Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Biotechnol Bioeng ; 120(2): 524-535, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36326175

RESUMO

As one of the most abundant components in human milk oligosaccharides, 2'-fucosyllactose (2'-FL) possesses versatile beneficial health effects. Although most studies focused on overexpressing or fine-tuning the expression of pathway enzymes and achieved a striking increase of 2'-FL production, directly facilitating the metabolic flux toward the key intermediate GDP-l-fucose seems to be ignored. Here, multienzyme complexes consisting of sequential pathway enzymes were constructed by using specific peptide interaction motifs in recombinant Escherichia coli to achieve a higher titer of 2'-FL. Specifically, we first fine-tuned the expression level of pathway enzymes and balanced the metabolic flux toward 2'-FL synthesis. Then, two key enzymes (GDP-mannose 4,6-dehydratase and GDP- l-fucose synthase) were self-assembled into enzyme complexes in vivo via a short peptide interaction pair RIAD-RIDD (RI anchoring disruptor-RI dimer D/D domains), resulting in noticeable improvement of 2'-FL production. Next, to further strengthen the metabolic flux toward 2'-FL, three pathway enzymes were further aggregated into multienzyme assemblies by using another orthogonal protein interaction motif (Spycatcher-SpyTag or PDZ-PDZlig). Intracellular multienzyme assemblies remarkably enlarged the flux toward 2'-FL biosynthesis and showed a 2.1-fold increase of 2'-FL production compared with a strain expressing free-floating and unassembled enzymes. The optimally engineered strain EZJ23 accumulated 4.8 g/L 2'-FL in shake flask fermentation and was capable of producing 25.1 g/L 2'-FL by fed-batch cultivation. This work provides novel approaches for further improvement and large-scale production of 2'-FL and demonstrates the effectiveness of spatial assembly of pathway enzymes to improve the production of valuable products in the engineered host strain.


Assuntos
Escherichia coli , Fucose , Trissacarídeos , Humanos , Escherichia coli/genética , Escherichia coli/metabolismo , Fucose/metabolismo , Guanosina Difosfato Fucose/metabolismo , Engenharia Metabólica/métodos , Complexos Multienzimáticos/metabolismo , Peptídeos/metabolismo , Trissacarídeos/biossíntese
2.
Plant J ; 100(5): 879-891, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31400245

RESUMO

Type 2 diabetes (T2D) affects over 320 million people worldwide. Healthy lifestyles, improved drugs and effective nutraceuticals are different components of a response against the growing T2D epidemic. The specialized metabolite montbretin A (MbA) is being developed for treatment of T2D and obesity due to its unique pharmacological activity as a highly effective and selective inhibitor of the human pancreatic α-amylase. MbA is an acylated flavonol glycoside found in small amounts in montbretia (Crocosmia × crocosmiiflora) corms. MbA cannot be obtained in sufficient quantities for drug development from its natural source or by chemical synthesis. To overcome these limitations through metabolic engineering, we are investigating the genes and enzymes of MbA biosynthesis. We previously reported the first three steps of MbA biosynthesis from myricetin to myricetin 3-O-(6'-O-caffeoyl)-glucosyl rhamnoside (mini-MbA). Here, we describe the sequence of reactions from mini-MbA to MbA, and the discovery and characterization of the gene and enzyme responsible for the glucosylation of mini-MbA. The UDP-dependent glucosyltransferase CcUGT3 (UGT703E1) catalyzes the 1,2-glucosylation of mini-MbA to produce myricetin 3-O-(glucosyl-6'-O-caffeoyl)-glucosyl rhamnoside. Co-expression of CcUGT3 with genes for myricetin and mini-MbA biosynthesis in Nicotiana benthamiana validated its biological function and expanded the set of genes available for metabolic engineering of MbA.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Flavonas/biossíntese , Glucosiltransferases/metabolismo , Hipoglicemiantes/metabolismo , Engenharia Metabólica/métodos , Trissacarídeos/biossíntese , Ácidos Cafeicos/química , Ácidos Cafeicos/metabolismo , Flavonas/química , Flavonas/farmacologia , Flavonas/uso terapêutico , Flavonoides/química , Flavonoides/metabolismo , Flavonóis/química , Flavonóis/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica de Plantas/genética , Glucose/química , Glucose/metabolismo , Glicosídeos/química , Glicosídeos/metabolismo , Glicosilação , Hipoglicemiantes/química , Hipoglicemiantes/farmacologia , Hipoglicemiantes/uso terapêutico , Iridaceae/química , Iridaceae/enzimologia , Filogenia , Proteínas de Plantas/metabolismo , Caules de Planta/química , Caules de Planta/metabolismo , Plantas Geneticamente Modificadas , Ramnose/química , Ramnose/metabolismo , Metabolismo Secundário , Biologia Sintética/métodos , Nicotiana/metabolismo , Transcriptoma/genética , Trissacarídeos/química , Trissacarídeos/farmacologia , Trissacarídeos/uso terapêutico , Xilose/química , Xilose/metabolismo
3.
Plant Physiol ; 180(3): 1277-1290, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31004005

RESUMO

The plant metabolite montbretin A (MbA) and its precursor mini-MbA are potential new drugs for treating type 2 diabetes. These complex acylated flavonol glycosides only occur in small amounts in the corms of the ornamental plant montbretia (Crocosmia × crocosmiiflora). Our goal is to metabolically engineer Nicotiana benthamiana using montbretia genes to achieve increased production of mini-MbA and MbA. Two montbretia UDP-dependent glycosyltransferases (UGTs), CcUGT1 and CcUGT2, catalyze the formation of the first two pathway-specific intermediates in MbA biosynthesis, myricetin 3-O-rhamnoside and myricetin 3-O-glucosyl rhamnoside. In previous work, expression of these UGTs in N. benthamiana resulted in small amounts of kaempferol glycosides but not myricetin glycosides, suggesting that myricetin was limiting. Here, we investigated montbretia genes and enzymes of flavonol biosynthesis to enhance myricetin formation in N. benthamiana We characterized two flavanone hydroxylases, a flavonol synthase, a flavonoid 3'-hydroxylase (F3'H), and a flavonoid 3'5'-hydroxylase (F3'5'H). Montbretia flavonol synthase converted dihydromyricetin into myricetin. Unexpectedly, montbretia F3'5'H shared higher sequence relatedness with F3'Hs in the CYP75B subfamily of cytochromes P450 than with those with known F3'5'H activity. Transient expression of combinations of montbretia flavonol biosynthesis genes and a montbretia MYB transcription factor in N. benthamiana resulted in availability of myricetin for MbA biosynthesis. Transient coexpression of montbretia flavonol biosynthesis genes combined with CcUGT1 and CcUGT2 in N. benthamiana resulted in 2 mg g-1 fresh weight of the MbA pathway-specific compound myricetin 3-O-glucosyl rhamnoside. Additional expression of the montbretia acyltransferase CcAT1 led to detectable levels of mini-MbA in N. benthamiana.


Assuntos
Vias Biossintéticas/genética , Flavonas/biossíntese , Flavonóis/biossíntese , Hipoglicemiantes/metabolismo , Engenharia Metabólica/métodos , Nicotiana/metabolismo , Trissacarídeos/biossíntese , Flavonas/química , Flavonóis/química , Regulação da Expressão Gênica de Plantas , Glicosídeos/química , Glicosídeos/metabolismo , Glicosiltransferases/genética , Glicosiltransferases/metabolismo , Hipoglicemiantes/química , Isoenzimas/genética , Isoenzimas/metabolismo , Quempferóis/química , Quempferóis/metabolismo , Manosídeos/química , Manosídeos/metabolismo , Modelos Químicos , Estrutura Molecular , Oxirredutases/genética , Oxirredutases/metabolismo , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Nicotiana/genética , Trissacarídeos/química
4.
Carbohydr Res ; 472: 1-15, 2019 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-30428394

RESUMO

Galectins are a family of carbohydrate-recognizing proteins that by interacting with specific glycoepitopes can mediate important biological processes, including immune cell homeostasis and activation of tolerogenic circuits. Among the different members of this family, Galectin 1 and 3 have shown pro-tumorigenic effects, being overexpressed in numerous neoplasic diseases, proving to be relevant in tumor immune escape, tumor progression and resistance to drug-induced apoptosis. Thus, generation of specific glycosides that could inhibit their pro-tumorigenic ability by blocking their carbohydrate recognition domain is one of the current major challenges in the field. Considering that galectin-ligand binding strength is closely related to the ligand structure, analysis of this relationship provides valuable information for rational design of high-affinity ligands that could work as effective galectin inhibitors. Taking profit of the ability of glycosidases to catalyze transglycosylation reactions we achieved the enzymatic synthesis of ß-d-Galp-(1 → 6)-ß-d-Galp-(1 → 4)-d-Glcp(2), a mixture of ß-d-Galp-(1 → 6)-ß-d-Glcp-(1 → 4)-d-Glcp(5) and ß-d-Galp-(1 → 3)-ß-d-Glcp-(1 → 4)-d-Glcp(6), and finally benzyl ß-d-galactopyranoside (9), with reaction yields between 16 and 27%. All the galactosides were purified, and characterized using 1H and 13C nuclear magnetic resonance spectroscopy. Docking results performed between the synthesized compounds and human Galectin 1 (hGal-1) and human Galectin 3 (hGal-3) showed that the replacement of a glucose moiety linked to the terminal galactose with a galactose moiety, decreases the affinity for these galectins. Moreover, regarding the interglycosidic bond the most favorable ß-Gal linkage seems to be ß(1 → 4) followed by ß(1 → 3) and ß(1 → 6) for hGal-1, and ß(1 → 4) followed by ß(1 → 6) and ß(1 → 3) for hGal-3. These results were in accordance with the IC50 values obtained with in vitro solid phase inhibition assays. Therefore, docking results obtained in this work proved to be a very good approximation for predicting binding affinity of novel galactosides.


Assuntos
Galactosídeos/biossíntese , Galectinas/antagonistas & inibidores , Glicosídeo Hidrolases/metabolismo , Trissacarídeos/biossíntese , Sítios de Ligação , Proteínas Sanguíneas , Espectroscopia de Ressonância Magnética Nuclear de Carbono-13 , Galactosídeos/química , Galactosídeos/farmacologia , Galectina 1/antagonistas & inibidores , Galectina 1/química , Galectina 3/antagonistas & inibidores , Galectina 3/química , Galectinas/química , Humanos , Modelos Moleculares , Simulação de Acoplamento Molecular , Espectroscopia de Prótons por Ressonância Magnética , Trissacarídeos/química , Trissacarídeos/farmacologia
5.
Appl Microbiol Biotechnol ; 100(15): 6693-6702, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27020280

RESUMO

Globotriose (Galα1-4Galß1-4Glc) is an important cell surface epitope that acts as the receptor for Shiga-like toxins, and it is also the core structure of Globo H and SSEA4 that are tumor-associated glycans. Hence, the enzymatic synthesis of globotriose would be necessary for the development of carbohydrate-based therapeutics for bacterial infections and cancers. Here, a novel GH27 α-galactosidase gene (agaBf3S), a 1521-bp DNA encoding 506 amino acids with a calculated molecular mass of 57.7 kDa, from Bacteroides fragilis NCTC9343 was cloned and heterogeneously expressed in Escherichia coli. The recombinant enzyme AgaBf3S preferentially hydrolyzed p-nitrophenyl-α-D-galactopyranoside (pNPαGal) in all tested nitrophenyl glycosides. It showed maximum activity at pH 4.5 and 40 °C, and it was stable at pH 4.0-11.0 below 40 °C and metal-independent. The K m and k cat values for pNPαGal, melibiose, and globotriose were 1.27 mM and 172.97 S(-1), 62.76 mM and 17.74 S(-1), and 4.62 mM and 388.45 S(-1), respectively. AgaBf3S could transfer galactosyl residue from pNPαGal to lactose (Galß1-4Glc) with high efficiency and strict α1-4 regioselectivity. The effects of initial substrate concentration, pH, temperature, and reaction time on transglycosylation reaction catalyzed by AgaBf3S were studied in detail. AgaBf3S could synthesize globotriose as a single transglycosylation product with a maximum yield of 32.4 % from 20 mM pNPαGal and 500 mM lactose (pH 4.5) at 40 °C for 30 min. This new one-enzyme one-step synthetic reaction is simple, fast, and low cost, which provides a promising alternative to the current synthetic methods for access to pharmaceutically important Galα1-4-linked oligosaccharides.


Assuntos
Bacteroides fragilis/enzimologia , Escherichia coli/metabolismo , Nitrofenilgalactosídeos/metabolismo , Trissacarídeos/biossíntese , alfa-Galactosidase/metabolismo , Sequência de Aminoácidos , Bacteroides fragilis/genética , Escherichia coli/genética , Melibiose/biossíntese , Alinhamento de Sequência , Especificidade por Substrato , alfa-Galactosidase/genética
6.
Appl Biochem Biotechnol ; 173(6): 1405-18, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24801404

RESUMO

Penicillium occitanis xylanase 2 expressed with a His-tag in Pichia pastoris, termed PoXyn2, was immobilized on nickel-chelate Eupergit C by covalent coupling reaction with a high immobilization yield up to 93.49 %. Characterization of the immobilized PoXyn2 was further evaluated. The optimum pH was not affected by immobilization, but the immobilized PoXyn2 exhibited more acidic and large optimum pH range (pH 2.0-4.0) than that of the free PoXyn2 (pH 3.0). The free PoXyn2 had an optimum temperature of 50 °C, whereas that of the immobilized enzyme was shifted to 65 °C. Immobilization increased both pH stability and thermostability when compared with the free enzyme. Time courses of the xylooligosaccharides (XOS) produced from corncob xylan indicated that the immobilized enzyme tends to use shorter xylan chains and to produce more xylobiose and xylotriose initially. At the end of 24-h reaction, XOS mixture contained a total of 21.3 and 34.2 % (w/w) of xylobiose and xylotriose with immobilized xylanase and free xylanase, respectively. The resulting XOS could be used as a special nutrient for lactic bacteria.


Assuntos
Endo-1,4-beta-Xilanases/metabolismo , Proteínas Fúngicas/metabolismo , Glucuronatos/biossíntese , Oligossacarídeos/biossíntese , Penicillium/enzimologia , Biotecnologia , Quelantes , Dissacarídeos/biossíntese , Estabilidade Enzimática , Enzimas Imobilizadas/genética , Enzimas Imobilizadas/metabolismo , Fermentação , Proteínas Fúngicas/genética , Glucuronatos/química , Concentração de Íons de Hidrogênio , Hidrólise , Níquel , Oligossacarídeos/química , Penicillium/genética , Pichia/enzimologia , Pichia/genética , Polímeros , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Temperatura , Trissacarídeos/biossíntese , Xilanos/metabolismo , Zea mays
7.
Eur J Med Chem ; 80: 423-7, 2014 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-24796882

RESUMO

Globotriose is involved in numerous pathogenic processes and drug development strategies. Recent studies have demonstrated that globotriosylceramide could be used in colon cancer therapy and as a crucial indicator for susceptibility to HIV-1 infection. Therefore, the cost-effective and facile approaches for large-scale production of globotiose as potential drugs are highly required. Here, a multi-enzyme one-pot system containing a galactokinase (SpGalK, E.C.2.7.1.6), a UDP-glucose pyrophosphorylase (SpGalU, E.C.2.7.7.9), a α-1,4-galactosyltransferase (LgtC, E.C. 2.4.1.44) and a commercial inorganic pyrophosphatase (PPase, EC 3.6.1.1) was designed to achieve globotriose on preparative scales. This method exploits a cheaper initial substrate, galactose, for donor UDP-galactose production. More importantly, the substrate specificity of SpGalK and SpGalU is highly promiscuous and various UDP-galactose derivatives obtained could be used as the donor substrates for LgtC. This pointcut of rapid preparation of globotriose derivatives is proposed for the first time. Finally, three globotriose analogs were achieved by this one-pot multi-enzyme system in our study.


Assuntos
Galactose/metabolismo , Transferases/metabolismo , Trissacarídeos/biossíntese , Trissacarídeos/química , Escherichia coli/genética , Transferases/genética , Trissacarídeos/isolamento & purificação
8.
Nutr Res ; 33(10): 831-8, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24074741

RESUMO

Human milk oligosaccharides help to prevent infectious diseases in breastfed infants. Larger scale testing, particularly in animal models and human clinical studies, is still limited due to shortened availability of more complex oligosaccharides. The purpose of this study was to evaluate 2'-fucosyllactose (2'-FL) and 3-fucosyllactose (3-FL) synthesized by whole-cell biocatalysis for their biological activity in vitro. Therefore, we have tested these oligosaccharides for their inhibitory potential of pathogen adhesion in two different human epithelial cell lines. 2'-FL could inhibit adhesion of Campylobacter jejuni, enteropathogenic Escherichia coli, Salmonella enterica serovar fyris, and Pseudomonas aeruginosa to the intestinal human cell line Caco-2 (reduction of 26%, 18%, 12%, and 17%, respectively), as could be shown for 3-FL (enteropathogenic E coli 29%, P aeruginosa 26%). Furthermore, adherence of P aeruginosa to the human respiratory epithelial cell line A549 was significantly inhibited by 2'-FL and 3-FL (reduction of 24% and 23%, respectively). These results confirm the biological and functional activity of biotechnologically synthesized human milk oligosaccharides. Mass-tailored human milk oligosaccharides could be used in the future to supplement infant formula ingredients or as preventatives to reduce the impact of infectious diseases.


Assuntos
Antibacterianos/farmacologia , Aderência Bacteriana/efeitos dos fármacos , Bactérias Gram-Negativas/efeitos dos fármacos , Intestinos/efeitos dos fármacos , Leite Humano/química , Sistema Respiratório/efeitos dos fármacos , Trissacarídeos/farmacologia , Antibacterianos/biossíntese , Biocatálise , Bioengenharia , Aleitamento Materno , Células CACO-2 , Campylobacter jejuni/efeitos dos fármacos , Campylobacter jejuni/patogenicidade , Escherichia coli Enteropatogênica/efeitos dos fármacos , Escherichia coli Enteropatogênica/patogenicidade , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/microbiologia , Bactérias Gram-Negativas/patogenicidade , Humanos , Infecções/microbiologia , Intestinos/microbiologia , Oligossacarídeos/biossíntese , Oligossacarídeos/farmacologia , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/patogenicidade , Sistema Respiratório/microbiologia , Salmonella enterica/efeitos dos fármacos , Salmonella enterica/patogenicidade , Trissacarídeos/biossíntese
9.
J Thorac Cardiovasc Surg ; 141(1): 269-75, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21168032

RESUMO

OBJECTIVES: Human subjects and Old World primates have high levels of antibody to galactose-α-1,3 galactose ß-1,4-N-acetylglucosamine (α-Gal). Commercially available bioprosthetic heart valves of porcine and bovine origin retain the Gal antigen despite current processing techniques. Gal-deficient pigs eliminate this xenoantigen. This study tests whether binding of human anti-Gal antibody effects calcification of wild-type and Gal-deficient glutaraldehyde-fixed porcine pericardium by using a standard subcutaneous implant model. METHODS: Expression of α-Gal was characterized by lectin Griffonia simplicifolia-IB4 staining. Glutaraldehyde-fixed pericardial disks from Gal-positive and Gal-deficient pigs were implanted into 12-day-old Wistar rats and 1.5-kg rabbits with and without prelabeling with affinity-purified human anti-Gal antibody. Calcification of the implants was determined after 3 weeks by using inductively coupled plasma spectroscopy. RESULTS: The α-Gal antigen was detected in wild-type but not Gal-deficient porcine pericardium. Wild-type disks prelabeled with human anti-Gal antibody exhibited significantly greater calcification compared with that seen in antibody-free wild-type samples (mean ± standard error of the mean: 111 ± 8.4 and 74 ± 9.6 mg/g, respectively; P = .01). In the presence of anti-Gal antibody, a significantly greater level of calcification was detected in wild-type compared with GTKO porcine pericardium (111 ± 8.4 and 55 ± 11.8 mg/g, respectively; P = .005). Calcification of Gal-deficient pericardium was not affected by the presence of anti-Gal antibody (51 ± 9.1 and 55 ± 11.8 mg/g). CONCLUSIONS: In this model anti-Gal antibody accelerates calcification of wild-type but not Gal-deficient glutaraldehyde-fixed pericardium. This study suggests that preformed anti-Gal antibody present in all patients might contribute to calcification of currently used bioprosthetic heart valves. Gal-deficient pigs might become the preferred source for new, potentially calcium-resistant bioprosthetic heart valves.


Assuntos
Antígenos Heterófilos/imunologia , Bioprótese , Calcinose/imunologia , Implante de Prótese de Valva Cardíaca/instrumentação , Próteses Valvulares Cardíacas , Pericárdio/transplante , Trissacarídeos/imunologia , Animais , Animais Geneticamente Modificados , Antígenos Heterófilos/biossíntese , Autoanticorpos/administração & dosagem , Fixadores , Galactosiltransferases/deficiência , Galactosiltransferases/genética , Glutaral , Implante de Prótese de Valva Cardíaca/efeitos adversos , Humanos , Microscopia de Fluorescência , Pericárdio/imunologia , Lectinas de Plantas , Desenho de Prótese , Coelhos , Ratos , Ratos Wistar , Suínos/genética , Fatores de Tempo , Transplante Heterólogo , Trissacarídeos/biossíntese
10.
Cancer Res ; 70(13): 5259-69, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20530670

RESUMO

Mucin 1 (MUC1), a bound mucin glycoprotein, is overexpressed and aberrantly glycosylated in >80% of human ductal pancreatic carcinoma. Evidence suggests that MUC1 can be used as a tumor marker and is a potential target for immunotherapy of pancreatic cancer. However, vaccination with MUC1 peptides fails to stimulate the immune response against cancer cells because immunity toward tumor-associated antigens (TAA), including MUC1, in cancer patients is relatively weak, and the presentation of these TAAs to the immune system is poor due to their low immunogenicity. We investigated whether vaccination with immunogenetically enhanced MUC1 (by expressing alpha-gal epitopes; Galalpha1-3Galbeta1-4GlcNAc-R) can elicit effective antibody production for MUC1 itself as well as certain TAAs derived from pancreatic cancer cells and induced tumor-specific T-cell responses. We also used alpha1,3galactosyltransferase (alpha1,3GT) knockout mice that were preimmunized with pig kidney and transplanted with B16F10 melanoma cells transfected with MUC1 expression vector. Vaccination of these mice with alpha-gal MUC1 resulted in marked inhibition of tumor growth and significant improvement of overall survival time compared with mice vaccinated with MUC1 alone (P = 0.003). Furthermore, vaccination with pancreatic cancer cells expressing alpha-gal epitopes induced immune responses against not only differentiated cancer cells but also cancer stem cells. The results suggested that vaccination using cells engineered to express alpha-gal epitopes is a novel strategy for treatment of pancreatic cancer.


Assuntos
Vacinas Anticâncer/imunologia , Imunoterapia Ativa/métodos , Mucina-1/imunologia , Neoplasias Pancreáticas/imunologia , Trissacarídeos/imunologia , Animais , Formação de Anticorpos , Linfócitos B/imunologia , Vacinas Anticâncer/genética , Vacinas Anticâncer/farmacologia , Epitopos/biossíntese , Epitopos/genética , Epitopos/imunologia , Humanos , Camundongos , Camundongos Knockout , Mucina-1/biossíntese , Mucina-1/genética , Células-Tronco Neoplásicas/imunologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/terapia , Engenharia de Proteínas/métodos , Suínos , Linfócitos T/imunologia , Transfecção , Trissacarídeos/biossíntese , Trissacarídeos/genética
11.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 41(2): 194-8, 207, 2010 Mar.
Artigo em Chinês | MEDLINE | ID: mdl-20506633

RESUMO

OBJECTIVES: To establish a human lung adenocarcinoma cell subline A549 that can stably express the Chinese Banna minipig inbred-line (BMI) alpha1 ,3-galactosyltransferase (alpha1 ,3GT) gene and alpha-galactosyl (Gala1-3Galb1-4GlcNAc-R, alpha-gal) epitopic, providing a cell model which expressed xenotransplantation antigens for the further research on the effect of complement dependent cytotoxic lysis of the tumor cells triggered by human natural serum. METHODS: The pEGFP-CMV-GT plasmid containing Banna minipig alpha1 ,3-GT gene was ransfected into A549 cells with lipofectin in vitro. After screened with G418,the single clones were got out and then amplified, the stable transfected cells was named A549-GT. The transcription of alpha1, 3-GT gene in A549-GT cells was detected by RT-PCR. Direct immunofluonrescence methods and flow cytometer were performed to observe the expression of alpha-gal and the binding conditions of IgM and complement C3 in human serum on A549-GT cells. The biological characters of A549-GT cells including morphology, proliferation, and tumorigenesis in nude mice were also examined. RESULTS: After G418 screening, A549-GT that stablely transfected with alpha1, 3-GT gene was obtained and has been passaged for 2 years. The expression of alpha1,3-GT mRNA and alpha-gal was detected continuously and stably in A549-GT. The expression rate of alpha-gal positive cells reached 80.1% +/- 3.2%. The binding of human serum IgM and C3 in human serum on A549-GT cells were founded. Compared with parental A549 cells, its biological characteristics did not change. CONCLUSION: A549-GT cell line stably and continuously expressing alpha1, 3-GT and alpha-gal was established successfully. It provided a useful cell model for the further study of pig alpha1,3-GT gene in tumor immunotherapy.


Assuntos
Adenocarcinoma/patologia , Galactosiltransferases/biossíntese , Neoplasias Pulmonares/patologia , Transfecção , Adenocarcinoma/imunologia , Adenocarcinoma/metabolismo , Animais , Linhagem Celular Tumoral , Galactosiltransferases/genética , Vetores Genéticos/genética , Humanos , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Suínos , Porco Miniatura , Trissacarídeos/biossíntese , Trissacarídeos/genética
12.
J Microbiol Biotechnol ; 19(12): 1514-9, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20075612

RESUMO

A gene encoding the xylanase of Bacillus subtilis AMX-4 isolated from soil was cloned into Escherichia coli, and the gene product was purified from the cell-free extract of the recombinant strain. The gene, designated xylA, consisted of 639 nucleotides encoding a polypeptide of 213 residues. The deduced amino acid sequence was highly homologous to those of xylanase belonging to glycosyl hydrolase family 11. The molecular mass of the purified xylanase was 23 kDa as estimated by SDS-PAGE. The enzyme had a pH optimum at 6.0-7.0 and a temperature optimum at 50-55 degrees C. Xylanase activity was significantly inhibited by 5 mM Cu2+ and 5 mM Mn2+, and noticeably enhanced by 5 mM Fe2+. The enzyme was active on xylans including arabinoxylan, birchwood xylan, and oat spelt xylan, but it did not exhibit activity toward carboxymethylcellulose or p-nitrophenyl-beta-xylopyranoside. The predominant products resulting from xylan and xylooligosaccharide hydrolysis were xylobiose and xylotriose. The enzyme could hydrolyze xylooligosaccharides larger than xylotriose.


Assuntos
Bacillus subtilis/enzimologia , Endo-1,4-beta-Xilanases/genética , Endo-1,4-beta-Xilanases/metabolismo , Bacillus subtilis/genética , Clonagem Molecular , Dissacarídeos/biossíntese , Endo-1,4-beta-Xilanases/química , Escherichia coli/metabolismo , Genes Bacterianos , Concentração de Íons de Hidrogênio , Metais Pesados/química , Peso Molecular , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Microbiologia do Solo , Especificidade por Substrato , Temperatura , Trissacarídeos/biossíntese , Xilanos/metabolismo
13.
Gynecol Oncol ; 90(1): 100-8, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12821349

RESUMO

OBJECTIVE: Poor presentation of tumor-associated antigens (TAA) to the immune system remains a major obstacle to effective anti-tumor vaccine therapy. The aim of this study is to demonstrate the feasibility of producing a novel autologous tumor vaccine from ovarian carcinoma that is expected to have increased immunogenicity. The strategy is based on the ability of the anti-Gal IgG antibody (a natural antibody comprising 1% of IgG in humans) to target tumor membranes expressing alpha-gal epitopes (Galalpha1-3Galbeta1-4GlcNAc-R) to antigen-presenting cells (APC). STUDY DESIGN: Freshly obtained ovarian carcinoma tumors are homogenized, washed, and incubated with a mixture of neuraminidase, recombinant alpha1,3 galactosyltransferase (ralpha1,3GT) and uridine diphosphate galactose (UDP-Gal) to synthesize alpha-gal epitopes on carbohydrate chains of glycoproteins of these membranes. Subsequently, the processed membranes are analyzed for expression of alpha-gal epitopes and for the binding of anti-Gal. RESULTS: Incubation of 3 g of ovarian carcinoma membranes, from five different patients, at 100 mg/ml, mixed together with ralpha1,3GT (50 microg/ml), neuraminidase (1 mU/ml), and UDP-Gal (2 mM), resulted in the effective synthesis of alpha-gal epitopes to the extent of approximately 2 x 10(11) epitopes/mg of tumor membranes. As a result of this de novo expression of alpha-gal epitopes, the tumor membranes readily bound purified anti-Gal antibody, as well as anti-Gal in autologous serum. CONCLUSIONS: The method described in this study is very effective in the synthesis of many alpha-gal epitopes on tumor membranes obtained from ovarian carcinoma. These novel epitopes readily bind the naturally occurring anti-Gal antibody. This technique of opsonization of alpha-gal-modified autologous tumor membranes carrying TAA is expected to increase effective uptake of the vaccine by APC, which is key to successful anti-tumor vaccination.


Assuntos
Vacinas Anticâncer/imunologia , Neoplasias Ovarianas/imunologia , Trissacarídeos/biossíntese , Trissacarídeos/imunologia , Células Apresentadoras de Antígenos/imunologia , Sequência de Carboidratos , Membrana Celular/imunologia , Membrana Celular/metabolismo , Ensaio de Imunoadsorção Enzimática , Epitopos/biossíntese , Epitopos/imunologia , Feminino , Galactosiltransferases/metabolismo , Galactosiltransferases/farmacologia , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Glicoproteínas de Membrana/imunologia , Dados de Sequência Molecular , Neuraminidase/metabolismo , Neuraminidase/farmacologia , Neoplasias Ovarianas/metabolismo , Uridina Difosfato Galactose/metabolismo , Uridina Difosfato Galactose/farmacologia
14.
J Am Chem Soc ; 125(19): 5745-53, 2003 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-12733914

RESUMO

To gain initial structure-activity relationships regarding the highly functionalized pentyl side chain attached at C-3 of mithramycin (MTM), we focused on a post-polyketide synthase (post-PKS) tailoring step of the MTM biosynthesis by Streptomyces argillaceus ATCC 12956, which was proposed to be catalyzed by ketoreductase (KR) MtmW. In this last step of the MTM biosynthesis, a keto group of the pentyl side chain is reduced to a secondary alcohol, and we anticipated the generation of an MTM derivative with an additional keto group in the 3-side chain. Insertional inactivation of mtmW, a gene located ca. 8 kb downstream of the mithramycin-PKS genes, yielded an S. argillaceus mutant, which accumulated three new mithramycin analogues, namely mithramycin SA, demycarosyl-mithramycin SK, and mithramycin SK (MTM-SK). The structures of these three compounds confirmed indirectly the proposed role of MtmW in MTM biosynthesis. However, the new mithramycin derivatives bear unexpectedly shorter 3-side chains (ethyl or butyl) than MTM, presumably caused by nonenzymatic rearrangement or cleavage reactions of the initially formed pentyl side chain with a reactive beta-dicarbonyl functional group. The major product, MTM-SK, was tested in vitro against a variety of human cancer cell lines, as well as in an in vitro toxicity assay, and showed an improved therapeutic index, in comparison to the parent drug, MTM.


Assuntos
Antibióticos Antineoplásicos/biossíntese , Plicamicina/análogos & derivados , Streptomyces/metabolismo , Antibióticos Antineoplásicos/farmacologia , Sequência de Carboidratos , Técnicas de Química Combinatória , Ensaios de Seleção de Medicamentos Antitumorais , Inativação Gênica , Humanos , Dados de Sequência Molecular , Mutagênese Insercional , Ressonância Magnética Nuclear Biomolecular/métodos , Oxirredutases/genética , Oxirredutases/metabolismo , Plicamicina/biossíntese , Plicamicina/farmacologia , Streptomyces/enzimologia , Streptomyces/genética , Trissacarídeos/biossíntese , Trissacarídeos/metabolismo , Células Tumorais Cultivadas
15.
Biosci Biotechnol Biochem ; 67(3): 646-50, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12723619

RESUMO

We isolated a beta-N-acetylglucosaminidase encoding gene from the filamentous fungus Aspergillus oryzae, and designated it nagA. The nagA gene encoded a polypeptide of 600 amino acids with significant similarity to glucosaminidases and hexosaminidases of various eukaryotes. A. oryzae strain carrying the nagA gene under the control of the improved glaA promoter produced large amounts of beta-N-acetylglucosaminidase in a wheat bran solid culture. The beta-N-acetylglucosaminidase was purified from crude extracts of the solid culture by column chromatographies on Q-Sepharose and Sephacryl S-200. This enzyme was used for synthesis of lacto-N-triose II, which is contained in human milk. By reverse hydrolysis reaction, lacto-N-triose II and its positional isomer were synthesized from lactose and D-N-acetylglucosamine in 0.21% and 0.15% yield, respectively.


Assuntos
Acetilglucosaminidase/genética , Acetilglucosaminidase/metabolismo , Aspergillus oryzae/enzimologia , Aspergillus oryzae/genética , Leite Humano/metabolismo , Trissacarídeos/biossíntese , Acetilglucosamina/metabolismo , Acetilglucosaminidase/biossíntese , Acetilglucosaminidase/isolamento & purificação , Sequência de Aminoácidos , Sequência de Carboidratos , Catálise , Clonagem Molecular , Genes Fúngicos/genética , Humanos , Hidrólise , Isomerismo , Lactose/metabolismo , Dados de Sequência Molecular , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Trissacarídeos/química
16.
Biochemistry ; 41(38): 11479-87, 2002 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-12234191

RESUMO

Three key regulatory enzymes in ganglioside biosynthesis, sialyltransferase I (ST1), sialyltransferase II (ST2), and N-acetylgalactosaminyltransferase I (GalNAcT), have been expressed as fusion proteins with green, yellow, or red fluorescent protein (GFP, YFP, or RFP) in F-11A cells. F-11A cells are a substrain of murine neuroblastoma F-11 cells that contain only low endogenous ST2 and GalNAcT activity. The subcellular localization of the fusion proteins has been determined by fluorescence microscopy, and the ganglioside composition of these cells was analyzed by high-performance thin-layer chromatography (HPTLC). ST2-GFP (85 kDa) shows a distinct Golgi localization, whereas ST1-YFP (85 kDa) and GalNAcT-RFP (115 kDa) are broadly distributed in ER and Golgi. Untransfected F-11A cells contain mainly GM3, whereas stable transfection with ST2 or GalNAcT results in the predominant expression of b-series complex gangliosides (BCGs). This result indicates that the expression of ST2 enhances the activity of endogenous GalNAcT and vice versa. The specificity of this reaction has been verified by in vitro activity assays with detergent-solubilized enzymes, suggesting the formation of an enzyme complex between ST2 and GalNAcT but not with ST1. Complex formation has also been verified by co-immunoprecipitation of ST2-GFP upon transient transfection with GalNAcT-HA-RFP and by GFP-to-RFP FRET signals that are confined to the Golgi. FRET analysis also suggests that ST2-GFP binds tightly to pyrene-labeled GM3 but not to ST1. We hypothesize that an ST2-GM3 complex is associated with GalNAcT, resulting in the enhanced conversion of GM3 to GD3 and BCGs in the Golgi. Taken together, our results support the concept that ganglioside biosynthesis is tightly regulated by the formation of glycosyltransferase complexes in the ER and/or Golgi.


Assuntos
Gangliosídeos/biossíntese , Glicosiltransferases/metabolismo , Sialiltransferases/metabolismo , Animais , Sequência de Bases , Sequência de Carboidratos , Clonagem Molecular , Técnicas de Cocultura , Primers do DNA , DNA Complementar , Gânglios Espinais/metabolismo , Genes Reporter , Proteínas de Fluorescência Verde , Homeostase , Proteínas Luminescentes/genética , Camundongos , Dados de Sequência Molecular , N-Acetilgalactosaminiltransferases/metabolismo , Neuroblastoma , Ratos , Proteínas Recombinantes/metabolismo , Transfecção , Trissacarídeos/biossíntese , Trissacarídeos/química , Células Tumorais Cultivadas
17.
Mol Ther ; 6(2): 252-7, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12161192

RESUMO

Expression of retrovirally transduced genes in bone marrow-derived cells can be used to establish stable long-term B- and T-cell tolerance. To determine whether preexisting antibodies may prohibit the use of gene therapy to establish tolerance, we examined the extent to which preexisting antibodies specific for the carbohydrate antigen Gal alpha1-3Gal beta1-4GlcNAc-R (alpha Gal) could affect engraftment and development of bone marrow progenitors expressing the enzyme UDPgalactose:beta-D-galactosyl-1,4-N-acetyl-D-glucosaminide alpha(1-3)galactosyltransferase (E.C. 2.4.1.151), or simply alpha GT, which synthesizes the alpha Gal epitope. Groups of alpha GT knockout mice (GT(0) mice) lacking alpha Gal were presensitized to alpha Gal by immunization and then lethally irradiated and reconstituted with varying numbers of alpha GT-transduced syngeneic bone marrow cells. Whereas unimmunized controls were reconstituted with as few as 2 x 10(6) transduced cells, a significant fraction of immunized mice reconstituted with 2 x 10(6) or 4 x 10(6) alpha GT-transduced cells failed to undergo bone marrow engraftment and died. Immunized mice in which radiation protection was achieved failed to express alpha Gal. However, radiation protection and expression of alpha Gal on bone marrow-derived cells, resulting in tolerance, could be achieved by increasing the number of transduced cells used to reconstitute immunized mice. Thus, although high levels of preexisting antibodies can be a significant barrier to engraftment, this barrier can be overcome by increasing the number of transduced cells used for reconstitution.


Assuntos
Transplante de Medula Óssea/imunologia , Animais , Formação de Anticorpos , Linfócitos B/imunologia , Galactosiltransferases/deficiência , Galactosiltransferases/genética , Expressão Gênica , Terapia Genética/efeitos adversos , Terapia Genética/métodos , Sobrevivência de Enxerto/imunologia , Tolerância Imunológica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Suínos , Porco Miniatura , Transplante Heterólogo , Trissacarídeos/biossíntese , Trissacarídeos/imunologia
18.
J Am Chem Soc ; 124(8): 1606-14, 2002 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-11853433

RESUMO

Mithramycin is an aureolic acid-type antimicrobial and antitumor agent produced by Streptomyces argillaceus. Modifying post-polyketide synthase (PKS) tailoring enzymes involved in the production of mithramycin is an effective way of gaining further information regarding the late steps of its biosynthetic pathway. In addition, new "unnatural" natural products of the aureolic acid-type class are likely to be produced. The role of two such post-PKS tailoring enzymes, encoded by mtmC and mtmTIII, was investigated, and four novel aureolic acid class drugs, two premithramycin-type molecules and two mithramycin derivatives, were isolated from mutant strains constructed by insertional gene inactivation of either of these two genes. From data bank comparisons, the corresponding proteins MtmC and MtmTIII were believed to act as a C-methyltransferase involved in the production of the D-mycarose (sugar E) of mithramycin and as a ketoreductase seemingly involved in the biosynthesis of the mithramycin aglycon, respectively. However, gene inactivation and analysis of the accumulated products revealed that both genes encode enzymes participating in the biosynthesis of the D-mycarose building block. Furthermore, the inactivation of MtmC seems to affect the ketoreductase responsible for 4-ketoreduction of sugar C, a D-olivose. Instead of obtaining premithramycin and mithramycin derivatives with a modified E-sugar upon inactivation of mtmC, compounds were obtained that completely lack the E-sugar moiety and that possess an unexpected 4-ketosugar moiety instead of the D-olivose at the beginning of the lower deoxysaccharide chain. The inactivation of mtmTIII led to the accumulation of 4E-ketomithramycin, showing that this ketoreductase is responsible for the 4-ketoreduction of the D-mycarose moiety. The new compounds of the mutant strains, 4A-ketopremithramycin A2, 4A-keto-9-demethylpremithramycin A2, 4C-keto-demycarosylmithramycin, and 4E-ketomithramycin, indicate surprising substrate flexibility of post-PKS enzymes of the mithramycin biosynthetic pathway. Although the glycosyltransferase responsible for the attachment of D-mycarose cannot transfer the unmethylated sugar to the existing lower disaccharide chain, it can transfer the 4-ketoform of sugar E. In addition, the glycosyltransferase MtmGIV, which is responsible for the linkage of sugar C, is also able to transfer an activated 4-ketosugar. The oxygenase MtmOIV, normally responsible for the oxidative cleavage of the tetracyclic premithramycin B into the tricyclic immediate precursor of mithramycin, can act on a substrate analogue with a modified or even incomplete trisaccharide chain. The same is true for glycosyltransferases MtmGI and MtmGII, both of which partake in the formation and attachment of the A-B disaccharide in mithramycin.


Assuntos
Antibióticos Antineoplásicos/biossíntese , Hexoses/metabolismo , Complexos Multienzimáticos/metabolismo , Plicamicina/biossíntese , Streptomyces/genética , Streptomyces/metabolismo , Trissacarídeos/biossíntese , Sequência de Carboidratos , Regulação Bacteriana da Expressão Gênica , Inativação Gênica , Metiltransferases/genética , Metiltransferases/metabolismo , Dados de Sequência Molecular
19.
J Biol Chem ; 272(37): 23104-10, 1997 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-9287311

RESUMO

The binding of xenoreactive natural antibodies to the Galalpha1-3Galbeta1-4GlcNAc (alpha-galactose) oligosaccharide epitope on pig cells activates the recipient's complement system in pig to primate xenotransplantation. Expression of human alpha-1, 2-fucosyltransferase in pigs has been proposed as a strategy for reducing the expression level of the alpha-galactose epitope, thereby rendering the pig organs more suitable for transplantation into humans. The aim of this study was to examine how the cell surface expression of alpha-galactose, H, and related fucosylated and sialylated structures on a pig liver endothelial cell line is affected by transfection of human alpha-1,2-fucosyltransferase cDNA. Nontransfected and mock-transfected cells expressed alpha-galactose, alpha-2,3-sialylated, and alpha-2,6-sialylated epitopes strongly, with low level expression of type 2 H and LewisX. By contrast, expression of the H epitope was increased 5-8-fold in transfected cells with a 40% reduction in the expression of alpha-galactose epitope and a 50% decrease in sialylation, as measured by binding of Maackia amurensis and Sambuccus nigra agglutinins. LewisX expression was reduced to background levels, while the LewisY neoepitope was induced in human alpha-1,2-fucosyltransferase-expressing pig cells. The activities of endogenous alpha-1,3-galactosyltransferase, alpha-1,3-fucosyltransferases, and alpha-2,3- and alpha-2, 6-sialyltransferases acting on lactosamine were unaffected. Our results show that a reduction in alpha-galactose epitope expression in porcine endothelial cells transfected with human alpha-1, 2-fucosyltransferase cDNA may be achieved but at the expense of considerable distortion of the overall cell surface glycosylation profile, including the appearance of carbohydrate epitopes that are absent from the parent cells.


Assuntos
Endotélio/imunologia , Epitopos/biossíntese , Fucosiltransferases/metabolismo , Galactosídeos/biossíntese , Oligossacarídeos/biossíntese , Trissacarídeos/biossíntese , Sistema ABO de Grupos Sanguíneos/biossíntese , Animais , Linhagem Celular , Clonagem Molecular , DNA Complementar , Endotélio/citologia , Endotélio/metabolismo , Citometria de Fluxo , Fucosiltransferases/genética , Galactosídeos/imunologia , Engenharia Genética/métodos , Glicosilação , Humanos , Imunoglobulina M/imunologia , Lectinas/metabolismo , Antígenos do Grupo Sanguíneo de Lewis/biossíntese , Antígenos CD15/biossíntese , Fígado/citologia , Fígado/imunologia , Fígado/metabolismo , Masculino , Oligossacarídeos/imunologia , Ligação Proteica , Proteínas Recombinantes/metabolismo , Suínos , Transplante de Tecidos , Transfecção , Trissacarídeos/imunologia , Galactosídeo 2-alfa-L-Fucosiltransferase
20.
Plant J ; 12(5): 1057-65, 1997 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9418047

RESUMO

By screening a cDNA library of artichoke (Cynara scolymus) blossom discs for fructosyltransferases, we isolated a clone designated Cy21. The deduced amino acid sequence shows homology to acid beta-fructosyl hydrolases and to the sucrose-fructan 6-fructosyltransferase (6-SFT) of barley. Transiently expressed in Nicotiana tabacum protoplasts, the Cy21 gene-product synthesized 1-kestose, indicating that Cy21 codes for a sucrose sucrose 1-fructosyltransferase (1-SST). The enzyme worked at physiologically relevant sucrose concentrations (25 mM sucrose). In the protoplast system, 1-kestose seemed to be the only fructan product of the 1-SST. The enzyme activity was not affected by pyridoxal-HCl, an inhibitor of both the beta-fructosyl hydrolase and the fructosyltransferase activity of invertases. The fructosyltransferase activity of the Cy21 gene-product, however, could be inhibited by Zn2+, Ag+ and Cu2+ ions. In artichoke plants the Cy21 transcript was highly abundant in primary roots and blossom discs. Transgenic potato tubers expressing Cy21 contain high levels of 1-kestose along with nystose and traces of fructosyl-nystose, supporting the conclusion that the Cy21 clone encodes a sucrose sucrose 1-fructosyltransferase.


Assuntos
Hexosiltransferases/metabolismo , Oligossacarídeos/biossíntese , Solanum tuberosum/metabolismo , Trissacarídeos/biossíntese , Verduras/enzimologia , Sequência de Aminoácidos , Cátions Bivalentes/farmacologia , Clonagem Molecular , Inibidores Enzimáticos/farmacologia , Biblioteca Gênica , Hexosiltransferases/biossíntese , Hexosiltransferases/química , Cinética , Dados de Sequência Molecular , Raízes de Plantas , Plantas Geneticamente Modificadas , Plantas Tóxicas , Protoplastos/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Sacarose/metabolismo , Nicotiana , Transfecção , Verduras/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA