Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Mol Genet Metab ; 116(1-2): 13-23, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26071627

RESUMO

Lysosomal storage diseases (LSDs) are caused by accumulation of partially degraded substrates within the lysosome, as a result of a function loss of a lysosomal protein. Recombinant lysosomal proteins are usually produced in mammalian cells, based on their capacity to carry out post-translational modifications similar to those observed in human native proteins. However, during the last years, a growing number of studies have shown the possibility to produce active forms of lysosomal proteins in other expression systems, such as plants and microorganisms. In this paper, we review the production and characterization of human lysosomal proteins, deficient in several LSDs, which have been produced in microorganisms. For this purpose, Escherichia coli, Saccharomyces cerevisiae, Pichia pastoris, Yarrowia lipolytica, and Ogataea minuta have been used as expression systems. The recombinant lysosomal proteins expressed in these hosts have shown similar substrate specificities, and temperature and pH stability profiles to those produced in mammalian cells. In addition, pre-clinical results have shown that recombinant lysosomal enzymes produced in microorganisms can be taken-up by cells and reduce the substrate accumulated within the lysosome. Recently, metabolic engineering in yeasts has allowed the production of lysosomal enzymes with tailored N-glycosylations, while progresses in E. coli N-glycosylations offer a potential platform to improve the production of these recombinant lysosomal enzymes. In summary, microorganisms represent convenient platform for the production of recombinant lysosomal proteins for biochemical and physicochemical characterization, as well as for the development of ERT for LSD.


Assuntos
Doenças por Armazenamento dos Lisossomos/tratamento farmacológico , Lisossomos/enzimologia , Proteínas/isolamento & purificação , Proteínas Recombinantes/biossíntese , Animais , Escherichia coli/metabolismo , Vetores Genéticos/metabolismo , Humanos , Plantas/genética , Proteínas/uso terapêutico , Proteínas Recombinantes/uso terapêutico , Saccharomycetales/metabolismo
2.
Food Sci Technol Int ; 20(5): 365-72, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23744119

RESUMO

In the food industry, osmotic dehydration can be an important stage to obtain partially dry foodstuffs. However, the remaining spent osmotic solution at the end of the process could become a waste with an important environmental impact due to the large amount of organic compounds that it might contain. Since one of the most important osmotic agents used in osmotic dehydration is sucrose, this spent osmotic solution could be used to be biotransformed to produce fructooligosaccharides by a fructosyltransferase. This study evaluated the production of fructooligosaccharides using the fructosyltransferase produced by Aspergillus oryzae N74, and the spent osmotic solution that resulted in the osmotic dehydration of Andes berry (Rubus glaucus) and tamarillo (Cyphomandra betacea). Assays were conducted at small and bioreactor scales, using spent osmotic solution with or without re-concentration. At small scale no significant difference (p > 0.05) was observed in the fructooligosaccharides production yield, ranging from 31.18% to 34.98% for spent osmotic solution from tamarillo osmotic dehydration, and from 33.16% to 37.52% for spent osmotic solution from Andes berry osmotic dehydration, using either the SOS with or without re-concentration. At bioreactor scale the highest fructooligosaccharides yield of 58.51 ± 1.73% was obtained with spent osmotic solution that resulted from tamarillo osmotic dehydration. With the spent osmotic solution from Andes berry osmotic dehydration the yield was 49.17 ± 2.82%. These results showed the feasibility of producing fructooligosaccharides from spent osmotic solution that is considered a waste in food industry.


Assuntos
Aspergillus oryzae/metabolismo , Dessecação/métodos , Manipulação de Alimentos/métodos , Oligossacarídeos/biossíntese , Soluções , Sacarose , Reatores Biológicos , Frutas , Resíduos Industriais , Osmose , Prebióticos , Reciclagem , Rosaceae , Solanum
3.
Environ Int ; 190: 108914, 2024 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-39079332

RESUMO

PFOA is a legacy Per- and Polyfluorinated Substances (PFAS), a group of chemicals widely used in various industrial applications and consumer products. Although there has been a voluntary phase out of PFOA since 2005, it is still widely detected in various water supplies. A growing body of evidence suggests an association between PFOA exposure, particularly during developmental stages, with increased risks of neurodegenerative diseases (NDs). The neurotoxic mechanism of developmental PFOA exposure, however, remains poorly understood. Utilizing human induced-pluripotent stem cell (hiPSC)-derived cortical neurons, we investigated the effect of PFOA exposure prior to differentiation and assessed changes in neuronal characteristics, transcriptome, and neurodegeneration markers mimicking a Developmental Origin of Health and Disease (DoHAD) paradigm. Exposure to PFOA before neuron differentiation resulted in persistent alterations in nuclear morphology, neuronal network, and calcium activity. RNA sequencing analysis further revealed transcriptomic changes aligning with Alzheimer's Disease (AD) after PFOA exposure. These observations were further corroborated by alterations in tau phosphorylation markers, the presence of fibrillar tau, an increase in liquid droplets, and a decrease in RNA translational efficiency characterized using a battery of biochemical assays. Taken together, our results revealed persistent deficits of key neuronal characteristics induced by pre-differentiation PFOA exposure, suggesting impairments in several AD-related pathways that can together contribute to the elevation of AD risk after pre-differentiation PFOA exposure.

4.
Sci Total Environ ; 908: 168307, 2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-37949145

RESUMO

Atrazine (ATZ) is one of the most used herbicides in the US and a known endocrine disruptor. ATZ is frequently detected in drinking water, especially in Midwestern regions of the United States, exceeding the EPA regulation of maximum contamination level (MCL) of 3 ppb. Epidemiology studies have suggested an association between ATZ exposure and neurodegeneration. Less, however, is known about the neurotoxic mechanism of ATZ, particularly for exposures at a developmental stage. Here, we exposed floor plate progenitors (FPPs) derived from human induced pluripotent stem cells (hiPSCs) to low concentrations of ATZ at 0.3 and 3 ppb for two days followed by differentiation into dopaminergic (DA) neurons in ATZ-free medium. We then examined the morphology, activity, pathological protein aggregation, and transcriptomic changes of differentiated DA neurons. We observed significant decrease in the complexity of neurite network, increase of neuronal activity, and elevated tau- and α-synuclein (aSyn) pathologies after ATZ exposure. The ATZ-induced neuronal changes observed here align with pathological characteristics in Parkinson's disease (PD). Transcriptomic analysis further corroborates our findings; and collectively provides a strong evidence base that low-concentration ATZ exposure during development can elicit increased risk of neurodegeneration.


Assuntos
Atrazina , Herbicidas , Células-Tronco Pluripotentes Induzidas , Doença de Parkinson , Humanos , Atrazina/toxicidade , Neurônios Dopaminérgicos , Herbicidas/toxicidade
5.
Heliyon ; 10(12): e32555, 2024 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-38952373

RESUMO

Mucopolysaccharidosis IVA (MPS IVA) is a lysosomal storage disease caused by mutations in the gene encoding the lysosomal enzyme N-acetylgalactosamine-6-sulfate sulfatase (GALNS), resulting in the accumulation of keratan sulfate (KS) and chondroitin-6-sulfate (C6S). Previously, it was reported the production of an active human recombinant GALNS (rGALNS) in E. coli BL21(DE3). However, this recombinant enzyme was not taken up by HEK293 cells or MPS IVA skin fibroblasts. Here, we leveraged a glyco-engineered E. coli strain to produce a recombinant human GALNS bearing the eukaryotic trimannosyl core N-glycan, Man3GlcNAc2 (rGALNSoptGly). The N-glycosylated GALNS was produced at 100 mL and 1.65 L scales, purified and characterized with respect to pH stability, enzyme kinetic parameters, cell uptake, and KS clearance. The results showed that the addition of trimannosyl core N-glycans enhanced both protein stability and substrate affinity. rGALNSoptGly was capture through a mannose receptor-mediated process. This enzyme was delivered to the lysosome, where it reduced KS storage in human MPS IVA fibroblasts. This study demonstrates the potential of a glyco-engineered E. coli for producing a fully functional GALNS enzyme. It may offer an economic approach for the biosynthesis of a therapeutic glycoprotein that could prove useful for MPS IVA treatment. This strategy could be extended to other lysosomal enzymes that rely on the presence of mannose N-glycans for cell uptake.

6.
Environ Pollut ; 308: 119684, 2022 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-35764183

RESUMO

Perfluorooctanoic acid (PFOA) is abundant in environment due to its historical uses in consumer products and industrial applications. Exposure to low doses of PFOA has been associated with various disease risks, including neurological disorders. The underlying mechanism, however, remains poorly understood. In this study, we examined the effects of low dose PFOA exposure at 0.4 and 4 µg/L on the morphology, epigenome, mitochondrion, and neuronal markers of dopaminergic (DA)-like SH-SY5Y cells. We observed persistent decreases in H3K4me3, H3K27me3 and 5 mC markers in nucleus along with alterations in nuclear size and chromatin compaction percentage in DA-like neurons differentiated from SH-SY5Y cells exposed to 0.4 and 4 µg/L PFOA. Among the selected epigenetic features, DNA methylation pattern can be used to distinguish between PFOA-exposed and naïve populations, suggesting the involvement of epigenetic regulation. Moreover, DA-like neurons with pre-differentiation PFOA exposure exhibit altered network connectivity, mitochondrial volume, and TH expression, implying impairment in DA neuron functionality. Collectively, our results revealed the prolonged effects of developmental PFOA exposure on the fitness of DA-like neurons and identified epigenome and mitochondrion as potential targets for bearing long-lasting changes contributing to increased risks of neurological diseases later in life.


Assuntos
Fluorocarbonos , Neuroblastoma , Biomarcadores/metabolismo , Caprilatos/metabolismo , Caprilatos/toxicidade , Metilação de DNA , Dopamina/metabolismo , Epigênese Genética , Fluorocarbonos/metabolismo , Fluorocarbonos/toxicidade , Humanos , Mitocôndrias/metabolismo , Neuroblastoma/metabolismo , Neurônios/metabolismo
7.
Curr Res Toxicol ; 3: 100061, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35005634

RESUMO

Lead (Pb) is a heavy metal contaminant commonly found in air, soil, and drinking water due to legacy uses. Excretion of ingested Pb can result in extensive kidney damages due to elevated oxidative stress. Epigenetic alterations induced by exposure to Pb have also been implied but remain poorly understood. In this work, we assessed changes in repressive epigenetic marks, namely DNA methylation (meCpG) and histone 3 lysine 9 tri-methylation (H3K9me3) after exposure to Pb. Live cell epigenetic probes coupled to bimolecular fluorescence complementation (BiFC) were used to monitor changes in the selected epigenetic marks. Exposure to Pb significantly lowered meCpG and H3K9me3 levels in HEK293T cells suggesting global changes in constitutive heterochromatin. A heterodimeric pair of probes that tags chromatin regions enriched in both meCpG and H3K9me3 further confirmed our findings. The observed epigenetic changes can be partially attributed to aberrant transcriptional changes induced by Pb, such as overexpression of TET1 after Pb exposure. Lastly, we monitored changes in selected heterochromatin marks after removal of Pb and found that changes in these markers do not immediately recover to their original level suggesting potential long-term damages to chromatin structure.

8.
Neurotoxicol Teratol ; 92: 107091, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35472415

RESUMO

Atrazine (ATZ) is the second most common agricultural herbicide used in the United States and is an endocrine disrupting chemical (EDC). Developmental exposure to ATZ can lead to significant behavioral and morphological alterations in exposed animals and their progeny suggesting the involvement of an epigenetic mechanism. Specific epigenetic mechanisms responsible for these alterations, however, are yet to be elucidated. In this study, we exposed zebrafish embryos to 0, 0.3, 3, or 30 ppb (µg/L) of ATZ from 1 to 72 h post fertilization (hpf). Chemical exposure was ceased and zebrafish maintained until 9 months post fertilization (mpf), when whole-genome bisulfite sequencing (WGBS) was performed to assess the effects of embryonic ATZ exposure on DNA methylation in female fish brains. The number of differentially methylated genes (DMGs) increased with increasing treatment concentration. DMGs were enriched in neurological pathways with extensive methylation changes consistently observed in neuroendocrine pathways. Specifically, DMGs with methylation changes in promoter regions showed hypomethylation in estrogen receptor signaling and hypermethylation in androgen signaling. DMGs with methylation changes in genebody were primarily enriched for mitochondrion-related pathways associated with healthy aging. Integrated analysis with transcriptomic data at 9 mpf exhibited a similar trend identifying CABLES1 and NDUFA4 as shared targets at all concentrations. We then compared the predicted upstream regulators of transcriptomic changes with DMGs and identified CALML3 as a common upstream regulator at both 0.3 and 30 ppb that exhibit significant methylation changes. Collectively, our study identified long-lasting DNA methylation changes in genome after embryonic ATZ exposure and elucidated potential gene targets whose aberrant methylation features may drive alterations in gene transcription in long-term.


Assuntos
Atrazina , Disruptores Endócrinos , Herbicidas , Animais , Atrazina/metabolismo , Atrazina/toxicidade , Metilação de DNA , Disruptores Endócrinos/toxicidade , Feminino , Herbicidas/toxicidade , Peixe-Zebra
9.
N Biotechnol ; 69: 18-27, 2022 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-35217201

RESUMO

Fructo-oligosaccharides (FOS) are one of the most well-studied and commercialized prebiotics. FOS can be obtained either by controlled hydrolysis of inulin or by sucrose transfructosylation. FOS produced from sucrose are typically classified as short-chain FOS (scFOS), of which the best known are 1-kestotriose (GF2), 1,1-kestotetraose (GF3), and 1,1,1-kestopentaose (GF4), produced by fructosyltransferases (FTases) or ß-fructofuranosidases. In previous work, FOS production was studied using the Aspergillus oryzae N74 strain, its ftase gene was heterologously expressed in Komagataella phaffii (Pichia pastoris), and the enzyme's tertiary structure modeled. More recently, residues that may be involved in protein-substrate interactions were predicted. In this study, the aim was to experimentally validate previous in silico results by independently producing recombinant wild-type A. oryzae N74 FTase and three single-point mutations in Komagataella phaffii (Pichia pastoris). The R163A mutation virtually abolished the transfructosylating activity, indicating a requirement for the positively charged arginine residue in the catalytic domain D. In contrast, transfructosylating activity was improved by introducing the mutations V242E or F254H, with V242E resulting in higher production of GF2 without affecting that of GF3. Interestingly, initial sucrose concentration, reaction temperature and the presence of metal cofactors did not affect the enhanced activity of mutant V242E. Overall, these results shed light on the mechanism of transfructosylation of the FTase from A. oryzae and expand considerations regarding the design of biotechnological processes for specific FOS production.


Assuntos
Aspergillus oryzae , Aspergillus oryzae/genética , Hexosiltransferases , Oligossacarídeos , Pichia/genética , Saccharomycetales , Sacarose
10.
Mol Biol Rep ; 38(2): 1151-61, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20563857

RESUMO

The fructooligosaccharides (FOS) represent an important source of prebiotic compounds that are widely used as an ingredient in functional foods. Recently, the strain Aspergillus oryzae N74 was reported as a potential microorganism for the industrial production of FOS, due to its high yields of FOS production. In this work, we used a PCR-cloning strategy to clone the A. oryzae N74 ftase gene as a previous step for recombinant enzyme production. Ftase showed a 1630 bp size with a 99% similarity with other A. oryzae strains and between 1 to 68% identities with other Aspergillus strains. This gene encodes for a 525 amino acids protein with 99% similarity with other A. oryzae strains and between 11 to 69% similarities with other Aspergillus strains. Finally, an A. oryzae N74 FTase tertiary structure model was predicted base on its similarity with other glycoside hydrolase 32 family members. The active site was located inside the ß-propeller domain and was formed for non-charged polar and charged amino acids. In summary, these results shows the high level of sequence conservation between A. oryzae strains and represent a first step towards the development of a FOS production industrial process using recombinant microorganism carrying the ftase gene from A. oryzae N74.


Assuntos
Aspergillus oryzae/enzimologia , Aspergillus oryzae/genética , Hexosiltransferases/genética , Sequência de Aminoácidos , Sequência de Bases , Domínio Catalítico , Clonagem Molecular , Regulação Fúngica da Expressão Gênica , Dados de Sequência Molecular , Oligossacarídeos/química , Filogenia , Reação em Cadeia da Polimerase , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Análise de Sequência de DNA , Homologia de Sequência do Ácido Nucleico
11.
Biochim Biophys Acta Gene Regul Mech ; 1864(8): 194725, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34174495

RESUMO

The 3D spatial organization of the genome controls gene expression and cell functionality. Heterochromatin (HC), which is the densely compacted and largely silenced part of the chromatin, is the driver for the formation and maintenance of nuclear organization in the mammalian nucleus. It is functionally divided into highly compact constitutive heterochromatin (cHC) and transcriptionally poised facultative heterochromatin (fHC). Long regarded as a static structure, the highly dynamic nature of the heterochromatin is being slowly understood and studied. These changes in HC occur on various temporal scales during the cell cycle and differentiation processes. Most methods that capture information about the heterochromatin are static techniques that cannot provide a readout of how the HC organization evolves with time. The delineation of specific areas such as fHC are also rendered difficult due to its diffusive nature and lack of specific features. Another degree of complexity in characterizing changes in heterochromatin occurs due to the heterogeneity in the HC organization of individual cells, necessitating single cell studies. Overall, there is a need for live cell compatible tools that can stably track the heterochromatin as it undergoes re-organization. In this work, we present an approach to track cHC and fHC based on the epigenetic hallmarks associated with them. Unlike conventional immunostaining approaches, we use small recombinant protein probes that allow us to dynamically monitor the HC by binding to modifications specific to the cHC and fHC, such as H3K9me3, DNA methylation and H3K27me3. We demonstrate the use of the probes to follow the changes in HC induced by drug perturbations at the single cell level. We also use the probe sets combinatorically to simultaneously track chromatin regions enriched in two selected epigenetic modifications using a FRET based approach that enabled us tracking distinctive chromatin features in situ.


Assuntos
Epigênese Genética , Transferência Ressonante de Energia de Fluorescência , Heterocromatina/metabolismo , Metilação de DNA , Corantes Fluorescentes , Células HEK293 , Código das Histonas , Histonas/metabolismo , Humanos , Análise de Célula Única
12.
Environ Pollut ; 271: 116379, 2021 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-33388679

RESUMO

Exposures to organic pesticides, particularly during a developmental window, have been associated with various neurodegenerative diseases later in life. Atrazine (ATZ), one of the most used pesticides in the U.S., is suspected to be associated with increased neurodegeneration later in life but few studies assessed the neurotoxicity of developmental ATZ exposure using human neuronal cells. Here, we exposed human SH-SY5Y cells to 0.3, 3, and 30 ppb of ATZ prior to differentiating them into dopaminergic-like neurons in ATZ-free medium to mimic developmental exposure. The differentiated neurons exhibit altered neurite outgrowth and SNCA pathology depending on the ATZ treatment doses. Epigenome changes, such as decreases in 5mC (for 0.3 ppb only), H3K9me3, and H3K27me3 were observed immediately after exposure. These alterations persist in a compensatory manner in differentiated neurons. Specifically, we observed significant reductions in 5mC and H3K9me3, as well as, an increase in H3K27me3 in ATZ-exposed cells after differentiation, suggesting substantial chromatin rearrangements after developmental ATZ exposure. Transcriptional changes of relevant epigenetic enzymes were also quantified but found to only partially explain the observed epigenome alteration. Our results thus collectively suggest that exposure to low-dose of ATZ prior to differentiation can result in long-lasting changes in epigenome and increase risks of SNCA-related Parkinson's Disease.


Assuntos
Atrazina , Herbicidas , Atrazina/toxicidade , Diferenciação Celular , Linhagem Celular , Humanos , Neurônios
13.
Chemosphere ; 264(Pt 1): 128486, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33032221

RESUMO

Lead (Pb) is a commonly found heavy metal due to its historical applications. Recent studies have associated early-life Pb exposure with the onset of various neurodegenerative disease. The molecular mechanisms of Pb conferring long-term neurotoxicity, however, is yet to be elucidated. In this study, we explored the persistency of alteration in epigenetic marks that arise from exposure to low dose of Pb using a combination of image-based and gene expression analysis. Using SH-SY5Y as a model cell line, we observed significant alterations in global 5-methycytosine (5 mC) and histone 3 lysine 27 tri-methylation (H3K27me3) and histone 3 lysine 9 tri-methylation (H3K9me3) levels in a dose-dependent manner immediately after Pb exposure. The changes are partially associated with alterations in epigenetic enzyme expression levels. Long term culturing (14 days) after cease of exposure revealed persistent changes in 5 mC, partial recovery in H3K9me3 and overcompensation in H3K27me3 levels. The observed alterations in H3K9me3 and H3K27me3 are reversed after neuronal differentiation, while reduction in 5 mC levels are amplified with significant changes in patterns as identified via texture clustering analysis. Moreover, correlation analysis demonstrates a strong positive correlation between trends of 5 mC alteration after differentiation and neuronal morphology. Collectively, our results suggest that exposure to low dose of Pb prior to differentiation can result in persistent epigenome alterations that can potentially be responsible for the observed phenotypic changes. Our work reveals that Pb induced changes in epigenetic repressive marks can persist through neuron differentiation, which provides a plausible mechanism underlying long-term neurotoxicity associated with developmental Pb-exposure.


Assuntos
Histonas , Doenças Neurodegenerativas , Diferenciação Celular , Heterocromatina , Humanos , Chumbo/toxicidade
14.
Gene ; 780: 145527, 2021 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-33636292

RESUMO

Mucopolysaccharidosis type IVA (MPS IVA) is a lysosomal storage disease produced by the deficiency of the N-acetylgalactosamine-6-sulfate sulfatase (GALNS) enzyme, leading to glycosaminoglycans (GAGs) accumulation. Since currently available treatments remain limited and unspecific, novel therapeutic approaches are essential for the disease treatment. In an attempt to reduce treatment limitations, gene therapy rises as a more effective and specific alternative. We present in this study the delivery assessment of GALNS and sulfatase-modifying factor 1 (SUMF1) genes via HIV-1 derived lentiviral vectors into fibroblasts from MPS IVA patients. After transduction, we determined GALNS enzymatic activity, lysosomal mass change, and autophagy pathway impairment. Additionally, we computationally assessed the effect of mutations over the enzyme-substrate interaction and phenotypic effects. The results showed that the co-transduction of MPS IVA fibroblasts with GALNS and SUMF1 cDNAs led to a significant increase in GALNS enzyme activity and a reduction of lysosomal mass. We show that patient-specific differences in cellular response are directly associated with the set of mutations on each patient. Lastly, we present new evidence supporting autophagy impairment in MPS IVA due to the presence and changes in autophagy proteins in treated MPS IVA fibroblasts. Our results offer new evidence that demonstrate the potential of lentiviral vectors as a strategy to correct GALNS deficiency.


Assuntos
Condroitina Sulfatases , Fibroblastos/metabolismo , Vetores Genéticos , HIV-1 , Mucopolissacaridose IV , Oxirredutases atuantes sobre Doadores de Grupo Enxofre , Transdução Genética , Condroitina Sulfatases/biossíntese , Condroitina Sulfatases/genética , Terapia Genética , Células HEK293 , Humanos , Mucopolissacaridose IV/genética , Mucopolissacaridose IV/metabolismo , Mucopolissacaridose IV/terapia , Oxirredutases atuantes sobre Doadores de Grupo Enxofre/biossíntese , Oxirredutases atuantes sobre Doadores de Grupo Enxofre/genética
15.
J Ind Microbiol Biotechnol ; 37(11): 1193-201, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20582614

RESUMO

Mucopolysaccharidosis IVA (MPS IVA) is an autosomal recessive disorder caused by N-acetylgalactosamine-6-sulfate sulfatase (GALNS) deficiency. Currently no effective therapies exist for MPS IVA. In this work, production of a recombinant GALNS enzyme (rGALNS) in Escherichia coli BL21 strain was studied. At shake scale, the effect of glucose concentration on microorganism growth, and microorganism culture and induction times on rGALNS production were evaluated. At bench scale, the effect of aeration and agitation on microorganism growth, and culture and induction times were evaluated. The highest enzyme activity levels at shake scale were observed in 12 h culture after 2-4 h induction. At bench scale the highest enzyme activity levels were observed after 2 h induction. rGALNS amounts in inclusion bodies fraction were up to 17-fold higher than those observed in the soluble fraction. However, the highest levels of active enzyme were found in the soluble fraction. Western blot analysis showed the presence of a 50-kDa band, in both soluble and inclusion bodies fractions. These results show for the first time the feasibility and potential of production of active rGALNS in a prokaryotic system for development of enzyme replacement therapy for MPS IVA disease.


Assuntos
Condroitina Sulfatases/biossíntese , Escherichia coli/metabolismo , Fermentação , Corpos de Inclusão/microbiologia , Western Blotting , Condroitina Sulfatases/genética , Clonagem Molecular , Meios de Cultura , Terapia de Reposição de Enzimas/métodos , Ensaio de Imunoadsorção Enzimática , Escherichia coli/genética , Mucopolissacaridose IV/terapia , Plasmídeos/genética , Plasmídeos/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética
16.
Cells ; 9(4)2020 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-32244528

RESUMO

Gap junction (GJ) channels and their connexins (Cxs) are complex proteins that have essential functions in cell communication processes in the central nervous system (CNS). Neurons, astrocytes, oligodendrocytes, and microglial cells express an extraordinary repertory of Cxs that are important for cell to cell communication and diffusion of metabolites, ions, neurotransmitters, and gliotransmitters. GJs and Cxs not only contribute to the normal function of the CNS but also the pathological progress of several diseases, such as cancer and neurodegenerative diseases. Besides, they have important roles in mediating neuroprotection by internal or external molecules. However, regulation of Cx expression by epigenetic mechanisms has not been fully elucidated. In this review, we provide an overview of the known mechanisms that regulate the expression of the most abundant Cxs in the central nervous system, Cx30, Cx36, and Cx43, and their role in brain cancer, CNS disorders, and neuroprotection. Initially, we focus on describing the Cx gene structure and how this is regulated by epigenetic mechanisms. Then, the posttranslational modifications that mediate the activity and stability of Cxs are reviewed. Finally, the role of GJs and Cxs in glioblastoma, Alzheimer's, Parkinson's, and Huntington's diseases, and neuroprotection are analyzed with the aim of shedding light in the possibility of using Cx regulators as potential therapeutic molecules.


Assuntos
Neoplasias Encefálicas/metabolismo , Conexinas/metabolismo , Doenças Neurodegenerativas/metabolismo , Neuroproteção , Animais , Neoplasias Encefálicas/genética , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Conexinas/química , Conexinas/genética , Epigênese Genética , Humanos , Doenças Neurodegenerativas/genética , Neuroproteção/genética , Processamento de Proteína Pós-Traducional
17.
Environ Pollut ; 258: 113712, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31875570

RESUMO

How environmental chemicals can affect and exert their toxic effect at a molecular level has gained significant interest in recent years, not only for understanding their immediate health implications over exposed individuals, but also for their subsequent progeny. Atrazine (ATZ) is a commonly used herbicide in the U.S. and a long-suspected endocrine disrupting chemical. The molecular mechanism conferring long-term adverse health outcomes, however, remain elusive. Here, we explored changes in epigenetic marks that arise after exposure to ATZ at selected doses using image-based analysis coupled with data clustering. Significant decreases in methylated CpG (meCpG) and histone 3 lysine 9 tri-methylated (H3K9me3) were observed in the selected human cell line with a clear spatial preference. Treating cells with ATZ leads to the loss of a subpopulation of cells with high meCpG levels as identified in our clustering and histogram analysis. A similar trend was observed in H3K9me3 potentially attributing to the cross-talking between meCpG and H3K9me3. Changes in meCpG are likely to be associated with alterations in epigenetic enzyme expression levels regulating meCpG and persist after the removal of ATZ source which collectively provide a plausible mechanism for long-term ATZ-induced toxicity.


Assuntos
Atrazina/toxicidade , Metilação de DNA , Epigênese Genética , Herbicidas/toxicidade , Linhagem Celular , Ilhas de CpG , Disruptores Endócrinos/toxicidade , Histonas/química , Humanos
18.
Front Neurorobot ; 14: 578834, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33117141

RESUMO

Although different physiological signals, such as electrooculography (EOG) have been widely used in the control of assistance systems for people with disabilities, customizing the signal classification system remains a challenge. In most interfaces, the user must adapt to the classification parameters, although ideally the systems must adapt to the user parameters. Therefore, in this work the use of a multilayer neural network (MNN) to model the EOG signal as a mathematical function is presented, which is optimized using genetic algorithms, in order to obtain the maximum and minimum amplitude threshold of the EOG signal of each person to calibrate the designed interface. The problem of the variation of the voltage threshold of the physiological signals is addressed by means of an intelligent calibration performed every 3 min; if an assistance system is not calibrated, it loses functionality. Artificial intelligence techniques, such as machine learning and fuzzy logic are used for classification of the EOG signal, but they need calibration parameters that are obtained through databases generated through prior user training, depending on the effectiveness of the algorithm, the learning curve, and the response time of the system. In this work, by optimizing the parameters of the EOG signal, the classification is customized and the domain time of the system is reduced without the need for a database and the training time of the user is minimized, significantly reducing the time of the learning curve. The results are implemented in an HMI for the generation of points in a Cartesian space (X, Y, Z) in order to control a manipulator robot that follows a desired trajectory by means of the movement of the user's eyeball.

19.
J Mol Med (Berl) ; 98(7): 931-946, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32529345

RESUMO

Lysosomal storage disorders (LSDs) are a group of monogenic diseases characterized by progressive accumulation of undegraded substrates into the lysosome, due to mutations in genes that encode for proteins involved in normal lysosomal function. In recent years, several approaches have been explored to find effective and successful therapies, including enzyme replacement therapy, substrate reduction therapy, pharmacological chaperones, hematopoietic stem cell transplantation, and gene therapy. In the case of gene therapy, genome editing technologies have opened new horizons to accelerate the development of novel treatment alternatives for LSD patients. In this review, we discuss the current therapies for this group of disorders and present a detailed description of major genome editing technologies, as well as the most recent advances in the treatment of LSDs. We will further highlight the challenges and current bioethical debates of genome editing.


Assuntos
Doenças por Armazenamento dos Lisossomos/tratamento farmacológico , Doenças por Armazenamento dos Lisossomos/genética , Lisossomos/genética , Animais , Edição de Genes/métodos , Terapia Genética/métodos , Transplante de Células-Tronco Hematopoéticas/métodos , Humanos , Proteínas/genética
20.
ACS Omega ; 4(8): 13250-13259, 2019 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-31460452

RESUMO

H3K9me3 (methylation of lysine 9 of histone H3) is an epigenetic modification that acts as a repressor mark. Several diseases, including cancers and neurological disorders, have been associated with aberrant changes in H3K9me3 levels. Different tools have been developed to enable detection and quantification of H3K9me3 levels in cells. Most techniques, however, lack live cell compatibility. To address this concern, we have engineered recombinant protein sensors for probing H3K9me3 in situ. A heterodimeric sensor containing a chromodomain and chromo shadow domain from HP1a was found to be optimal in recognizing H3K9me3 and exhibited similar spatial resolution to commercial antibodies. Our sensor offers similar quantitative accuracy in characterizing changes in H3K9me3 compared to antibodies but claims single cell resolution. The sensor was applied to evaluate changes in H3K9me3 responding to environmental chemical atrazine (ATZ). ATZ was found to result in significant reductions in H3K9me3 levels after 24 h of exposure. Its impact on the distribution of H3K9me3 among cell populations was also assessed and found to be distinctive. We foresee the application of our sensors in multiple toxicity and drug-screening applications.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA