Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 355
Filtrar
1.
Sci Transl Med ; 16(728): eadg7740, 2024 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-38170790

RESUMO

Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer with poor prognosis. TNBCs with high homologous recombination deficiency (HRD) scores benefit from DNA-damaging agents, including platinum drugs and poly(ADP-ribose) polymerase (PARP) inhibitors, whereas those with low HRD scores still lack therapeutic options. Therefore, we sought to exploit metabolic alterations to induce HRD and sensitize DNA-damaging agents in TNBCs with low HRD scores. We systematically analyzed TNBC metabolomics and identified a metabolite, guanosine diphosphate (GDP)-mannose (GDP-M), that impeded homologous recombination repair (HRR). Mechanistically, the low expression of the upstream enzyme GDP-mannose-pyrophosphorylase-A (GMPPA) led to the endogenous up-regulation of GDP-M in TNBC. The accumulation of GDP-M in tumor cells further reduced the interaction between breast cancer susceptibility gene 2 (BRCA2) and ubiquitin-specific peptidase 21 (USP21), which promoted the ubiquitin-mediated degradation of BRCA2 to inhibit HRR. Therapeutically, we illustrated that the supplementation of GDP-M sensitized DNA-damaging agents to impair tumor growth in both in vitro (cancer cell line and patient-derived organoid) and in vivo (xenograft in immunodeficient mouse) models. Moreover, the combination of GDP-M with DNA-damaging agents activated STING-dependent antitumor immunity in immunocompetent syngeneic mouse models. Therefore, GDP-M supplementation combined with PARP inhibition augmented the efficacy of anti-PD-1 antibodies. Together, these findings suggest that GDP-M is a crucial HRD-related metabolite and propose a promising therapeutic strategy for TNBCs with low HRD scores using the combination of GDP-M, PARP inhibitors, and anti-PD-1 immunotherapy.


Assuntos
Neoplasias de Mama Triplo Negativas , Animais , Camundongos , Humanos , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Reparo de DNA por Recombinação , Manose/uso terapêutico , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico , Recombinação Homóloga , Guanosina Difosfato Manose , Proteína BRCA1/genética , DNA , Ubiquitina Tiolesterase/metabolismo
2.
Physiol Plant ; 173(3): 1244-1252, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34380178

RESUMO

Cell-wall polysaccharides are synthesized from nucleotide sugars by glycosyltransferases. However, in what way the level of nucleotide sugars affects the structure of the polysaccharides is not entirely clear. guanosine diphosphate (GDP)-mannose (GDP-Man) is one of the major nucleotide sugars in plants and serves as a substrate in the synthesis of mannan polysaccharides. GDP-Man is synthesized from mannose 1-phosphate and GTP by a GDP-Man pyrophosphorylase, VITAMIN C DEFECTIVE1 (VTC1), which is positively regulated by the interacting protein KONJAC1 (KJC1) in Arabidopsis. Since seed-coat mucilage can serve as a model of the plant cell wall, we examined the influence of vtc1 and kjc1 mutations on the synthesis of mucilage galactoglucomannan. Sugar composition analysis showed that mannose content in adherent mucilage of kjc1 and vtc1 mutants was only 42% and 11% of the wild-type, respectively, indicating a drastic decrease of galactoglucomannan. On the other hand, structural analysis based on specific oligosaccharides released by endo-ß-1,4-mannanase indicated that galactoglucomannan had a patterned glucomannan backbone consisting of alternating residues of glucose and mannose and the frequency of α-galactosyl branches was also similar to the wild type structure. These results suggest that the structure of mucilage galactoglucomannan is mainly determined by properties of glycosyltransferases rather than the availability of nucleotide sugars.


Assuntos
Proteínas de Arabidopsis , Arabidopsis , Arabidopsis/genética , Proteínas de Arabidopsis/genética , Guanosina Difosfato Manose , Mananas , Manose , Polissacarídeos , Sementes
3.
NMR Biomed ; 34(10): e4576, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34155714

RESUMO

Guanosine diphosphate mannose (GDP-Man) is the donor substrate required for mannosylation in the synthesis of glycoproteins, glycolipids and the newly discovered glycoRNA. Normal GDP-Man biosynthesis plays a crucial role in support of a variety of cellular functions, including cell recognition, cell communication and immune responses against viruses. Here, we report the detection of GDP-Man in human brain for the first time, using 31 P MRS at 7 T. The presence of GDP-Man is evidenced by the detection of a weak 31 P doublet at -10.7 ppm that can be assigned to the phosphomannosyl group (Pß) of the GDP-Man molecule. This weak but well-resolved signal lies 0.9 ppm upfield of UDP(G) Pß-multiplet from a mixture of UDP-Glc, UDP-Gal, UDP-GlcNAc and UDP-GalNAc. In reference to ATP (2.8 mM), the concentration of GDP-Man in human brain was estimated to be 0.02 ± 0.01 mM, about 15-fold lower than the total concentration of UDP(G) (0.30 ± 0.04, N = 17) and consistent with previous reports of UDP-Man in cells and brain tissue extracts measured by high-performance liquid chromatography. The reproducibility of the measured GDP-Man between test and 2-week retest was 21% ± 15% compared with 5% ± 4% for UDP(G) (N = 7). The measured concentrations of GDP-Man and UDP(G) are linearly correlated ([UDP(G)] = 4.3 [GDP-Man] + 0.02, with R = 0.66 and p = 0.0043), likely reflecting the effect of shared sugar precursors, which may vary among individuals in response to variation in nutritional intake and consumption. Given that GDP-Man has another set of doublet (Pα) at -8.3 ppm that overlaps with NAD(H) and UDP(G)-Pα signals, the amount of GDP-Man could potentially interfere with the deconvolution of these mixed signals in composition analysis. Importantly, this new finding may be useful in advancing our understanding of glycosylation and its role in the development of cancer, as well as infectious and neurodegenerative diseases.


Assuntos
Encéfalo/diagnóstico por imagem , Guanosina Difosfato Manose/análise , Espectroscopia de Ressonância Magnética , Adulto , Idoso , Feminino , Guanosina Difosfato Manose/química , Humanos , Masculino , Pessoa de Meia-Idade , Fósforo , Reprodutibilidade dos Testes , Difosfato de Uridina/metabolismo , Adulto Jovem
4.
Nat Struct Mol Biol ; 28(5): 1-12, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33986552

RESUMO

GDP-mannose (GDP-Man) is a key metabolite essential for protein glycosylation and glycophosphatidylinositol anchor synthesis, and aberrant cellular GDP-Man levels have been associated with multiple human diseases. How cells maintain homeostasis of GDP-Man is unknown. Here, we report the cryo-EM structures of human GMPPA-GMPPB complex, the protein machinery responsible for GDP-Man synthesis, in complex with GDP-Man or GTP. Unexpectedly, we find that the catalytically inactive subunit GMPPA displays a much higher affinity to GDP-Man than the active subunit GMPPB and, subsequently, inhibits the catalytic activity of GMPPB through a unique C-terminal loop of GMPPA. Importantly, disruption of the interactions between GMPPA and GMPPB or the binding of GDP-Man to GMPPA in zebrafish leads to abnormal brain development and muscle abnormality, analogous to phenotypes observed in individuals carrying GMPPA or GMPPB mutations. We conclude that GMPPA acts as a cellular sensor to maintain mannose homeostasis through allosterically regulating GMPPB.


Assuntos
Guanosina Difosfato Manose/metabolismo , Nucleotidiltransferases , Animais , Microscopia Crioeletrônica , Células HEK293 , Humanos , Nucleotidiltransferases/química , Nucleotidiltransferases/metabolismo , Ligação Proteica , Domínios Proteicos , Peixe-Zebra
5.
J Clin Invest ; 131(9)2021 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-33755596

RESUMO

GDP-mannose-pyrophosphorylase-B (GMPPB) facilitates the generation of GDP-mannose, a sugar donor required for glycosylation. GMPPB defects cause muscle disease due to hypoglycosylation of α-dystroglycan (α-DG). Alpha-DG is part of a protein complex, which links the extracellular matrix with the cytoskeleton, thus stabilizing myofibers. Mutations of the catalytically inactive homolog GMPPA cause alacrima, achalasia, and mental retardation syndrome (AAMR syndrome), which also involves muscle weakness. Here, we showed that Gmppa-KO mice recapitulated cognitive and motor deficits. As structural correlates, we found cortical layering defects, progressive neuron loss, and myopathic alterations. Increased GDP-mannose levels in skeletal muscle and in vitro assays identified GMPPA as an allosteric feedback inhibitor of GMPPB. Thus, its disruption enhanced mannose incorporation into glycoproteins, including α-DG in mice and humans. This increased α-DG turnover and thereby lowered α-DG abundance. In mice, dietary mannose restriction beginning after weaning corrected α-DG hyperglycosylation and abundance, normalized skeletal muscle morphology, and prevented neuron degeneration and the development of motor deficits. Cortical layering and cognitive performance, however, were not improved. We thus identified GMPPA defects as the first congenital disorder of glycosylation characterized by α-DG hyperglycosylation, to our knowledge, and we have unraveled underlying disease mechanisms and identified potential dietary treatment options.


Assuntos
Distroglicanas , Guanosina Difosfato Manose , Músculo Esquelético/metabolismo , Doenças Neuromusculares , Nucleotidiltransferases/deficiência , Animais , Distroglicanas/genética , Distroglicanas/metabolismo , Glicosilação , Guanosina Difosfato Manose/genética , Guanosina Difosfato Manose/metabolismo , Humanos , Camundongos , Camundongos Knockout , Doenças Neuromusculares/dietoterapia , Doenças Neuromusculares/genética , Doenças Neuromusculares/metabolismo , Nucleotidiltransferases/metabolismo
6.
Mol Microbiol ; 116(1): 245-259, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33629421

RESUMO

Aspergillus fumigatus is an opportunistic mold responsible for severe life-threatening fungal infections in immunocompromised patients. The cell wall, an essential structure composed of glucan, chitin, and galactomannan, is considered to be a target for the development of antifungal drugs. The nucleotide sugar donor GDP-mannose (GDP-Man) is required for the biosynthesis of galactomannan, glycosylphosphatidylinositol (GPI) anchors, glycolipid, and protein glycosylation. Starting from fructose-6-phosphate, GDP-Man is produced by the sequential action of the enzymes phosphomannose isomerase, phosphomannomutase (Pmm), and GDP-mannose pyrophosphorylase. Here, using heterokaryon rescue and gene knockdown approaches we demonstrate that the phosphomannomutase encoding gene in A. fumigatus (pmmA) is essential for survival. Reduced expression of pmmA is associated with significant morphological defects including retarded germination, growth, reduced conidiation, and abnormal polarity. Moreover, the knockdown strain exhibited an altered cell wall organization and sensitivity toward cell wall perturbing agents. By solving the first crystal structure of A. fumigatus phosphomannomutase (AfPmmA) we identified non-conservative substitutions near the active site when compared to the human orthologues. Taken together, this work provides a genetic and structural foundation for the exploitation of AfPmmA as a potential antifungal target.


Assuntos
Aspergillus fumigatus/genética , Guanosina Difosfato Manose/metabolismo , Fosfotransferases (Fosfomutases)/genética , Fosfotransferases (Fosfomutases)/metabolismo , Antifúngicos/farmacologia , Aspergilose/tratamento farmacológico , Aspergilose/patologia , Aspergillus fumigatus/efeitos dos fármacos , Aspergillus fumigatus/metabolismo , Parede Celular/metabolismo , Deleção de Genes , Humanos , Virulência/genética
7.
Plant Sci ; 302: 110676, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33288001

RESUMO

Ascorbic acid (AsA) is an antioxidant and enzyme co-factor that is vital to plant development and abiotic stress tolerance. However, the regulation mechanisms of AsA biosynthesis in plants remain poorly understood. Here, we report a basic helix-loop-helix 55 (ZmbHLH55) transcription factor that regulates AsA biosynthesis in maize. Analysis of publicly available transcriptomic data revealed that ZmbHLH55 is co-expressed with several genes of the GDP-mannose pathway. Experimental data showed that ZmbHLH55 forms homodimers localized to the cell nuclei, and it exhibits DNA binding and transactivation activity in yeast. Under salt stress conditions, knock down mutant (zmbhlh55) in maize accumulated lower levels of AsA compared with wild type, accompanied by lower antioxidant enzymes activity, shorter root length, and higher malondialdehyde (MDA) level. Gene expression data from the WT and zmbhlh55 mutant, showed that ZmbHLH55 positively regulates the expression of ZmPGI2, ZmGME1, and ZmGLDH, but negatively regulates ZmGMP1 and ZmGGP. Furthermore, ZmbHLH55-overexpressing Arabidopsis, under salt conditions, showed higher AsA levels, increased rates of germination, and elevated antioxidant enzyme activities. In conclusion, these results have identified previously unknown regulation mechanisms for AsA biosynthesis, indicating that ZmbHLH55 may be a potential candidate to enhance plant salt stress tolerance in the future.


Assuntos
Ácido Ascórbico/biossíntese , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Genes de Plantas/fisiologia , Guanosina Difosfato Manose/metabolismo , Redes e Vias Metabólicas/genética , Proteínas de Plantas/fisiologia , Tolerância ao Sal , Zea mays/metabolismo , Ácido Ascórbico/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Clonagem Molecular , Ensaio de Desvio de Mobilidade Eletroforética , Regulação da Expressão Gênica de Plantas/fisiologia , Técnicas de Silenciamento de Genes , Genes de Plantas/genética , Malondialdeído/metabolismo , Redes e Vias Metabólicas/fisiologia , Filogenia , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Tolerância ao Sal/genética , Tolerância ao Sal/fisiologia , Técnicas do Sistema de Duplo-Híbrido , Zea mays/genética , Zea mays/fisiologia
8.
Int J Biol Macromol ; 165(Pt B): 1862-1868, 2020 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-33075338

RESUMO

GDP-mannose 3,5-epimerase (GM35E) catalyzes the double epimerization of GDP-mannose to yield GDP-l-galactose. GDP-l-gulose (C5-epimer) has previously been detected as a byproduct of this reaction, indicating that C3,5-epimerization occurs through an initial epimerization at C5. Given these products, GM35E constitutes a valuable bridge between d- and l-hexoses. In order to fully exploit this potential, the enzyme might be subjected to specificity engineering for which profound mechanistic insights are beneficial. Accordingly, this study further elucidated GM35E's reaction mechanism. For the first time, the production of the C3-epimer GDP-altrose was demonstrated, resulting in an adjustment of the acknowledged reaction mechanism. As GM35E converts GDP-mannose to GDP-l-gulose, GDP-altrose and GDP-l-galactose in a 72:4:4:20 ratio, this indicates that the enzyme does not discriminate between the C3 and C5 position as initial epimerization site. This was also confirmed by a structural investigation. Based on a mutational analysis of the active site, residues S115 and R281 were attributed a stabilizing function, which is believed to support the reactivation process of the catalytic residues. This paper eventually reflected on some engineering strategies that aim to change the enzyme towards a single specificity.


Assuntos
Guanosina Difosfato Manose/metabolismo , Guanosina Difosfato/metabolismo , Hexoses/metabolismo , Racemases e Epimerases/metabolismo , Biocatálise , Domínio Catalítico , Guanosina Difosfato/química , Hexoses/química , Engenharia de Proteínas , Especificidade por Substrato
9.
Microb Cell Fact ; 19(1): 32, 2020 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-32054531

RESUMO

BACKGROUND: Bleomycin is a broad-spectrum glycopeptide antitumor antibiotic produced by Streptomyces verticillus. Clinically, the mixture of bleomycin A2 and bleomycin B2 is widely used in combination with other drugs for the treatment of various cancers. As a secondary metabolite, the biosynthesis of bleomycin is precisely controlled by the complex extra-/intracellular regulation mechanisms, it is imperative to investigate the global metabolic and regulatory system involved in bleomycin biosynthesis for increasing bleomycin production. RESULTS: N-acetylglucosamine (GlcNAc), the vital signaling molecule controlling the onset of development and antibiotic synthesis in Streptomyces, was found to increase the yields of bleomycins significantly in chemically defined medium. To mine the gene information relevant to GlcNAc metabolism, the DNA sequences of dasR-dasA-dasBCD-nagB and nagKA in S. verticillus were determined by chromosome walking. From the results of Real time fluorescence quantitative PCR (RT-qPCR) and electrophoretic mobility shift assays (EMSAs), the repression of the expression of nagB and nagKA by the global regulator DasR was released under induction with GlcNAc. The relief of blmT expression repression by BlmR was the main reason for increased bleomycin production. DasR, however, could not directly affect the expression of the pathway-specific repressor BlmR in the bleomycins gene cluster. With at the beginning of bleomycin synthesis, the supply of the specific precursor GDP-mannose played the key role in bleomycin production. Genetic engineering of the GDP-mannose synthesis pathway indicated that phosphomannose isomerase (ManA) and phosphomannomutase (ManB) were key enzymes for bleomycins synthesis. Here, the blmT, manA and manB co-expression strain OBlmT/ManAB was constructed. Based on GlcNAc regulation and assisted metabolic profiling analysis, the yields of bleomycin A2 and B2 were ultimately increased to 61.79 and 36.9 mg/L, respectively. CONCLUSIONS: Under GlcNAc induction, the elevated production of bleomycins was mainly associated with the alleviation of the inhibition of BlmT, so blmT and specific precursor synthesis pathways were genetically engineered for bleomycins production improvement. Combination with subsequent metabolomics analysis not only effectively increased the bleomycin yield, but also extended the utilization of chitin-derived substrates in microbial-based antibiotic production.


Assuntos
Acetilglucosamina/metabolismo , Bleomicina/biossíntese , Guanosina Difosfato Manose/metabolismo , Streptomyces/metabolismo , Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica , Redes e Vias Metabólicas , Metabolômica/métodos , Metabolismo Secundário
10.
Neuromuscul Disord ; 29(8): 614-617, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31378432

RESUMO

Mutations in GMPPB cause a wide spectrum of neuromuscular syndromes, including muscular dystrophies and congenital myasthenic syndrome. The mechanisms by which GMPPB mutations impair neuromuscular transmission however remain incompletely understood. We expand here upon a previous report of one such patient presenting with a myopathy-congenital myasthenic syndrome overlap phenotype. Fatigable proximal muscle weakness developed gradually between 13 and 25 years of age, with subsequent stabilization. Low-frequency repetitive nerve stimulation showed a decrement, while a muscle biopsy demonstrated the presence of a centronuclear myopathy. Genetic testing identified a homozygous c.458C > T (p.Thr153Ile) variant in GMPPB. In-vitro microelectrode recordings and ultrastructural studies showed impairment of both pre- and postsynaptic neuromuscular transmission, thus demonstrating the presence of not only postsynaptic, but also presynaptic pathology in GMPPB-related disorders.


Assuntos
Guanosina Difosfato Manose/genética , Síndromes Miastênicas Congênitas/genética , Miopatias Congênitas Estruturais/genética , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Síndromes Miastênicas Congênitas/diagnóstico , Síndromes Miastênicas Congênitas/fisiopatologia , Miopatias Congênitas Estruturais/diagnóstico , Miopatias Congênitas Estruturais/fisiopatologia
11.
Int J Mol Sci ; 20(14)2019 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-31330931

RESUMO

GDP-mannose 3,5-epimerase (GM35E) catalyzes the conversion of GDP-mannose towards GDP-l-galactose and GDP-l-gulose. Although this reaction represents one of the few enzymatic routes towards the production of l-sugars and derivatives, it has not yet been exploited for that purpose. One of the reasons is that so far only GM35Es from plants have been characterized, yielding biocatalysts that are relatively unstable and difficult to express heterologously. Through the mining of sequence databases, we succeeded in identifying a promising bacterial homologue. The gene from the thermophilic organism Methylacidiphilum fumariolicum was codon optimized for expression in Escherichia coli, resulting in the production of 40 mg/L of recombinant protein. The enzyme was found to act as a self-sufficient GM35E, performing three chemical reactions in the same active site. Furthermore, the biocatalyst was highly stable at temperatures up to 55 °C, making it well suited for the synthesis of new carbohydrate products with application in the pharma industry.


Assuntos
Proteínas de Bactérias , Carboidratos Epimerases/química , Carboidratos Epimerases/metabolismo , Sequência de Aminoácidos , Catálise , Ativação Enzimática , Estabilidade Enzimática , Guanosina Difosfato Manose/química , Guanosina Difosfato Manose/metabolismo , Concentração de Íons de Hidrogênio , Modelos Moleculares , Estrutura Molecular , Conformação Proteica , Proteínas Recombinantes , Relação Estrutura-Atividade , Termodinâmica
12.
Int J Mol Med ; 44(1): 262-272, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31115488

RESUMO

Congenital disorder of glycosylation (CDG) type Ia is a multisystem disorder that occurs due to mutations in the phosphomannomutase 2 (PMM2) gene, which encodes for an enzyme involved in the N­glycosylation pathway. Mutated PMM2 leads to the reduced conversion of mannose­6­P to mannose­1­P, which results in low concentration levels of guanosine 5'­diphospho­D­mannose (GDP­Man), a nucleotide­activated sugar essential for the construction of protein oligosaccharide chains. In the present study, an in vitro therapeutic approach was used, based on GDP­Man­loaded poly (D,L­lactide­co­glycolide) (PLGA) nanoparticles (NPs), which were used to treat CDG­Ia fibroblast cultures, thus bypassing the glycosylation pathway reaction catalysed by PMM2. To assess the degree of hypoglycosylation in vitro, the present study examined the activities of α­mannosidase, ß­glucoronidase and ß­galactosidase in defective and normal fibroblasts. GDP­Man (30 µg/ml GDP­Man PLGA NPs) was incubated for 48 h with the cells and the specific activities of α­mannosidase and ß­galactosidase were estimated at 69 and 92% compared with healthy controls. The residual activity of ß­glucoronidase increased from 6.5 to 32.5% and was significantly higher compared with that noted in the untreated CDG­Ia fibroblasts. The glycosylation process of fibroblasts was also analysed by two­dimensional electrophoresis. The results demonstrated that treatment caused the reappearance of several glycosylated proteins. The data in vitro showed that GDP­Man PLGA NPs have desirable efficacy and warrant further evaluation in a preclinical validation animal model.


Assuntos
Defeitos Congênitos da Glicosilação/tratamento farmacológico , Portadores de Fármacos , Guanosina Difosfato Manose , Nanopartículas , Fosfotransferases (Fosfomutases)/deficiência , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Células Cultivadas , Defeitos Congênitos da Glicosilação/genética , Defeitos Congênitos da Glicosilação/metabolismo , Defeitos Congênitos da Glicosilação/patologia , Portadores de Fármacos/química , Portadores de Fármacos/farmacologia , Fibroblastos , Glicosilação/efeitos dos fármacos , Guanosina Difosfato Manose/química , Guanosina Difosfato Manose/farmacologia , Humanos , Nanopartículas/química , Nanopartículas/uso terapêutico , Fosfotransferases (Fosfomutases)/genética , Fosfotransferases (Fosfomutases)/metabolismo , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/farmacologia
13.
Am J Hum Genet ; 103(6): 1030-1037, 2018 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-30503518

RESUMO

FUK encodes fucokinase, the only enzyme capable of converting L-fucose to fucose-1-phosphate, which will ultimately be used for synthesizing GDP-fucose, the donor substrate for all fucosyltransferases. Although it is essential for fucose salvage, this pathway is thought to make only a minor contribution to the total amount of GDP-fucose. A second pathway, the major de novo pathway, involves conversion of GDP-mannose to GDP-fucose. Here we describe two unrelated individuals who have pathogenic variants in FUK and who presented with severe developmental delays, encephalopathy, intractable seizures, and hypotonia. The first individual was compound heterozygous for c.667T>C (p.Ser223Pro) and c.2047C>T (p.Arg683Cys), and the second individual was homozygous for c.2980A>C (p.Lys994Gln). Skin fibroblasts from the first individual confirmed the variants as loss of function and showed significant decreases in total GDP-[3H] fucose and [3H] fucose-1-phosphate. There was also a decrease in the incorporation of [5,6-3H]-fucose into fucosylated glycoproteins. Lys994 has previously been shown to be an important site for ubiquitin conjugation. Here, we show that loss-of-function variants in FUK cause a congenital glycosylation disorder characterized by a defective fucose-salvage pathway.


Assuntos
Anormalidades Congênitas/genética , Variação Genética/genética , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Sequência de Aminoácidos , Encefalopatias/genética , Criança , Deficiências do Desenvolvimento/genética , Feminino , Fibroblastos/patologia , Fucosiltransferases/genética , Glicosilação , Guanosina Difosfato Fucose/genética , Guanosina Difosfato Manose/genética , Humanos , Masculino , Hipotonia Muscular/genética , Convulsões/genética , Alinhamento de Sequência , Pele/patologia , Ubiquitina/genética
14.
Microbiology (Reading) ; 164(4): 614-624, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29493491

RESUMO

Actinomycete bacteria use polyprenol phosphate mannose as a lipid linked sugar donor for extra-cytoplasmic glycosyl transferases that transfer mannose to cell envelope polymers, including glycoproteins and glycolipids. We showed recently that strains of Streptomyces coelicolor with mutations in the gene ppm1 encoding polyprenol phosphate mannose synthase were both resistant to phage φC31 and have greatly increased susceptibility to antibiotics that mostly act on cell wall biogenesis. Here we show that mutations in the genes encoding enzymes that act upstream of Ppm1 in the polyprenol phosphate mannose synthesis pathway can also confer phage resistance and antibiotic hyper-susceptibility. GDP-mannose is a substrate for Ppm1 and is synthesised by GDP-mannose pyrophosphorylase (GMP; ManC) which uses GTP and mannose-1-phosphate as substrates. Phosphomannomutase (PMM; ManB) converts mannose-6-phosphate to mannose-1-phosphate. S. coelicolor strains with knocked down GMP activity or with a mutation in sco3028 encoding PMM acquire phenotypes that resemble those of the ppm1- mutants i.e. φC31 resistant and susceptible to antibiotics. Differences in the phenotypes of the strains were observed, however. While the ppm1- strains have a small colony phenotype, the sco3028 :: Tn5062 mutants had an extremely small colony phenotype indicative of an even greater growth defect. Moreover we were unable to generate a strain in which GMP activity encoded by sco3039 and sco4238 is completely knocked out, indicating that GMP is also an important enzyme for growth. Possibly GDP-mannose is at a metabolic branch point that supplies alternative nucleotide sugar donors.


Assuntos
Antibacterianos/farmacologia , Vias Biossintéticas , Guanosina Difosfato Manose/metabolismo , Nucleotidiltransferases/genética , Fosfotransferases (Fosfomutases)/genética , Streptomyces coelicolor/efeitos dos fármacos , Streptomyces coelicolor/fisiologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Bacteriófagos/fisiologia , Manosiltransferases/genética , Manosiltransferases/metabolismo , Mutação , Nucleotidiltransferases/metabolismo , Fenótipo , Fosfotransferases (Fosfomutases)/metabolismo , Streptomyces coelicolor/virologia
15.
J Neurol Neurosurg Psychiatry ; 89(7): 762-768, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29437916

RESUMO

BACKGROUND: Defects in glycosylation of alpha-dystroglycan (α-DG) cause autosomal-recessive disorders with wide clinical and genetic heterogeneity, with phenotypes ranging from congenital muscular dystrophies to milder limb girdle muscular dystrophies. Patients show variable reduction of immunoreactivity to antibodies specific for glycoepitopes of α-DG on a muscle biopsy. Recessive mutations in 18 genes, including guanosine diphosphate mannose pyrophosphorylase B (GMPPB), have been reported to date. With no specific clinical and pathological handles, diagnosis requires parallel or sequential analysis of all known genes. METHODS: We describe clinical, genetic and biochemical findings of 21 patients with GMPPB-associated dystroglycanopathy. RESULTS: We report eight novel mutations and further expand current knowledge on clinical and muscle MRI features of this condition. In addition, we report a consistent shift in the mobility of beta-dystroglycan (ß-DG) on Western blot analysis of all patients analysed by this mean. This was only observed in patients with GMPPB in our large dystroglycanopathy cohort. We further demonstrate that this mobility shift in patients with GMPPB was due to abnormal N-linked glycosylation of ß-DG. CONCLUSIONS: Our data demonstrate that a change in ß-DG electrophoretic mobility in patients with dystroglycanopathy is a distinctive marker of the molecular defect in GMPPB.


Assuntos
Distroglicanas/metabolismo , Guanosina Difosfato Manose/genética , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Mutação/genética , Nucleotidiltransferases/genética , Adolescente , Idoso , Biomarcadores/metabolismo , Criança , Pré-Escolar , Estudos de Coortes , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Distrofias Musculares/patologia
16.
Med Mycol ; 56(5): 621-630, 2018 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-29420778

RESUMO

Systemic human fungal infections are increasingly common. Aspergillus species cause most of the airborne fungal infections. Life-threatening invasive aspergillosis was formerly found only in immune-suppressed patients, but recently some strains of A. fumigatus have become primary pathogens. Many fungal cell wall components are absent from mammalian systems, so they are potential drug targets. Cell-wall-targeting drugs such as echinocandins are used clinically, although echinocandin-resistant strains were discovered shortly after their introduction. Currently there are no fully effective anti-fungal drugs. Fungal cell wall glycoconjugates modulate human immune responses, as well as fungal cell adhesion, biofilm formation, and drug resistance. Guanosine diphosphate (GDP) mannose transporters (GMTs) transfer GDP-mannose from the cytosol to the Golgi lumen prior to mannosylation. Aspergillus nidulans GMTs are encoded by gmtA and gmtB. Here we elucidate the roles of A. nidulans GMTs. Strains engineered to lack either or both GMTs were assessed for hyphal and colonial morphology, cell wall ultrastructure, antifungal susceptibility, spore hydrophobicity, adherence and biofilm formation. The gmt-deleted strains had smaller colonies with reduced sporulation and with thicker hyphal walls. The gmtA deficient spores had reduced hydrophobicity and were less adherent and less able to form biofilms in vitro. Thus, gmtA not only participates in maintaining the cell wall integrity but also plays an important role in biofilm establishment and adherence of A. nidulans. These findings suggested that GMTs have roles in A. nidulans growth and cell-cell interaction and could be a potential target for new antifungals that target virulence determinants.


Assuntos
Aspergillus nidulans/crescimento & desenvolvimento , Aspergillus nidulans/genética , Proteínas de Transporte/genética , Proteínas Fúngicas/genética , Antifúngicos/farmacologia , Aspergillus nidulans/ultraestrutura , Biofilmes/crescimento & desenvolvimento , Adesão Celular/fisiologia , Parede Celular/ultraestrutura , Equinocandinas/farmacologia , Guanosina Difosfato Manose/metabolismo , Hifas/crescimento & desenvolvimento , Lipopeptídeos/farmacologia , Micafungina , Testes de Sensibilidade Microbiana , Viabilidade Microbiana/efeitos dos fármacos , Morfogênese , Deleção de Sequência , Esporos Fúngicos/crescimento & desenvolvimento , Esporos Fúngicos/ultraestrutura
17.
Biotechnol Bioeng ; 115(1): 192-205, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28922469

RESUMO

Glycosylation of proteins is a key function of the biosynthetic-secretory pathway in the endoplasmic reticulum (ER) and Golgi apparatus. Glycosylated proteins play a crucial role in cell trafficking and signaling, cell-cell adhesion, blood-group antigenicity, and immune response. In addition, the glycosylation of proteins is an important parameter in the optimization of many glycoprotein-based drugs such as monoclonal antibodies. In vitro glycoengineering of proteins requires glycosyltransferases as well as expensive nucleotide sugars. Here, we present a designed pathway consisting of five enzymes, glucokinase (Glk), phosphomannomutase (ManB), mannose-1-phosphate-guanyltransferase (ManC), inorganic pyrophosphatase (PmPpA), and 1-domain polyphosphate kinase 2 (1D-Ppk2) expressed in E. coli for the cell-free production and regeneration of GDP-mannose from mannose and polyphosphate with catalytic amounts of GDP and ADP. It was shown that GDP-mannose is produced at various conditions, that is pH 7-8, temperature 25-35°C and co-factor concentrations of 5-20 mM MgCl2 . The maximum reaction rate of GDP-mannose achieved was 2.7 µM/min at 30°C and 10 mM MgCl2 producing 566 nmol GDP-mannose after a reaction time of 240 min. With respect to the initial GDP concentration (0.8 mM) this is equivalent to a yield of 71%. Additionally, the cascade was coupled to purified, transmembrane-deleted Alg1 (ALG1ΔTM), the first mannosyltransferase in the ER-associated lipid-linked oligosaccharide (LLO) assembly. Thereby, in a one-pot reaction, phytanyl-PP-(GlcNAc)2 -Man1 was produced with efficient nucleotide sugar regeneration for the first time. Phytanyl-PP-(GlcNAc)2 -Man1 can serve as a substrate for the synthesis of LLO for the cell-free in vitro glycosylation of proteins. A high-performance anion exchange chromatography method with UV and conductivity detection (HPAEC-UV/CD) assay was optimized and validated to determine the enzyme kinetics. The established kinetic model enabled the optimization of the GDP-mannose regenerating cascade and can further be used to study coupling of the GDP-mannose cascade with glycosyltransferases. Overall, the study envisages a first step towards the development of a platform for the cell-free production of LLOs as precursors for in vitro glycoengineering of proteins.


Assuntos
Enzimas/metabolismo , Escherichia coli/genética , Guanosina Difosfato Manose/metabolismo , Lipopolissacarídeos/metabolismo , Proteínas Recombinantes/metabolismo , Coenzimas/metabolismo , Enzimas/genética , Enzimas/isolamento & purificação , Escherichia coli/metabolismo , Expressão Gênica , Concentração de Íons de Hidrogênio , Cloreto de Magnésio/metabolismo , Manose/metabolismo , Polifosfatos/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Temperatura
18.
Carbohydr Res ; 452: 91-96, 2017 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-29080432

RESUMO

We have previously developed a new class of inhibitors and chemical probes for glycosyltransferases through base-modification of the sugar-nucleotide donor. The key feature of these donor analogues is the presence of an additional substituent at the nucleobase. To date, the application of this general concept has been limited to UDP-sugars and UDP-sugar-dependent glycosyltransferases. Herein, we report for the first time the application of our approach to a GDP-mannose-dependent mannosyltransferase. We have prepared four GDP-mannose derivatives with an additional substituent at either position 6 or 8 of the nucleobase. These donor analogues were recognised as donor substrates by the mannosyltransferase Kre2p from yeast, albeit with significantly lower turnover rates than the natural donor GDP-mannose. The presence of the additional substituent also redirected enzyme activity from glycosyl transfer to donor hydrolysis. Taken together, our results suggest that modification of the donor nucleobase is, in principle, a viable strategy for probe and inhibitor development against GDP-mannose-dependent GTs.


Assuntos
Guanosina Difosfato Manose/metabolismo , Manosiltransferases/metabolismo , Saccharomyces cerevisiae/metabolismo , Guanosina Difosfato Manose/química , Especificidade por Substrato
19.
Biochem J ; 474(6): 897-905, 2017 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-28104756

RESUMO

The obligate intracellular lifestyle of Plasmodium falciparum and the difficulties in obtaining sufficient amounts of biological material have hampered the study of specific metabolic pathways in the malaria parasite. Thus, for example, the pools of sugar nucleotides required to fuel glycosylation reactions have never been studied in-depth in well-synchronized asexual parasites or in other stages of its life cycle. These metabolites are of critical importance, especially considering the renewed interest in the presence of N-, O-, and other glycans in key parasite proteins. In this work, we adapted a liquid chromatography tandem mass spectrometry (LC-MS/MS) method based on the use of porous graphitic carbon (PGC) columns and MS-friendly solvents to quantify sugar nucleotides in the malaria parasite. We report the thorough quantification of the pools of these metabolites throughout the intraerythrocytic cycle of P. falciparum The sensitivity of the method enabled, for the first time, the targeted analysis of these glycosylation precursors in gametocytes, the parasite sexual stages that are transmissible to the mosquito vector.


Assuntos
Guanosina Difosfato Fucose/metabolismo , Guanosina Difosfato Manose/metabolismo , Açúcares de Guanosina Difosfato/metabolismo , Plasmodium falciparum/metabolismo , Uridina Difosfato Galactose/metabolismo , Uridina Difosfato Glucose/metabolismo , Uridina Difosfato N-Acetilgalactosamina/metabolismo , Cromatografia Líquida , Eritrócitos/parasitologia , Gametogênese/fisiologia , Guanosina Difosfato Fucose/análise , Guanosina Difosfato Manose/análise , Açúcares de Guanosina Difosfato/análise , Humanos , Estágios do Ciclo de Vida/fisiologia , Plasmodium falciparum/crescimento & desenvolvimento , Espectrometria de Massas em Tandem , Uridina Difosfato Galactose/análise , Uridina Difosfato Glucose/análise , Uridina Difosfato N-Acetilgalactosamina/análise
20.
Microb Cell Fact ; 15(1): 142, 2016 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-27527078

RESUMO

BACKGROUND: In this study we examine the integrity of the cell wall during scale up of a yeast fermentation process from laboratory scale (10 L) to industrial scale (10,000 L). In a previous study we observed a clear difference in the volume fraction occupied by yeast cells as revealed by wet cell weight (WCW) measurements between these scales. That study also included metabolite analysis which suggested hypoxia during scale up. Here we hypothesize that hypoxia weakens the yeast cell wall during the scale up, leading to changes in cell permeability, and/or cell mechanical resistance, which in turn may lead to the observed difference in WCW. We tested the cell wall integrity by probing the cell wall sensitivity to Zymolyase. Also exometabolomics data showed changes in supply of precursors for the glycosylation pathway. RESULTS: The results show a more sensitive cell wall later in the production process at industrial scale, while the sensitivity at early time points was similar at both scales. We also report exometabolomics data, in particular a link with the protein glycosylation pathway. Significantly lower levels of Man6P and progressively higher GDP-mannose indicated partially impaired incorporation of this sugar nucleotide during co- or post-translational protein glycosylation pathways at the 10,000 L compared to the 10 L scale. This impairment in glycosylation would be expected to affect cell wall integrity. Although cell viability from samples obtained at both scales were similar, cells harvested from 10 L bioreactors were able to re-initiate growth faster in fresh shake flask media than those harvested from the industrial scale. CONCLUSIONS: The results obtained help explain the WCW differences observed at both scales by hypoxia-triggered weakening of the yeast cell wall during the scale up.


Assuntos
Parede Celular/fisiologia , Fermentação , Saccharomyces cerevisiae/crescimento & desenvolvimento , Saccharomyces cerevisiae/metabolismo , Anaerobiose , Técnicas de Cultura Celular por Lotes , Reatores Biológicos , Glicosilação , Guanosina Difosfato Manose/metabolismo , Hidrolases/metabolismo , Microbiologia Industrial , Manosefosfatos/metabolismo , Metaboloma , Processamento de Proteína Pós-Traducional , Saccharomyces cerevisiae/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA