Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 23(2)2022 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-35055170

RESUMO

Tuberculosis (TB) infection, caused by the airborne pathogen Mycobacterium tuberculosis (M.tb), resulted in almost 1.4 million deaths in 2019, and the number of deaths is predicted to increase by 20% over the next 5 years due to the COVID-19 pandemic. Upon reaching the alveolar space, M.tb comes into close contact with the lung mucosa before and after its encounter with host alveolar compartment cells. Our previous studies show that homeostatic, innate soluble components of the alveolar lining fluid (ALF) can quickly alter the cell envelope surface of M.tb upon contact, defining subsequent M.tb-host cell interactions and infection outcomes in vitro and in vivo. We also demonstrated that ALF from 60+ year old elders (E-ALF) vs. healthy 18- to 45-year-old adults (A-ALF) is dysfunctional, with loss of homeostatic capacity and impaired innate soluble responses linked to high local oxidative stress. In this study, a targeted transcriptional assay shows that M.tb exposure to human ALF alters the expression of its cell envelope genes. Specifically, our results indicate that A-ALF-exposed M.tb upregulates cell envelope genes associated with lipid, carbohydrate, and amino acid metabolism, as well as genes associated with redox homeostasis and transcriptional regulators. Conversely, M.tb exposure to E-ALF shows a lesser transcriptional response, with most of the M.tb genes unchanged or downregulated. Overall, this study indicates that M.tb responds and adapts to the lung alveolar environment upon contact, and that the host ALF status, determined by factors such as age, might play an important role in determining infection outcome.


Assuntos
Cápsulas Bacterianas/genética , Cápsulas Bacterianas/metabolismo , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/metabolismo , Adolescente , Adulto , Fatores Etários , Idoso , Líquido da Lavagem Broncoalveolar , Estruturas Celulares , Feminino , Regulação Bacteriana da Expressão Gênica , Humanos , Lipopolissacarídeos/biossíntese , Lipopolissacarídeos/genética , Masculino , Manosídeos/biossíntese , Manosídeos/genética , Manosiltransferases/biossíntese , Manosiltransferases/genética , Pessoa de Meia-Idade , Adulto Jovem
2.
Int J Lab Hematol ; 42(4): 464-472, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32359033

RESUMO

INTRODUCTION: The development of drug resistance is the main obstacle for successful treatment in acute myeloid leukemia (AML). Noncoding RNAs have been implicated in biological function in AML drug resistance. Aberrant protein glycosylation is associated with AML progression. The aim of the study was to explore the potential regulatory mechanism of lncRNA MEG3/miR-155/ALG9 axis in drug resistance of AML. METHODS: QRT-PCR and Western blot were used for comparison analyses of ALG9, MEG3, and miR-155 levels. CCK-8 and colony formation assays were determined for drug sensitivity and proliferative capability of AML cells. Luciferase reporter assay was used to confirm the targets of miR-155. RESULTS: The mannosyltransferase ALG9 and MEG3 was downregulated in peripheral blood mononuclear cells (PBMCs) of M5/multidrug resistance (MDR) AML patients and adriamycin (ADR)-resistant AML cell lines, which determined a positive correlation in AML patients. Low expression of ALG9 and MEG3 predicted poor prognosis of AML patients. The altered level of ALG9 was found corresponding to the drug-resistant phenotype and sphere formation of AML cells. MiR-155 was overexpressed in M5/MDR patients and ADR-resistant AML cells, as well as inversely correlated to ALG9 expression. MEG3 was a direct target of miR-155 and could sponge miR-155 in AML cells. MEG3 interacted with miR-155 to regulate ALG9 expression, which reversed the effects of ALG9 regulation on proliferation and drug resistance in AML cells. CONCLUSION: MEG3 sponged miR-155 by competing endogenous RNA (ceRNA) mechanism, which further modulated ALG9 expression and AML procession, providing a novel therapeutic target for AML chemoresistance.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Regulação Leucêmica da Expressão Gênica , Leucemia Mieloide Aguda/metabolismo , Manosiltransferases/biossíntese , Proteínas de Membrana/biossíntese , MicroRNAs/metabolismo , Proteínas de Neoplasias/biossíntese , RNA Longo não Codificante/metabolismo , RNA Neoplásico/metabolismo , Feminino , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Masculino , Manosiltransferases/genética , Proteínas de Membrana/genética , MicroRNAs/genética , Proteínas de Neoplasias/genética , RNA Longo não Codificante/genética , RNA Neoplásico/genética , Células THP-1 , Células U937
3.
Sci Rep ; 9(1): 1974, 2019 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-30760814

RESUMO

Several pioneering work have established that apart from genetic alterations, epigenetic modifications contribute significantly in tumor progression. Remarkable role of EZH2 in cancer highlights the importance of identifying its targets. Although much emphasis has been placed in recent years in designing drugs and inhibitors targeting EZH2, less effort has been given in exploring its existing targets that will help in understanding the oncogenic role of EZH2 in turn which may provide a more stringent method of targeting EZH2. In the present study, we validated six direct targets of EZH2 that are GPNMB, PMEPA1, CoL5A1, VGLL4, POMT2 and SUMF1 associated with cancer related pathways. Upon EZH2 knockdown, more than two fold increase in the target gene expression was evident. CHIP-qPCR performed in both MCF-7 and MDA-MDA-231 confirmed the in-vivo binding of EZH2 on its identified target. Thirty invasive breast carcinoma cases with their adjacent normal tissues were included in the study. Immunohistochemistry in primary breast tumor tissue array showed tumor dependent expression of EZH2. Array of MERAV expression database revealed the strength of association of EZH2 with its target genes. Real time PCR performed with RNA extracted from breast tumor tissues further authenticated the existing negative correlation between EZH2 and its target genes. Pearson correlation coefficient & statistical significance computed using the matrix provided in the database strengthened the negative correlation between identified target genes and EZH2. KM plotter analysis showed improved relapse-free survival with increased expression of PMEPA1, POMT2, VGLL4 and SUMF1 in breast cancer patients indicating their therapeutic potential. While investigating the relevance of these target genes, different mutations of them were found in breast cancer patients. Seeking the clinical relevance of our study, following our recent publication that reports the role of EZH2 in nicotine-mediated breast cancer development and progression, we observed significant reduced expression of SUMF1 in breast cancer patient samples with smoking history in comparison to never-smoked patient samples.


Assuntos
Neoplasias da Mama/genética , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Manosiltransferases/genética , Proteínas de Membrana/genética , Oxirredutases atuantes sobre Doadores de Grupo Enxofre/genética , Fatores de Transcrição/genética , Mama/patologia , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Células MCF-7 , Manosiltransferases/biossíntese , Proteínas de Membrana/biossíntese , Recidiva Local de Neoplasia/genética , Oxirredutases atuantes sobre Doadores de Grupo Enxofre/biossíntese , Receptores de Estrogênio/metabolismo , Fumar/efeitos adversos , Fatores de Transcrição/biossíntese
4.
Appl Microbiol Biotechnol ; 103(5): 2277-2293, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30706115

RESUMO

The traditional yeast Saccharomyces cerevisiae has been widely used as a host for the production of recombinant proteins and metabolites with industrial potential. However, its thick and rigid cell wall presents problems for the effective recovery of products. In this study, we modulated the expression of ScOCH1, encoding the α-1,6-mannosyltransferase responsible for outer chain biosynthesis of N-glycans, and ScCHS3, encoding the chitin synthase III required for synthesis of the majority of cell wall chitin, by exploiting the repressible ScMET3 promoter. The conditional single mutants PMET3-OCH1 and PMET3-CHS3 and the double mutant PMET3-OCH1/PMET3-CHS3 showed comparable growth to the wild-type strain under normal conditions but exhibited increased sensitivity to temperature and cell wall-disturbing agents in the presence of methionine. Such conditional growth defects were fully recovered by supplementation with 1 M sorbitol. The osmotic lysis of the conditional mutants cultivated with methionine was sufficient to release the intracellularly expressed recombinant protein, nodavirus capsid protein, with up to 60% efficiency, compared to lysis by glass bead breakage. These mutant strains also showed approximately three-fold-enhanced secretion of a recombinant extracellular glycoprotein, Saccharomycopsis fibuligera ß-glucosidase, with markedly reduced hypermannosylation, particularly in the PMET3-OCH1 mutants. Furthermore, a substantial increase of extracellular glutathione production, up to four-fold, was achieved with the conditional mutant yeast cells. Together, our data support that the conditional cell wall lysis mutants constructed based on the modulation of ScOCH1 and ScCHS3 expression would likely be useful hosts for the improved recovery of proteins and metabolites with industrial application.


Assuntos
Proteínas do Capsídeo/metabolismo , Quitina Sintase/biossíntese , Regulação Fúngica da Expressão Gênica/genética , Manosiltransferases/biossíntese , Glicoproteínas de Membrana/biossíntese , Proteínas de Saccharomyces cerevisiae/biossíntese , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas do Capsídeo/genética , Parede Celular/metabolismo , Quitina/biossíntese , Quitina Sintase/genética , Expressão Gênica/genética , Glutationa/biossíntese , Manosiltransferases/genética , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Metionina/farmacologia , Nodaviridae/genética , Proteínas de Saccharomyces cerevisiae/genética , beta-Glucosidase/metabolismo
5.
Ann Clin Microbiol Antimicrob ; 17(1): 24, 2018 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-29859115

RESUMO

BACKGROUND: Acinetobacter baumannii causes difficult-to-treat nosocomial infections, which often lead to morbidity due to the development of antimicrobial drug resistance and expression of virulence genes. Data regarding the association of resistance to colistin, a last treatment option, and the virulence gene expression of A. baumannii is scarce. METHODS: We evaluated the MLVA genotype, antimicrobial resistance, and biofilm formation of 100 A. baumannii isolates from burn patients, and further compared the in vitro and in vivo expression of four virulence genes among five colistin-resistant A. baumannii (Cst-R-AB) isolates. Five Cst-R-AB isolates were tested; one from the present study, and four isolated previously. RESULTS: Our results showed that reduced expression of recA, along with increased in vivo expression of lpsB, dnaK, and blsA; are associated with colistin resistance among Cst-R-AB isolates. Differences in virulence gene expressions among Cst-R-AB isolates, may in part explain common discrepant in vitro vs. in vivo susceptibility data during treatment of infections caused by Cst-R-AB. CONCLUSIONS: Our findings highlight the intricate relationship between colistin-resistance and virulence among A. baumannii isolates, and underscore the importance of examining the interactions between virulence and antimicrobial resistance toward efforts to control the spread of multidrug-resistant A. baumannii (MDR-AB) isolates, and also to reduce disease severity in burn patients with MDR-AB infection.


Assuntos
Acinetobacter baumannii/genética , Acinetobacter baumannii/patogenicidade , Antibacterianos/uso terapêutico , Biofilmes/crescimento & desenvolvimento , Colistina/uso terapêutico , Farmacorresistência Bacteriana Múltipla/genética , Infecções por Acinetobacter/tratamento farmacológico , Infecções por Acinetobacter/microbiologia , Acinetobacter baumannii/efeitos dos fármacos , Proteínas da Membrana Bacteriana Externa/biossíntese , Proteínas da Membrana Bacteriana Externa/genética , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/genética , Queimaduras/microbiologia , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Humanos , Manosiltransferases/biossíntese , Manosiltransferases/genética , Testes de Sensibilidade Microbiana , Recombinases Rec A/biossíntese , Recombinases Rec A/genética , Virulência/genética
6.
PLoS Genet ; 11(3): e1005082, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25807459

RESUMO

Glycosylphosphatidylinositol (GPI) is a post-translational modification resulting in the attachment of modified proteins to the outer leaflet of the plasma membrane. Tissue culture experiments have shown GPI-anchored proteins (GPI-APs) to be targeted to the apical membrane of epithelial cells. However, the in vivo importance of this targeting has not been investigated since null mutations in GPI biosynthesis enzymes in mice result in very early embryonic lethality. Missense mutations in the human GPI biosynthesis enzyme pigv are associated with a multiple congenital malformation syndrome with a high frequency of Hirschsprung disease and renal anomalies. However, it is currently unknown how these phenotypes are linked to PIGV function. Here, we identify a temperature-sensitive hypomorphic allele of PIGV in Caenorhabditis elegans, pigv-1(qm34), enabling us to study the role of GPI-APs in development. At the restrictive temperature we found a 75% reduction in GPI-APs at the surface of embryonic cells. Consequently, ~80% of pigv-1(qm34) embryos arrested development during the elongation phase of morphogenesis, exhibiting internal cysts and/or surface ruptures. Closer examination of the defects revealed them all to be the result of breaches in epithelial tissues: cysts formed in the intestine and excretory canal, and ruptures occurred through epidermal cells, suggesting weakening of the epithelial membrane or membrane-cortex connection. Knockdown of piga-1, another GPI biosynthesis enzymes resulted in similar phenotypes. Importantly, fortifying the link between the apical membrane and actin cortex by overexpression of the ezrin/radixin/moesin ortholog ERM-1, significantly rescued cyst formation and ruptures in the pigv-1(qm34) mutant. In conclusion, we discovered GPI-APs play a critical role in maintaining the integrity of the epithelial tissues, allowing them to withstand the pressure and stresses of morphogenesis. Our findings may help to explain some of the phenotypes observed in human syndromes associated with pigv mutations.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Proteínas do Citoesqueleto/genética , Desenvolvimento Embrionário/genética , Glicosilfosfatidilinositóis/biossíntese , Doença de Hirschsprung/genética , Manosiltransferases/genética , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/crescimento & desenvolvimento , Proteínas de Caenorhabditis elegans/biossíntese , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas do Citoesqueleto/biossíntese , Proteínas do Citoesqueleto/metabolismo , Células Epiteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Glicosilfosfatidilinositóis/genética , Doença de Hirschsprung/metabolismo , Humanos , Manosiltransferases/biossíntese , Manosiltransferases/metabolismo , Camundongos , Mutação , Processamento de Proteína Pós-Traducional/genética , Temperatura
7.
Biosci Biotechnol Biochem ; 78(3): 400-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25036826

RESUMO

We cloned the mouse mannosyltransferase IV/V gene (mALG11) from FM3A cells by a bioinformatic approach. The ORF contained 1476 bp encoding 492 amino acids. The cloned mALG11 complemented the growth defect of the Saccharomyces cerevisiae ALG11Δ mutant. In addition, we detected a variant cDNA by alternate splicing that had an additional four-nucleotide ATGC insertion at base 276 of the ORF. Consequently the variant cDNA encoded a truncated protein with 92 amino acids, lacking the glycosyltransferase group-1 domain. The variant cDNA occurs in many mouse strains according to EST database searches. Moreover, we detected it in FM3A cDNA, but we did not detect any such variants in the human EST database or in HeLa cDNA, although human ALG11 (hALG11) genomic DNA has the same sequence around the intron-exon boundaries as those of mALG11 genomic DNA. Hence, we concluded that there is different transcriptional control mechanism between mALG11 and hALG11.


Assuntos
Clonagem Molecular , Lipopolissacarídeos/biossíntese , Manosiltransferases/genética , Transcrição Gênica , Processamento Alternativo/genética , Animais , DNA Complementar/genética , Células HeLa , Humanos , Lipopolissacarídeos/genética , Manosiltransferases/biossíntese , Camundongos
8.
J Biol Chem ; 289(14): 9865-79, 2014 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-24563464

RESUMO

Human immunodeficiency virus type 1 enhancer-binding protein 3 (Hivep3) suppresses osteoblast differentiation by inducing proteasomal degradation of the osteogenesis master regulator Runx2. In this study, we tested the possibility of cooperation of Hivep1, Hivep2, and Hivep3 in osteoblast and/or chondrocyte differentiation. Microarray analyses with ST-2 bone stroma cells demonstrated that expression of any known osteochondrogenesis-related genes was not commonly affected by the three Hivep siRNAs. Only Hivep3 siRNA promoted osteoblast differentiation in ST-2 cells, whereas all three siRNAs cooperatively suppressed differentiation in ATDC5 chondrocytes. We further used microarray analysis to identify genes commonly down-regulated in both MC3T3-E1 osteoblasts and ST-2 cells upon knockdown of Hivep3 and identified asparagine-linked glycosylation 2 (Alg2), which encodes a mannosyltransferase residing on the endoplasmic reticulum. The Hivep3 siRNA-mediated promotion of osteoblast differentiation was negated by forced Alg2 expression. Alg2 suppressed osteoblast differentiation and bone formation in cultured calvarial bone. Alg2 was immunoprecipitated with Runx2, whereas the combined transfection of Runx2 and Alg2 interfered with Runx2 nuclear localization, which resulted in suppression of Runx2 activity. Chondrocyte differentiation was promoted by Hivep3 overexpression, in concert with increased expression of Creb3l2, whose gene product is the endoplasmic reticulum stress transducer crucial for chondrogenesis. Alg2 silencing suppressed Creb3l2 expression and chondrogenesis of ATDC5 cells, whereas infection of Alg2-expressing virus promoted chondrocyte maturation in cultured cartilage rudiments. Thus, Alg2, as a downstream mediator of Hivep3, suppresses osteogenesis, whereas it promotes chondrogenesis. To our knowledge, this study is the first to link a mannosyltransferase gene to osteochondrogenesis.


Assuntos
Diferenciação Celular/fisiologia , Condrócitos/metabolismo , Proteínas de Ligação a DNA/metabolismo , Manosiltransferases/biossíntese , Osteoblastos/metabolismo , Osteogênese/fisiologia , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Linhagem Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Condrócitos/citologia , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Proteínas de Ligação a DNA/genética , Estresse do Retículo Endoplasmático/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Técnicas de Silenciamento de Genes , Manosiltransferases/genética , Camundongos , Osteoblastos/citologia
9.
PLoS One ; 9(1): e87673, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24489949

RESUMO

The glycosylphosphatidylinositol (GPI) anchor is an essential glycolipid that tethers certain eukaryotic proteins to the cell surface. The core structure of the GPI anchor is remarkably well conserved across evolution and consists of NH2-CH2-CH2-PO4-6Manα1,2Manα1,6Manα1,4-GlcNα1,6-myo-inositol-PO4-lipid. The glycan portion of this structure may be modified with various side-branching sugars or other compounds that are heterogeneous and differ from organism to organism. One such modification is an α(1,2)-linked fourth mannose (Man-IV) that is side-branched to the third mannose (Man-III) of the trimannosyl core. In fungi and mammals, addition of Man-III and Man-IV occurs by two distinct Family 22 α(1,2)-mannosyltransferases, Gpi10/PigB and Smp3/PigZ, respectively. However, in the five protozoan parasite genomes we examined, no genes encoding Smp3/PigZ proteins were observed, despite reports of tetramannosyl-GPI structures (Man4-GPIs) being produced by some parasites. In this study, we tested the hypothesis that the Gpi10/PigB proteins produced by protozoan parasites have the ability to add both Man-III and Man-IV to GPI precursors. We used yeast genetics to test the in vivo specificity of Gpi10/PigB proteins from several Plasmodium and Trypanosoma species by examining their ability to restore viability to Saccharomyces cerevisiae strains harboring lethal defects in Man-III (gpi10Δ) or Man-IV (smp3Δ) addition to GPI precursor lipids. We demonstrate that genes encoding PigB enzymes from T. cruzi, T. congolense and P. falciparum are each capable of separately complementing essential gpi10Δ and smp3Δ mutations, while PIGB genes from T. vivax and T. brucei only complement gpi10Δ. Additionally, we show the ability of T. cruzi PIGB to robustly complement a gpi10Δ/smp3Δ double mutant. Our data suggest that certain Plasmodium and Trypanosoma PigB mannosyltransferases can transfer more than one mannose to GPI precursors in vivo, and suggest a novel biosynthetic mechanism by which Man4-GPIs may be synthesized in these organisms.


Assuntos
Manosiltransferases/genética , Proteínas de Protozoários/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Sequência de Aminoácidos , Sequência Conservada , Deleção de Genes , Teste de Complementação Genética , Glicosilfosfatidilinositóis/biossíntese , Manosiltransferases/biossíntese , Viabilidade Microbiana , Dados de Sequência Molecular , Plasmodium falciparum/enzimologia , Plasmodium falciparum/genética , Proteínas de Protozoários/biossíntese , Saccharomyces cerevisiae/enzimologia , Especificidade por Substrato , Trypanosoma/enzimologia , Trypanosoma/genética
10.
Mol Microbiol ; 80(3): 715-25, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21375589

RESUMO

Protein-O-mannosyltransferases (Pmt) transfer mannosyl residues to secretory proteins. Five isoforms of Pmt proteins in the human fungal pathogen Candida albicans have distinct functions in growth, morphogenesis and antifungal resistance. We found that PMT genes encoding the major isoforms Pmt1, Pmt2, Pmt4 are regulated differently in response to impaired glycostructures. While the PMT1 transcript level increased in cell wall mutants and under inhibition of N-glycosylation by tunicamycin, PMT2 and PMT4 transcripts were upregulated only by inhibition of Pmt1 activity. Reporter fusions revealed specific promoter sequences to be required for PMT1 repression in undamaged cells, which was de-repressed by tunicamycin. Constitutive PMT1 de-repression was observed in mutants lacking the Cek1 MAP kinase and its upstream sensor Msb2. In contrast, in msb2 and cek1 mutants, upregulation of PMT2/PMT4 by Pmt1 inhibition did not occur and basal expression of both transcripts were decreased. We identified Ace2 as a novel transcription factor, which upregulates PMT basal expression and induction in response to glycostructure damage. Mutants lacking Msb2, Cek1 and Ace2 were supersensitive to glycosylation and cell wall inhibitors. We propose that a Msb2, Cek1 and Ace2 signalling pathway addresses PMT genes as downstream targets and that different modes of regulation have evolved for PMT1 and PMT2/PMT4 genes.


Assuntos
Candida albicans/fisiologia , Proteínas Fúngicas/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Regulação Fúngica da Expressão Gênica , Manosiltransferases/biossíntese , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Estresse Fisiológico , Candida albicans/metabolismo , Parede Celular/metabolismo , Proteínas Fúngicas/genética , Proteínas Ativadoras de GTPase/genética , Técnicas de Inativação de Genes , Humanos , Proteína Quinase 3 Ativada por Mitógeno/genética , Transdução de Sinais
11.
Glycobiology ; 20(5): 542-52, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20053626

RESUMO

Protein O-mannosyltransferases (PMTs) initiate O-mannosylation of secretory proteins, which are of fundamental importance in eukaryotes. The human fungal pathogen Aspergillus fumigatus possesses three genes encoding for PMTs, namely, Afpmt1, Afpmt2 and Afpmt4. We have previously shown that lack of AfPmt1 leads to a temperature-sensitive phenotype featured with severe defects in hyphal growth, conidiation, cell wall integrity and morphology at elevated temperatures. In this study, a conditional mutant P2 was constructed by replacing the native promoter of the Afpmt2 with the Aspergillus nidulans alcA promoter. Reduced expression of the Afpmt2 gene led to a lagged germination, retarded hyphal growth, reduced conidiation and defect in cell wall integrity; however, no temperature-sensitive growth was observed. Further analysis revealed that reduced expression of the Afpmt2 caused a failure of the actin re-arrangement. Our results suggest that Afpmt2 gene was required for growth and played a role distinct from that of the Afpmt1 in A. fumigatus.


Assuntos
Aspergillus fumigatus/citologia , Aspergillus fumigatus/enzimologia , Polaridade Celular , Parede Celular , Regulação para Baixo , Manosiltransferases/deficiência , Aspergillus fumigatus/crescimento & desenvolvimento , Manosiltransferases/biossíntese , Manosiltransferases/genética , Mutação , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
J Biol Chem ; 284(32): 21613-25, 2009 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-19520856

RESUMO

Phosphatidyl-myo-inositol mannosyltransferase A (PimA) is an essential glycosyltransferase (GT) involved in the biosynthesis of phosphatidyl-myo-inositol mannosides (PIMs), which are key components of the mycobacterial cell envelope. PimA is the paradigm of a large family of peripheral membrane-binding GTs for which the molecular mechanism of substrate/membrane recognition and catalysis is still unknown. Strong evidence is provided showing that PimA undergoes significant conformational changes upon substrate binding. Specifically, the binding of the donor GDP-Man triggered an important interdomain rearrangement that stabilized the enzyme and generated the binding site for the acceptor substrate, phosphatidyl-myo-inositol (PI). The interaction of PimA with the beta-phosphate of GDP-Man was essential for this conformational change to occur. In contrast, binding of PI had the opposite effect, inducing the formation of a more relaxed complex with PimA. Interestingly, GDP-Man stabilized and PI destabilized PimA by a similar enthalpic amount, suggesting that they formed or disrupted an equivalent number of interactions within the PimA complexes. Furthermore, molecular docking and site-directed mutagenesis experiments provided novel insights into the architecture of the myo-inositol 1-phosphate binding site and the involvement of an essential amphiphatic alpha-helix in membrane binding. Altogether, our experimental data support a model wherein the flexibility and conformational transitions confer the adaptability of PimA to the donor and acceptor substrates, which seems to be of importance during catalysis. The proposed mechanism has implications for the comprehension of the peripheral membrane-binding GTs at the molecular level.


Assuntos
Proteínas de Bactérias/química , Manosiltransferases/biossíntese , Mycobacterium smegmatis/metabolismo , Proteínas de Bactérias/biossíntese , Calorimetria/métodos , Catálise , Membrana Celular/metabolismo , Dicroísmo Circular/métodos , Guanosina Difosfato/química , Manosiltransferases/química , Manosiltransferases/metabolismo , Modelos Biológicos , Modelos Químicos , Conformação Molecular , Desnaturação Proteica , Estrutura Terciária de Proteína , Especificidade por Substrato , Termodinâmica
13.
FEMS Yeast Res ; 7(8): 1248-55, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17627774

RESUMO

ybl006cDelta was selected using an Alcian-blue-based screen aimed to identify nonessential genes involved in the regulation of mannosylphosphorylation. When cells of this deletant were mixed with the cationic dye Alcian blue in a typical assay, they remained white, indicating a low number of mannosylphosphate groups on the cell surface. ybl006cDelta cells did not show any defect in growth rate nor in the glycosylation or secretion rate of the major exoglucanase Exg1. Transcriptome analysis of ybl006cDelta using macroarrays showed at least two-fold changes in the expression of 52 genes (<0.9% of the genome). Three of these have previously been reported to be directly (MNN6 and MNN4) or indirectly (MNN1) implicated in the transfer of mannosylphosphate to the N- and O-oligosaccharides. Alterations in the expression of these genes were confirmed by Northern analysis. YBL006C product was recently identified as a subunit (Rsc14) of the RSC chromatin-remodelling complex of Saccharomyces cerevisiae. It follows that remodelling of chromatin can be an important regulatory mechanism for the maturation of cell wall mannoproteins.


Assuntos
Parede Celular/metabolismo , Proteínas Cromossômicas não Histona/fisiologia , Regulação Fúngica da Expressão Gênica/fisiologia , Manosiltransferases/biossíntese , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/biossíntese , Proteínas de Saccharomyces cerevisiae/biossíntese , Proteínas de Saccharomyces cerevisiae/fisiologia , Saccharomyces cerevisiae/fisiologia , Azul Alciano/metabolismo , Northern Blotting , Proteínas Cromossômicas não Histona/genética , Deleção de Genes , Genes Fúngicos , Glucana 1,3-beta-Glucosidase/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , RNA Fúngico/biossíntese , RNA Mensageiro/biossíntese , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
14.
Am J Pathol ; 170(5): 1659-68, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17456771

RESUMO

Walker-Warburg syndrome (WWS) is the most severe of a group of congenital disorders that have in common defects in the O-glycosylation of alpha-dystroglycan. WWS is characterized by congenital muscular dystrophy coupled with severe ocular and brain malformations. Moreover, in at least one-fifth of the reported cases, mutations in the POMT1 gene are responsible for this disease. During embryonic development (E8.5 to E11.5), the mouse Pomt1 gene is expressed in the tissues most severely affected in WWS, the muscle, eye, and brain. In this study, we show that mPomt1 expression is maintained in the muscle and eye in later developmental stages and, notably, that its expression is particularly strong in regions of brain and cerebellum that, when affected, could generate the defects observed in patients with WWS. We show that the Pomt1 protein is localized to the sarcoplasmic reticulum of muscle tissue cells in adult mice, where alpha-dystroglycan is O-glycosylated. Furthermore, the Pomt1 protein is localized to the acrosome of maturing spermatids, where alpha-dystroglycan is not glycosylated, so that Pomt1 might have a different target for O-mannosylation in the testes. This expression pattern in the testes could also be related to the gonadal anomalies observed in some patients with WWS.


Assuntos
Anormalidades Múltiplas/enzimologia , Encéfalo/enzimologia , Manosiltransferases/biossíntese , Músculo Esquelético/enzimologia , Distrofia Muscular Animal/enzimologia , Acrossomo/enzimologia , Animais , Western Blotting , Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Distroglicanas/metabolismo , Embrião de Mamíferos , Imunofluorescência , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Masculino , Camundongos , Músculo Esquelético/embriologia , Músculo Esquelético/crescimento & desenvolvimento , Miocárdio/enzimologia , RNA Mensageiro/análise , Retículo Sarcoplasmático/enzimologia
15.
Glycobiology ; 16(9): 863-73, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16704966

RESUMO

Mammalian O-mannosylation, although an uncommon type of protein modification, is essential for normal brain and muscle development. Defective O-mannosylation causes congenital muscular dystrophy with abnormal neuronal migration [Walker-Warburg syndrome (WWS)]. Here, we have identified and cloned rat Pomt1 and Pomt2, which are homologues of human POMT1 and POMT2, with identities of 86 and 90%, respectively, at the amino acid level. Coexpression of both genes was found to be necessary for enzymatic activity, as is the case with human POMT1 and POMT2. Northern blot and reverse transcriptase polymerase chain reaction (RT-PCR) analyses revealed that rat Pomt1 and Pomt2 are expressed in all tissues but most strongly in testis. In situ hybridization histochemistry of rat brain revealed that Pomt1 and Pomt2 mRNA are coexpressed in neurons (dentate gyrus and CA1-CA3 region of the hippocampus and cerebellar Purkinje cells). Two transcription-initiation sites were observed in rat Pomt2, resulting in two forms: a testis form and a somatic form. The two forms had equal protein O-mannosyltransferase activity when coexpressed with rat Pomt1. Coexpression studies also showed that the human and rat protein O-mannosyltransferases are interchangeable, providing further evidence for the closeness of their structures.


Assuntos
Regulação da Expressão Gênica , Manosiltransferases/genética , Animais , Sequência de Bases , Encéfalo/citologia , Encéfalo/metabolismo , Clonagem Molecular , Humanos , Masculino , Manosiltransferases/biossíntese , Dados de Sequência Molecular , Especificidade de Órgãos , Filogenia , Ratos , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Testículo/citologia , Testículo/metabolismo
16.
Fungal Genet Biol ; 43(6): 422-9, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16527501

RESUMO

Protein secretion in Trichoderma reesei could be stimulated by overexpression of the yeast Saccharomyces cerevisiae DPM1 gene encoding dolichyl phosphate mannose synthase (DPMS) a key enzyme in the O-glycosylation pathway. The secreted proteins were glycosylated to the wild type level. On the other hand, the elevated concentration of GDP-mannose, a direct substrate for DPMS, resulting from overexpression in T. reesei of the mpg1 gene coding for guanyltransferase, did not affect secretion of proteins but did affect the degree of their O- and N-glycosylation. In this paper, we examined the effects of dolichol, an indispensable carrier of sugar residues in protein glycosylation, on the synthesis of glycosylated proteins. An increase in dolichol synthesis was obtained by overexpression of the yeast gene encoding cis-prenyltransferase, the first enzyme of the mevalonate pathway committed to dolichol biosynthesis. We observed that, an increased concentration of dolichol resulted in an increased expression of the dpm1 gene and DPMS activity and in overglycosylation of secreted proteins.


Assuntos
Dimetilaliltranstransferase/biossíntese , Proteínas de Saccharomyces cerevisiae/biossíntese , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética , Trichoderma/genética , Quitina/metabolismo , Dimetilaliltranstransferase/genética , Dolicóis/metabolismo , Glicosilação , Manosiltransferases/biossíntese , Manosiltransferases/genética , Manosiltransferases/metabolismo , Saccharomyces cerevisiae/crescimento & desenvolvimento , Proteínas de Saccharomyces cerevisiae/genética , Transferases/metabolismo , Trichoderma/enzimologia
17.
Hum Mutat ; 23(5): 477-86, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15108280

RESUMO

A defect of the dolichyl-P-Man:Man5GlcNAc2-PP-dolichyl mannosyltransferase encoded by the ALG3 gene (alias NOT56L) causes congenital disorder of glycosylation type Id (CDG-Id). In this work, a new mutation in the ALG3 gene causing atypical splicing is described with characterization of expression levels and transcript stabilities of the different splice products. A silent mutation in exon 1 of the ALG3 gene (c.165C

Assuntos
Erros Inatos do Metabolismo dos Carboidratos/genética , Códon de Terminação , Glicosilação , Manosiltransferases/genética , Sítios de Splice de RNA , Deleção de Sequência , Sequência de Aminoácidos , Sequência de Bases , Erros Inatos do Metabolismo dos Carboidratos/metabolismo , Códon sem Sentido , Cicloeximida/farmacologia , Análise Mutacional de DNA , Éxons , Humanos , Manosiltransferases/biossíntese , Manosiltransferases/fisiologia , Dados de Sequência Molecular , Precursores de RNA/metabolismo , Splicing de RNA , Estabilidade de RNA/efeitos dos fármacos , RNA Mensageiro/metabolismo
18.
J Biol Chem ; 278(42): 40842-50, 2003 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-12904287

RESUMO

The glycosyl donor, polyprenyl monophosphomannose (PPM), has been shown to be involved in the biosynthesis of the mycobacterial lipoglycans: lipomannan and lipoarabinomannan. The mycobacterial PPM synthase (Mt-ppm1) catalyzes the transfer of mannose from GDP-mannose to polyprenyl phosphates. Based on sequence homology to Mt-ppm1, we have identified the PPM synthase from Corynebacterium glutamicum. In the present study, we demonstrate that the corynebacterial synthase is composed of two distinct domains; a catalytic domain (Cg-ppm1) and a membrane domain (Cg-ppm2). Through the inactivation of Cg-ppm1, we observed a complex phenotype that included altered cell growth rate and inability to synthesize PPM molecules and lipoglycans. When Cg-ppm2 was deleted, no observable phenotype was noted, indicating the clear organization of the two domains. The complementation of the inactivated Cg-ppm1 strain with the corresponding mycobacterial enzyme (Mt-Ppm1/D2) led to the restoration of a wild type phenotype. The present study illustrates, for the first time, the generation of a lipoglycan-less mutant based on a molecular strategy in a member of the Corynebacterianeae family. Lipoglycans are important immunomodulatory molecules involved in determining the outcome of infection, and so the generation of defined mutants and their subsequent immunological characterization is timely.


Assuntos
Proteínas de Bactérias/genética , Corynebacterium/enzimologia , Manose/biossíntese , Manosiltransferases/química , Manosiltransferases/genética , Mutação , Proteínas de Bactérias/química , Cromatografia em Camada Fina , DNA/metabolismo , Eletroforese em Gel de Poliacrilamida , Escherichia coli/metabolismo , Teste de Complementação Genética , Lipopolissacarídeos/metabolismo , Manose/química , Manosiltransferases/biossíntese , Manosiltransferases/metabolismo , Modelos Genéticos , Fenótipo , Filogenia , Plasmídeos/metabolismo , Estrutura Terciária de Proteína , Fatores de Tempo
19.
Glycobiology ; 10(10): 983-91, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11030744

RESUMO

Dolichol phosphate mannose (DPM) synthase activity, which is required in N:-glycosylation, O-mannosylation, and glycosylphosphatidylinositol membrane anchoring of protein, has been postulated to regulate the Trichoderma reesei secretory pathway. We have cloned a T.reesei cDNA that encodes a 243 amino acid protein whose amino acid sequence shows 67% and 65% identity, respectively, to the Schizosaccharomyces pombe and human DPM synthases, and which lacks the COOH-terminal hydrophobic domain characteristic of the Saccharomyces cerevisiae class of synthase. The Trichoderma dpm1 (Trdpm1) gene complements a lethal null mutation in the S.pombe dpm1(+) gene, but neither restores viability of a S.cerevisiae dpm1-disruptant nor complements the temperature-sensitivity of the S. cerevisiae dpm1-6 mutant. The T.reesei DPM synthase is therefore a member of the "human" class of enzyme. Overexpression of Trdpm1 in a dpm1(+)::his7/dpm1(+) S.pombe diploid resulted in a 4-fold increase in specific DPM synthase activity. However, neither the wild type T. reesei DPM synthase, nor a chimera consisting of this protein and the hydrophobic COOH terminus of the S.cerevisiae DPM synthase, complemented an S.cerevisiae dpm1 null mutant or gave active enzyme when expressed in E.coli. The level of the Trdpm1 mRNA in T.reesei QM9414 strain was dependent on the composition of the culture medium. Expression levels of Trdpm1 were directly correlated with the protein secretory capacity of the fungus.


Assuntos
Manosiltransferases/classificação , Trichoderma/enzimologia , Sequência de Aminoácidos , Clonagem Molecular , Escherichia coli/genética , Proteínas Fúngicas/metabolismo , Genes Fúngicos , Humanos , Manosiltransferases/biossíntese , Manosiltransferases/genética , Dados de Sequência Molecular , Estrutura Secundária de Proteína , Proteínas Recombinantes/biossíntese , Saccharomyces cerevisiae/genética , Schizosaccharomyces/genética , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Trichoderma/genética , Regulação para Cima
20.
EMBO J ; 19(11): 2475-82, 2000 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-10835346

RESUMO

Dolichol-phosphate-mannose (DPM) synthase generates mannosyl donors for glycosylphosphatidylinositols, N-glycan and protein O- and C-mannosylation. In Saccharomyces cerevisiae, this enzyme is encoded by DPM1. We reported previously that mammalian DPM synthase contains catalytic DPM1 and regulatory DPM2 subunits, and that DPM1 requires DPM2 for its stable expression in the endoplasmic reticulum. Here we report that human DPM synthase consists of three subunits. The third subunit, DPM3, comprises 92 amino acids associated with DPM1 via its C-terminal domain and with DPM2 via its N-terminal portion. The stability of DPM3 was dependent upon DPM2. However, overexpression of DPM3 in Lec15 cells, a null mutant of DPM2, restored the biosynthesis of DPM with an increase in DPM1, indicating that DPM3 directly stabilized DPM1. Therefore, DPM2 stabilizes DPM3 and DPM3 stabilizes DPM1. DPM synthase activity was 10 times higher in the presence of DPM2, indicating that DPM2 also plays a role in the enzymatic reaction. Schizosaccharomyces pombe has proteins that resemble three human subunits; S.pombe DPM3 restored biosynthesis of DPM in Lec15 cells, indicating its orthologous relationship to human DPM3.


Assuntos
Manosiltransferases/química , Sequência de Aminoácidos , Animais , Sequência de Bases , Células CHO , Cricetinae , Cricetulus , Retículo Endoplasmático/enzimologia , Etiquetas de Sequências Expressas , Proteínas Fúngicas/química , Teste de Complementação Genética , Humanos , Lipossomos , Manosiltransferases/biossíntese , Manosiltransferases/genética , Dados de Sequência Molecular , Conformação Proteica , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Schizosaccharomyces/enzimologia , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...