Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
BMC Biol ; 21(1): 123, 2023 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-37226244

RESUMO

BACKGROUND: Changes in gene expression levels during brain development are thought to have played an important role in the evolution of human cognition. With the advent of high-throughput sequencing technologies, changes in brain developmental expression patterns, as well as human-specific brain gene expression, have been characterized. However, interpreting the origin of evolutionarily advanced cognition in human brains requires a deeper understanding of the regulation of gene expression, including the epigenomic context, along the primate genome. Here, we used chromatin immunoprecipitation sequencing (ChIP-seq) to measure the genome-wide profiles of histone H3 lysine 4 trimethylation (H3K4me3) and histone H3 lysine 27 acetylation (H3K27ac), both of which are associated with transcriptional activation in the prefrontal cortex of humans, chimpanzees, and rhesus macaques. RESULTS: We found a discrete functional association, in which H3K4me3HP gain was significantly associated with myelination assembly and signaling transmission, while H3K4me3HP loss played a vital role in synaptic activity. Moreover, H3K27acHP gain was enriched in interneuron and oligodendrocyte markers, and H3K27acHP loss was enriched in CA1 pyramidal neuron markers. Using strand-specific RNA sequencing (ssRNA-seq), we first demonstrated that approximately 7 and 2% of human-specific expressed genes were epigenetically marked by H3K4me3HP and H3K27acHP, respectively, providing robust support for causal involvement of histones in gene expression. We also revealed the co-activation role of epigenetic modification and transcription factors in human-specific transcriptome evolution. Mechanistically, histone-modifying enzymes at least partially contribute to an epigenetic disturbance among primates, especially for the H3K27ac epigenomic marker. In line with this, peaks enriched in the macaque lineage were found to be driven by upregulated acetyl enzymes. CONCLUSIONS: Our results comprehensively elucidated a causal species-specific gene-histone-enzyme landscape in the prefrontal cortex and highlighted the regulatory interaction that drove transcriptional activation.


Assuntos
Epigênese Genética , Histonas , Animais , Humanos , Lisina , Macaca mulatta/genética , Córtex Pré-Frontal , Expressão Gênica
2.
Nutr Neurosci ; 23(9): 714-723, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30474509

RESUMO

A sub-optimal nutritional environment from early life can be envisaged as a stressor that translates into mental health problems in adulthood. After considering (a) the widespread prevalence of vitamin B12 deficiency especially amongst women in developing countries and (b) the importance of vitamin B12 in normal brain function, in this study we have elucidated the behavioural correlates of chronic severe and moderate vitamin B12 deficiency in C57BL/6 mice. Female weanling mice were assigned to three dietary groups: (a) control AIN-76A diet with cellulose as dietary fibre (b) vitamin B12 restricted AIN-76A diet with pectin as dietary fibre (severe deficiency group) and (c) vitamin B12 restricted AIN-76A diet with cellulose as dietary fibre (moderate deficiency group). The mice received these diets throughout pregnancy, lactation and thereafter. Nest-building, maternal care, anxiety and depressive behaviours were evaluated. Oxidative stress, activities of antioxidant enzymes and expression of various histone modifying enzymes in brain were investigated to unravel the probable underlying mechanisms. Our data suggests that both severe and moderate vitamin B12 deficiency induced anxiety and impaired maternal care. However, only severe vitamin B12 deficiency induced depression. Oxidative stress and poor antioxidant defense underlie the deleterious effects of both severe and moderate vitamin B12 deficiency. Altered expression of histone modifying enzymes in the brain of severely deficient mice is suggestive of epigenetic reprogramming. This study suggests that chronic vitamin B12 deficiency leads to behavioural anomalies in female C57BL/6 mice and the severity of these outcomes can be correlated to the level of deficiency.


Assuntos
Encéfalo/enzimologia , Histona Desacetilases/metabolismo , Estresse Oxidativo , Deficiência de Vitamina B 12/metabolismo , Animais , Ansiedade/etiologia , Ansiedade/metabolismo , Comportamento Animal/fisiologia , Depressão/etiologia , Depressão/metabolismo , Feminino , Camundongos Endogâmicos C57BL , Comportamento de Nidação/fisiologia , Deficiência de Vitamina B 12/psicologia
3.
Adv Exp Med Biol ; 1202: 259-279, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32034718

RESUMO

Signal transduction pathways directly communicate and transform chromatin to change the epigenetic landscape and regulate gene expression. Chromatin acts as a dynamic platform of signal integration and storage. Histone modifications and alteration of chromatin structure play the main role in chromatin-based gene expression regulation. Alterations in genes coding for histone modifying enzymes and chromatin modifiers result in malfunction of proteins that regulate chromatin modification and remodeling. Such dysregulations culminate in profound changes in chromatin structure and distorted patterns of gene expression. Gliomagenesis is a multistep process, involving both genetic and epigenetic alterations. Recent applications of next generation sequencing have revealed that many chromatin regulation-related genes, including ATRX, ARID1A, SMARCA4, SMARCA2, SMARCC2, BAF155 and hSNF5 are mutated in gliomas. In this review we summarize newly identified mechanisms affecting expression or functions of selected histone modifying enzymes and chromatin modifiers in gliomas. We focus on selected examples of pathogenic mechanisms involving ATRX, histone methyltransferase G9a, histone acetylases/deacetylases and chromatin remodeling complexes SMARCA2/4. We discuss the impact of selected epigenetics alterations on glioma pathobiology, signaling and therapeutic responses. We assess the attempts of targeting defective pathways with new inhibitors.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Cromatina/metabolismo , Glioma/tratamento farmacológico , Glioma/patologia , Histonas/metabolismo , Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/genética , Cromatina/genética , Montagem e Desmontagem da Cromatina/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Glioma/enzimologia , Glioma/genética , Código das Histonas/efeitos dos fármacos , Histonas/química , Humanos
4.
Proc Natl Acad Sci U S A ; 110(33): 13510-5, 2013 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-23904479

RESUMO

Lysine methylation of the p65 subunit of nuclear factor κB (NF-κB) on K218 and K221 together or K37 alone strongly enhances gene expression in response to cytokines. We analyzed the effects of K-to-Q mutations in the REL homology domain of p65 on the response to IL-1ß in 293 cells with low levels of p65. The K218/221Q mutation greatly reduced the expression of 39 of 82 genes, whereas the K37Q mutation reduced the expression of 23 different genes. Enhanced expression of the lysine demethylase FBXL11, which catalyzes the demethylation of K218 and K221 specifically, inhibited the expression of most of the genes that were inhibited by the DKQ mutation. CHIP-Seq analysis showed that the K218/221Q mutation greatly reduces the affinity of p65 for many promoters and that the K37Q mutation does not. Structural modeling showed that the newly introduced methyl groups of K218 and K221 interact directly with DNA to increase the affinity of p65 for specific κB sites. Thus, the K218/221Q and K37Q mutations have dramatically different effects because methylations of these residues affect different genes by distinct mechanisms.


Assuntos
Proteínas F-Box/metabolismo , Regulação da Expressão Gênica/imunologia , Lisina/metabolismo , NF-kappa B/metabolismo , Oxirredutases N-Desmetilantes/metabolismo , Western Blotting , Imunoprecipitação da Cromatina , Citocinas/farmacologia , Primers do DNA/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Interleucina-1beta/farmacologia , Histona Desmetilases com o Domínio Jumonji , Metilação , Análise em Microsséries , Mutagênese Sítio-Dirigida , Mutação/genética , NF-kappa B/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA
5.
Future Oncol ; 11(18): 2587-601, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26289459

RESUMO

The epigenetic landscape is deregulated in cancer due to aberrant activation or inactivation of enzymes maintaining and modifying the epigenome. Histone modifications and global aberrations at the histone level may result in distorted patterns of gene expression, and malfunction of proteins that regulate chromatin modification and remodeling. Recent whole genome studies demonstrated that histones and chaperone proteins harbor mutations that may result in gross alterations of the epigenome leading to genome instability. Glioma development is a multistep process, involving genetic and epigenetic alterations. This review summarizes newly identified mechanisms affecting expression/functions of histone-modifying enzymes and chromatin modifiers in gliomas. We discuss recent approaches to overcome epigenetic alterations with histone-modifying enzyme inhibitors and their prospects for glioma therapy.


Assuntos
Cromatina/genética , Cromatina/metabolismo , Glioma/etiologia , Glioma/metabolismo , Histonas/metabolismo , Fatores Etários , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Epigênese Genética/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioma/tratamento farmacológico , Glioma/mortalidade , Glioma/patologia , Inibidores de Histona Desacetilases/farmacologia , Inibidores de Histona Desacetilases/uso terapêutico , Histonas/genética , Humanos , Mutação , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Nucleossomos/metabolismo
6.
Clin Epigenetics ; 15(1): 146, 2023 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-37697409

RESUMO

Dysregulation of histone modifications has been implicated in the pathogenesis of both inflammatory bowel disease (IBD) and colorectal cancer (CRC). These diseases are characterized by chronic inflammation, and alterations in histone modifications have been linked to their development and progression. Furthermore, the gut microbiota plays a crucial role in regulating immune responses and maintaining gut homeostasis, and it has been shown to exert effects on histone modifications and gene expression in host cells. Recent advances in our understanding of the roles of histone-modifying enzymes and their associated chromatin modifications in IBD and CRC have provided new insights into potential therapeutic interventions. In particular, inhibitors of histone-modifying enzymes have been explored in clinical trials as a possible therapeutic approach for these diseases. This review aims to explore these potential therapeutic interventions and analyze previous and ongoing clinical trials that examined the use of histone-modifying enzyme inhibitors for the treatment of IBD and CRC. This paper will contribute to the current body of knowledge by exploring the latest advances in the field and discussing the limitations of existing approaches. By providing a comprehensive analysis of the potential benefits of targeting histone-modifying enzymes for the treatment of IBD and CRC, this review will help to inform future research in this area and highlight the significance of understanding the functions of histone-modifying enzymes and their associated chromatin modifications in gastrointestinal disorders for the development of potential therapeutic interventions.


Assuntos
Histonas , Doenças Inflamatórias Intestinais , Humanos , Metilação de DNA , Doenças Inflamatórias Intestinais/tratamento farmacológico , Inflamação , Cromatina
7.
Adv Protein Chem Struct Biol ; 130: 189-243, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35534108

RESUMO

Histone modifying enzymes regulate chromatin architecture through covalent modifications and ultimately control multiple aspects of cellular function. Disruption of histone modification leads to changes in gene expression profiles and may lead to disease. Both small molecule inhibitors and intermediary metabolites have been shown to modulate histone modifying enzyme activity although our ability to identify successful drug candidates or novel metabolic regulators of these enzymes has been limited. Using a combination of large scale in silico screens and in vivo phenotypic analysis, we identified several small molecules and intermediary metabolites with distinctive HME activity. Our approach using unsupervised learning identifies the chemical fingerprints of both small molecules and metabolites that facilitate recognition by the enzymes active sites which can be used as a blueprint to design novel inhibitors. Furthermore, this work supports the idea that histone modifying enzymes sense intermediary metabolites integrating genes, environment and cellular physiology.


Assuntos
Cromatina , Histonas , Montagem e Desmontagem da Cromatina , Desenho de Fármacos , Histonas/metabolismo , Processamento de Proteína Pós-Traducional
8.
FEBS J ; 289(5): 1191-1213, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-33415821

RESUMO

In the last 15 years, increasing evidence linking epigenetics to various aspects of cancer biology has prompted the investigation of histone post-translational modifications (PTMs) and histone variants in the context of clinical samples. The studies performed so far demonstrated the potential of this type of investigations for the discovery of both potential epigenetic biomarkers for patient stratification and novel epigenetic mechanisms potentially targetable for cancer therapy. Although traditionally the analysis of histones in clinical samples was performed through antibody-based methods, mass spectrometry (MS) has emerged as a more powerful tool for the unbiased, comprehensive, and quantitative investigation of histone PTMs and variants. MS has been extensively used for the analysis of epigenetic marks in cell lines and animal tissue and, thanks to recent technological advances, is now ready to be applied also to clinical samples. In this review, we will provide an overview on the quantitative MS-based analysis of histones, their PTMs and their variants in cancer clinical samples, highlighting current achievements and future perspectives for this novel field of research. Among the different MS-based approaches currently available for histone PTM profiling, we will focus on the 'bottom-up' strategy, namely the analysis of short proteolytic peptides, as it has been already successfully employed for the analysis of clinical samples.


Assuntos
Epigênese Genética , Histonas/genética , Espectrometria de Massas/tendências , Neoplasias/genética , Peptídeos/análise , Processamento de Proteína Pós-Traducional , Antineoplásicos/uso terapêutico , Cromatina/química , Cromatina/metabolismo , Regulação Neoplásica da Expressão Gênica , Histonas/metabolismo , Humanos , Espectrometria de Massas/métodos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/diagnóstico , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Mapeamento de Peptídeos/métodos , Peptídeos/genética , Peptídeos/metabolismo , Proteômica
9.
Clin Epigenetics ; 14(1): 145, 2022 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-36371348

RESUMO

Epigenetics includes a complex set of processes that alter gene activity without modifying the DNA sequence, which ultimately determines how the genetic information common to all the cells of an organism is used to generate different cell types. Dysregulation in the deposition and maintenance of epigenetic features, which include histone posttranslational modifications (PTMs) and histone variants, can result in the inappropriate expression or silencing of genes, often leading to diseased states, including cancer. The investigation of histone PTMs and variants in the context of clinical samples has highlighted their importance as biomarkers for patient stratification and as key players in aberrant epigenetic mechanisms potentially targetable for therapy. Mass spectrometry (MS) has emerged as the most powerful and versatile tool for the comprehensive, unbiased and quantitative analysis of histone proteoforms. In recent years, these approaches-which we refer to as "epi-proteomics"-have demonstrated their usefulness for the investigation of epigenetic mechanisms in pathological conditions, offering a number of advantages compared with the antibody-based methods traditionally used to profile clinical samples. In this review article, we will provide a critical overview of the MS-based approaches that can be employed to study histone PTMs and variants in clinical samples, with a strong focus on the latest advances in this area, such as the analysis of uncommon modifications and the integration of epi-proteomics data into multi-OMICs approaches, as well as the challenges to be addressed to fully exploit the potential of this novel field of research.


Assuntos
Histonas , Proteômica , Humanos , Histonas/metabolismo , Proteômica/métodos , Metilação de DNA , Epigenômica , Processamento de Proteína Pós-Traducional , Epigênese Genética
10.
Exp Gerontol ; 125: 110658, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31302168

RESUMO

Changes in histone acetylation and methylation status with aging affect gene expression and phenotype in several tissues; however, age-related changes in histone modification in the skeletal muscle have not been elucidated yet. This study investigated age-related global changes in histone modification in rat gastrocnemius muscle. Male Wistar rats (n = 28) were assigned to one of four age groups (n = 7 per group) corresponding to different life stages: 3 months old (3-mo; young), 6 months old (adult), 12 months old (12-mo; middle-aged), and 24 months old (24-mo; old). The gastrocnemius muscle was removed and global histone modification (acetylation and tri-methylation) at K9 and K27 was evaluated by western blotting. Relative muscle mass decreased in the 12- and 24-mo rats accompanied with reduction in type IIb myosin heavy chain isoforms and Myh4 (MHC IIB) mRNA expression. Histone H3 acetylation decreased in an age-dependent manner, with lower levels in 12- and 24-mo groups than in the 3-mo group. K9 and K27 acetylation decreased with age. Although there was no significant change in K27 tri-methylation, K9 tri-methylation showed an age-dependent decline. Histone modification status (acetylation at K9 and K27 and tri-methylation at K9) was positively associated with relative gastrocnemius muscle weight, the percentage of type IIb myosin heavy chain isoform, myosin heavy chain type IIb protein expression, and the level of Myh4 mRNA. Thus, global histone H3 methylation and acetylation decrease with age, and the latter might be associated with age-related muscle atrophy of rat gastrocnemius muscle.


Assuntos
Envelhecimento/metabolismo , Código das Histonas , Músculo Esquelético/metabolismo , Acetilação , Animais , Masculino , Metilação , Cadeias Pesadas de Miosina/metabolismo , Ratos Wistar
11.
Expert Rev Mol Diagn ; 19(1): 27-36, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30563379

RESUMO

Introduction: Histone modifying enzymes (HMEs)-catalyzed histone modifications are important epigenetic markers that play critical roles in the regulation of a variety of cellular functions, especially the regulation of gene expression. The aberrant histone modifying enzyme activity and the abnormal histone modification level are closely associated with various human diseases including cancers, making them become the promising and attractive disease biomarkers. Consequently, the development of efficient assays for accurate and sensitive detection of histone modifications and HMEs are crucial for disease diagnosis. Areas covered: In this review, we summarize the advances in histone modifications and HMEs assays in recent 5 years (2013-2018), including the development of various methods based on fluorescent, bioluminescent, colorimetric, electrochemical, surface-enhanced Raman scattering, and mass spectrometry strategies. Their principles and applications for in vitro and in vivo assays are reviewed, and the future directions are discussed as well. Expert commentary: In comparison with the conventional radioactive and Western blot assays, the newly developed histone modifications and HMEs assays exhibit distinct advantages. Especially, the introduction of novel nanomaterials and advanced analytical techniques in recent years has greatly improved the assay performances, promoting their further applications in biomedical research and clinical diagnosis.


Assuntos
Ensaios Enzimáticos Clínicos/métodos , Código das Histonas , Biomarcadores/análise , Biomarcadores/metabolismo , Ensaios Enzimáticos Clínicos/normas , Histona Acetiltransferases/análise , Histona Acetiltransferases/metabolismo , Histona Metiltransferases/análise , Histona Metiltransferases/metabolismo , Humanos
12.
Cells ; 7(9)2018 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-30150556

RESUMO

Histone post-translational modifications influence many fundamental cellular events by regulating chromatin structure and gene transcriptional activity. These modifications are highly dynamic and tightly controlled, with many enzymes devoted to the addition and removal of these modifications. Interestingly, these modifying enzymes are themselves fine-tuned and precisely regulated at the level of protein turnover by ubiquitin-proteasomal processing. Here, we focus on recent progress centered on the mechanisms regulating ubiquitination of histone modifying enzymes, including ubiquitin proteasomal degradation and the reverse process of deubiquitination. We will also discuss the potential pathophysiological significance of these processes.

13.
Epigenetics ; 11(4): 303-19, 2016 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-27019326

RESUMO

Epigenetic mechanisms have not been characterized in ticks despite their importance as vectors of human and animal diseases worldwide. The objective of this study was to characterize the histones and histone modifying enzymes (HMEs) of the tick vector Ixodes scapularis and their role during Anaplasma phagocytophilum infection. We first identified 5 histones and 34 HMEs in I. scapularis in comparison with similar proteins in model organisms. Then, we used transcriptomic and proteomic data to analyze the mRNA and protein levels of I. scapularis histones and HMEs in response to A. phagocytophilum infection of tick tissues and cultured cells. Finally, selected HMEs were functionally characterized by pharmacological studies in cultured tick cells. The results suggest that A. phagocytophilum manipulates tick cell epigenetics to increase I. scapularis p300/CBP, histone deacetylase, and Sirtuin levels, resulting in an inhibition of cell apoptosis that in turn facilitates pathogen infection and multiplication. These results also suggest that a compensatory mechanism might exist by which A. phagocytophilum manipulates tick HMEs to regulate transcription and apoptosis in a tissue-specific manner to facilitate infection, but preserving tick fitness to guarantee survival of both pathogens and ticks. Our study also indicates that the pathogen manipulates arthropod and vertebrate cell epigenetics in similar ways to inhibit the host response to infection. Epigenetic regulation of tick biological processes is an essential element of the infection by A. phagocytophilum and the study of the mechanisms and principal actors involved is likely to provide clues for the development of anti-tick drugs and vaccines.


Assuntos
Anaplasma phagocytophilum/genética , Epigênese Genética , Código das Histonas/genética , Histonas/genética , Animais , Apoptose/genética , Linhagem Celular , Histonas/metabolismo , Interações Hospedeiro-Patógeno/genética , Humanos , Insetos Vetores/genética , Ixodes/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Transcriptoma/genética , Fatores de Transcrição de p300-CBP/biossíntese , Fatores de Transcrição de p300-CBP/genética
14.
Expert Rev Mol Diagn ; 16(3): 297-306, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26750583

RESUMO

Histones are the chief components of chromatin. When being catalyzed by a series of histone modifying enzymes, histones may undergo various post-translational modifications such as acetylation, methylation, phosphorylation, ubiquitylation and SUMOylation. The dysregulation of histone modifying enzymes will alter the histone post-modification patterns and cause diverse diseases including cancers. Consequently, the histone modifying enzymes have emerged as the promising biomarkers for disease diagnosis and prognosis. In this review, we summarize the recent researches about the histone modifying enzymes as the disease biomarkers, and highlight the development of methods for histone modifying enzyme assays.


Assuntos
Biomarcadores Tumorais/metabolismo , Histona Acetiltransferases/metabolismo , Histona Desacetilases/metabolismo , Histona Desmetilases/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Neoplasias/diagnóstico , Protamina Quinase/metabolismo , Animais , Ensaios Enzimáticos/métodos , Histona Metiltransferases , Humanos , Técnicas de Diagnóstico Molecular/métodos
15.
Mem. Inst. Oswaldo Cruz ; 106(7): 794-801, Nov. 2011. ilus
Artigo em Inglês | LILACS | ID: lil-606641

RESUMO

Only one drug is currently available for the treatment and control of schistosomiasis and the increasing risk of selecting strains of schistosome that are resistant to praziquantel means that the development of new drugs is urgent. With this objective we have chosen to target the enzymes modifying histones and in particular the histone acetyltransferases and histone deacetylases (HDAC). Inhibitors of HDACs (HDACi) are under intense study as potential anti-cancer drugs and act via the induction of cell cycle arrest and/or apoptosis. Schistosomes like other parasites can be considered as similar to tumours in that they maintain an intense metabolic activity and rate of cell division that is outside the control of the host. We have shown that HDACi can induce apoptosis and death of schistosomes maintained in culture and have set up a consortium (Schistosome Epigenetics: Targets, Regulation, New Drugs) funded by the European Commission with the aim of developing inhibitors specific for schistosome histone modifying enzymes as novel lead compounds for drug development.


Assuntos
Animais , Cromatina/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Histona Acetiltransferases/antagonistas & inibidores , Histona Desacetilases/metabolismo , Schistosoma/efeitos dos fármacos , Cromatina/metabolismo , Desenho de Fármacos , Histona Acetiltransferases/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Schistosoma/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA