/metabolismo , Proteína HMGB1/metabolismo , Schistosoma mansoni/metabolismo , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Animales , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Citosol/metabolismo , ADN Superhelicoidal/metabolismo , Pruebas de Enzimas , Femenino , Granuloma/metabolismo , Proteína HMGB1/química , Proteína HMGB1/genética , Células HeLa , Humanos , Hígado/metabolismo , Hígado/parasitología , Hígado/patología , Hígado/ultraestructura , Ratones , Datos de Secuencia Molecular , Fosforilación , Unión Proteica , Mapas de Interacción de Proteínas , Schistosoma mansoni/citología , Schistosoma mansoni/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA