Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Mol Microbiol ; 115(6): 1086-1093, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33434389

RESUMO

The deoxyhexose sugar L-fucose is important for many biological processes within the human body and the associated microbiota. This carbohydrate is abundant in host gut mucosal surfaces, numerous microbial cell surface structures, and some dietary carbohydrates. Fucosylated oligosaccharides facilitate the establishment of a healthy microbiota and provide protection from infection. However, there are instances where pathogens can also exploit these fucosylated structures to cause infection. Furthermore, deficiencies in host fucosylation are associated with specific disease outcomes. This review focuses on our current understanding of the impact of fucosylation within the mucosal environment of the gastrointestinal tract with a specific emphasis on the mediatory effects in host-microbe interactions.


Assuntos
Bactérias/metabolismo , Fucose/metabolismo , Microbioma Gastrointestinal/fisiologia , Interações entre Hospedeiro e Microrganismos/fisiologia , Mucosa Intestinal/metabolismo , Animais , Carboidratos da Dieta/metabolismo , Trato Gastrointestinal/fisiologia , Glicosilação , Humanos , Camundongos
2.
Cell Microbiol ; 23(1): e13269, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32975882

RESUMO

Endogenous carbohydrates released from the intestinal mucus represent a constant source of nutrients to the intestinal microbiota. Mucus-derived carbohydrates can also be used as building blocks in the biosynthesis of bacterial cell wall components, thereby influencing host mucosal immunity. To assess the uptake of endogenous carbohydrates by gut microbes in healthy mice and during intestinal inflammation, we applied azido-monosaccharides that can be tracked on bacterial cell walls after conjugation with fluorophores. In interleukin-10 deficient mice, changes in the gut microbiota were accompanied by decreased carbohydrate hydrolase activities and increased lumenal concentrations of host glycan-derived monosaccharides. Tracking of the monosaccharide N-azidoacetylglucosamine (GlcNAz) in caecum bacteria revealed a preferential incorporation of this carbohydrate by Xanthomonadaceae in healthy mice and by Bacteroidaceae in interleukin-10 deficient mice. These GlcNAz-positive Bacteroidaceae fractions mainly belonged to the species B. acidifaciens and B. vulgatus. Growth of Bacteroides species in the presence of specific monosaccharides changed their stimulatory activity toward CD11c+ dendritic cells. Expression of activation markers and cytokine production was highest after stimulation of dendritic cells with B. vulgatus. The variable incorporation of monosaccharides by related Bacteroides species underline the necessity to investigate intestinal bacteria down to the species level when addressing microbiota-host interactions.


Assuntos
Células Dendríticas/metabolismo , Microbioma Gastrointestinal , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Monossacarídeos/metabolismo , Polissacarídeos/metabolismo , Animais , Bacteroides/metabolismo , Metabolismo dos Carboidratos , Parede Celular/metabolismo , Interações entre Hospedeiro e Microrganismos , Hidrolases/metabolismo , Imunidade nas Mucosas , Inflamação/metabolismo , Interleucina-10/genética , Interleucina-10/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Xanthomonadaceae/metabolismo
3.
Trends Biochem Sci ; 41(6): 508-518, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27093946

RESUMO

The importance of breast milk for the growing infant is undisputed; breastfeeding decreases infantile mortality by tenfold and decreases the incidence of infectious diseases. Despite its recognized benefits, the structural richness of breast milk has also impeded the characterization of the multiple effects of milk components on infant physiology. However, the important roles of some components of breast milk are beginning to be dissected. For instance, molecules such as immunoglobulin A (IgA) and milk oligosaccharides protect from gastrointestinal infections and influence the development of the gut microbiota. Deciphering the complex composition of breast milk brings to light multifaceted contributions that combine to make breast milk the ultimate personalized medicine.


Assuntos
Aleitamento Materno , Citocinas/imunologia , Microbioma Gastrointestinal/imunologia , Lactalbumina/imunologia , Leite Humano/imunologia , Sequência de Carboidratos , Citocinas/genética , Feminino , Galactosiltransferases/genética , Galactosiltransferases/imunologia , Regulação da Expressão Gênica , Humanos , Imunidade , Imunoglobulina A/biossíntese , Lactente , Lactalbumina/genética , Receptores de Lipopolissacarídeos/genética , Receptores de Lipopolissacarídeos/imunologia , Leite Humano/química , Oligossacarídeos/biossíntese , Oligossacarídeos/imunologia , Transportador 1 de Glucose-Sódio/genética , Transportador 1 de Glucose-Sódio/imunologia
4.
Genes Immun ; 21(4): 224-239, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32753697

RESUMO

Carbohydrate-specific antibodies are widespread among all classes of immunoglobulins. Despite their broad occurrence, little is known about their formation and biological significance. Carbohydrate-specific antibodies are often classified as natural antibodies under the assumption that they arise without prior exposure to exogenous antigens. On the other hand, various carbohydrate-specific antibodies, including antibodies to ABO blood group antigens, emerge after the contact of immune cells with the intestinal microbiota, which expresses a vast diversity of carbohydrate antigens. Here we explore the development of carbohydrate-specific antibodies in humans, addressing the definition of natural antibodies and the production of carbohydrate-specific antibodies upon antigen stimulation. We focus on the significance of the intestinal microbiota in shaping carbohydrate-specific antibodies not just in the gut, but also in the blood circulation. The structural similarity between bacterial carbohydrate antigens and surface glycoconjugates of protists, fungi and animals leads to the production of carbohydrate-specific antibodies protective against a broad range of pathogens. Mimicry between bacterial and human glycoconjugates, however, can also lead to the generation of carbohydrate-specific antibodies that cross-react with human antigens, thereby contributing to the development of autoimmune disorders.


Assuntos
Anticorpos/imunologia , Carboidratos/química , Carboidratos/imunologia , Microbioma Gastrointestinal/imunologia , Animais , Apresentação de Antígeno , Autoimunidade , Glicoconjugados/imunologia , Humanos
5.
Trends Biochem Sci ; 40(7): 377-84, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25840516

RESUMO

Glycosylation is a ubiquitous modification of lipids and proteins. Despite the essential contribution of glycoconjugates to the viability of all living organisms, diseases of glycosylation in humans have only been identified over the past few decades. The recent development of next-generation DNA sequencing techniques has accelerated the pace of discovery of novel glycosylation defects. The description of multiple mutations across glycosylation pathways not only revealed tremendous diversity in functional impairments, but also pointed to phenotypic similarities, emphasizing the interconnected flow of substrates underlying glycan assembly. The current list of 100 known glycosylation disorders provides an overview of the significance of glycosylation in human development and physiology.


Assuntos
Defeitos Congênitos da Glicosilação/metabolismo , Glicocálix/metabolismo , Processamento de Proteína Pós-Traducional , Animais , Defeitos Congênitos da Glicosilação/patologia , Retículo Endoplasmático/metabolismo , Glicoproteínas/metabolismo , Glicosilação , Glicosiltransferases/fisiologia , Complexo de Golgi/metabolismo , Humanos
6.
Ann Neurol ; 84(6): 843-853, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30412317

RESUMO

OBJECTIVE: Approximately 5% of cerebral small vessel diseases are hereditary, which include COL4A1/COL4A2-related disorders. COL4A1/COL4A2 encode type IV collagen α1/2 chains in the basement membranes of cerebral vessels. COL4A1/COL4A2 mutations impair the secretion of collagen to the extracellular matrix, thereby resulting in vessel fragility. The diagnostic yield for COL4A1/COL4A2 variants is around 20 to 30%, suggesting other mutated genes might be associated with this disease. This study aimed to identify novel genes that cause COL4A1/COL4A2-related disorders. METHODS: Whole exome sequencing was performed in 2 families with suspected COL4A1/COL4A2-related disorders. We validated the role of COLGALT1 variants by constructing a 3-dimensional structural model, evaluating collagen ß (1-O) galactosyltransferase 1 (ColGalT1) protein expression and ColGalT activity by Western blotting and collagen galactosyltransferase assays, and performing in vitro RNA interference and rescue experiments. RESULTS: Exome sequencing demonstrated biallelic variants in COLGALT1 encoding ColGalT1, which was involved in the post-translational modification of type IV collagen in 2 unrelated patients: c.452 T > G (p.Leu151Arg) and c.1096delG (p.Glu366Argfs*15) in Patient 1, and c.460G > C (p.Ala154Pro) and c.1129G > C (p.Gly377Arg) in Patient 2. Three-dimensional model analysis suggested that p.Leu151Arg and p.Ala154Pro destabilized protein folding, which impaired enzymatic activity. ColGalT1 protein expression and ColGalT activity in Patient 1 were undetectable. RNA interference studies demonstrated that reduced ColGalT1 altered COL4A1 secretion, and rescue experiments showed that mutant COLGALT1 insufficiently restored COL4A1 production in cells compared with wild type. INTERPRETATION: Biallelic COLGALT1 variants cause cerebral small vessel abnormalities through a common molecular pathogenesis with COL4A1/COL4A2-related disorders. Ann Neurol 2018;84:843-853.


Assuntos
Doenças de Pequenos Vasos Cerebrais/genética , Colágeno Tipo IV/genética , Predisposição Genética para Doença/genética , Mutação/genética , Linhagem Celular Transformada , Doenças de Pequenos Vasos Cerebrais/diagnóstico por imagem , Criança , Análise Mutacional de DNA , Glucosiltransferases/metabolismo , Humanos , Imageamento por Ressonância Magnética , Masculino , Modelos Moleculares , Mutagênese , RNA Mensageiro/metabolismo , Transfecção
7.
Cell Mol Life Sci ; 74(16): 2959-2977, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28352996

RESUMO

The composition of the gut microbiota is in constant flow under the influence of factors such as the diet, ingested drugs, the intestinal mucosa, the immune system, and the microbiota itself. Natural variations in the gut microbiota can deteriorate to a state of dysbiosis when stress conditions rapidly decrease microbial diversity and promote the expansion of specific bacterial taxa. The mechanisms underlying intestinal dysbiosis often remain unclear given that combinations of natural variations and stress factors mediate cascades of destabilizing events. Oxidative stress, bacteriophages induction and the secretion of bacterial toxins can trigger rapid shifts among intestinal microbial groups thereby yielding dysbiosis. A multitude of diseases including inflammatory bowel diseases but also metabolic disorders such as obesity and diabetes type II are associated with intestinal dysbiosis. The characterization of the changes leading to intestinal dysbiosis and the identification of the microbial taxa contributing to pathological effects are essential prerequisites to better understand the impact of the microbiota on health and disease.


Assuntos
Disbiose/etiologia , Microbioma Gastrointestinal , Intestinos/microbiologia , Animais , Dieta , Disbiose/imunologia , Disbiose/metabolismo , Disbiose/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Humanos , Imunidade , Inflamassomos/imunologia , Mucosa Intestinal/metabolismo , Intestinos/patologia , Estresse Oxidativo , Preparações Farmacêuticas/metabolismo
8.
J Biol Chem ; 291(35): 18514-24, 2016 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-27402836

RESUMO

Collagen is post-translationally modified by prolyl and lysyl hydroxylation and subsequently by glycosylation of hydroxylysine. Despite the widespread occurrence of the glycan structure Glc(α1-2)Gal linked to hydroxylysine in animals, the functional significance of collagen glycosylation remains elusive. To address the role of glycosylation in collagen expression, folding, and secretion, we used the CRISPR/Cas9 system to inactivate the collagen galactosyltransferase GLT25D1 and GLT25D2 genes in osteosarcoma cells. Loss of GLT25D1 led to increased expression and intracellular accumulation of collagen type I, whereas loss of GLT25D2 had no effect on collagen secretion. Inactivation of the GLT25D1 gene resulted in a compensatory induction of GLT25D2 expression. Loss of GLT25D1 decreased collagen glycosylation by up to 60% but did not alter collagen folding and thermal stability. Whereas cells harboring individually inactivated GLT25D1 and GLT25D2 genes could be recovered and maintained in culture, cell clones with simultaneously inactive GLT25D1 and GLT25D2 genes could be not grown and studied, suggesting that a complete loss of collagen glycosylation impairs osteosarcoma cell proliferation and viability.


Assuntos
Neoplasias Ósseas/metabolismo , Colágeno/biossíntese , Galactosiltransferases , Proteínas de Neoplasias/metabolismo , Osteossarcoma/metabolismo , Neoplasias Ósseas/genética , Linhagem Celular Tumoral , Colágeno/genética , Galactosiltransferases/biossíntese , Galactosiltransferases/genética , Deleção de Genes , Regulação Enzimológica da Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Proteínas de Neoplasias/genética , Osteossarcoma/genética , Dobramento de Proteína
9.
Biochem J ; 473(20): 3451-3462, 2016 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-27433018

RESUMO

Acanthamoeba polyphaga mimivirus is a giant virus encoding 1262 genes among which many were previously thought to be exclusive to cellular life. For example, mimivirus genes encode enzymes involved in the biosynthesis of nucleotide sugars and putative glycosyltransferases. We identified in mimivirus a glycogenin-1 homologous gene encoded by the open reading frame R707. The R707 protein was found to be active as a polymerizing glucosyltransferase enzyme. Like glycogenin-1, R707 activity was divalent-metal-ion-dependent and relied on an intact DXD motif. In contrast with glycogenin-1, R707 was, however, not self-glucosylating. Interestingly, the product of R707 catalysis featured α1-6, ß1-6 and α1-4 glycosidic linkages. Mimivirus R707 is the first reported glycosyltransferase able to catalyse the formation of both α and ß linkages. Mimivirus-encoded glycans play a role in the infection of host amoebae. Co-infection of Acanthamoeba with mimivirus and amylose and chitin hydrolysate reduced the number of infected amoebae, thus supporting the importance of polysaccharide chains in the uptake of mimivirus by amoebae. The identification of a glycosyltransferase capable of forming α and ß linkages underlines the peculiarity of mimivirus and enforces the concept of a host-independent glycosylation machinery in mimivirus.


Assuntos
Acanthamoeba/virologia , Glucose/metabolismo , Glucosiltransferases/metabolismo , Glicoproteínas/metabolismo , Mimiviridae/metabolismo , Mimiviridae/patogenicidade , Proteínas Virais/metabolismo , Glucose/química , Glucosiltransferases/química , Glicoproteínas/química , Glicosídeos/química , Glicosídeos/metabolismo , Glicosilação , Glicosiltransferases/metabolismo , Proteínas Virais/química
10.
Br J Nutr ; 116(8): 1356-1368, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27719686

RESUMO

The gut microbiota has been established as an important player influencing many aspects of human physiology. Breast milk, the first diet for an infant, contains human milk oligosaccharides (HMO) that shape the infant's gut microbiota by selectively stimulating the growth of specific bacteria, especially bifidobacteria. In addition to their bifidogenic activity, the ability of HMO to modulate immune function and the gut barrier makes them prime candidates to restore a beneficial microbiota in dysbiotic adults and provide health benefits. We conducted a parallel, double-blind, randomised, placebo-controlled, HMO-supplementation study in 100 healthy, adult volunteers, consuming chemically produced 2'-O-fucosyllactose (2'FL) and/or lacto-N-neotetraose (LNnT) at various daily doses and mixes or placebo for 2 weeks. All participants completed the study without premature discontinuation. Supplementation of 2'FL and LNnT at daily doses up to 20 g was shown to be safe and well tolerated, as assessed using the gastrointestinal symptoms rating scale. 16S rRNA sequencing analysis showed that HMO supplementation specifically modified the adult gut microbiota with the primary impact being substantial increases in relative abundance of Actinobacteria and Bifidobacterium in particular and a reduction in relative abundance of Firmicutes and Proteobacteria. This study provides the first set of data on safety, tolerance and impact of HMO on the adult gut microbiota. Collectively, the results from this study show that supplementing the diet with HMO is a valuable strategy to shape the human gut microbiota and specifically promote the growth of beneficial bifidobacteria.


Assuntos
Actinobacteria/crescimento & desenvolvimento , Bifidobacterium/crescimento & desenvolvimento , Disbiose/prevenção & controle , Microbioma Gastrointestinal , Oligossacarídeos/uso terapêutico , Prebióticos , Trissacarídeos/uso terapêutico , Actinobacteria/classificação , Actinobacteria/isolamento & purificação , Adulto , Bifidobacterium/classificação , Bifidobacterium/isolamento & purificação , Biomarcadores/análise , Biomarcadores/sangue , Dinamarca , Método Duplo-Cego , Disbiose/sangue , Disbiose/metabolismo , Disbiose/microbiologia , Fezes/química , Fezes/microbiologia , Feminino , Firmicutes/classificação , Firmicutes/crescimento & desenvolvimento , Firmicutes/isolamento & purificação , Humanos , Masculino , Pessoa de Meia-Idade , Tipagem Molecular , Oligossacarídeos/administração & dosagem , Oligossacarídeos/efeitos adversos , Prebióticos/administração & dosagem , Prebióticos/efeitos adversos , Análise de Componente Principal , Proteobactérias/classificação , Proteobactérias/crescimento & desenvolvimento , Proteobactérias/isolamento & purificação , Trissacarídeos/administração & dosagem , Trissacarídeos/efeitos adversos , Adulto Jovem
11.
Proc Natl Acad Sci U S A ; 110(43): 17444-9, 2013 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-24101501

RESUMO

Breast milk oligosaccharides shape the intestinal environment by affecting mucosal immunity and bacterial colonization. To clarify the role of milk oligosaccharide sialyl(α2,3)lactose (3SL) in intestinal physiology and disease, we investigated colitis development in Il10(-/-) mice exposed to normal or 3SL-deficient milk during lactation. Onset and progression of intestinal inflammation were delayed in Il10(-/-) mice deficient for the α2,3 sialyltransferase 4 (ST3GAL4) responsible for 3SL biosynthesis. The proinflammatory role of 3SL was confirmed by showing that oral supplementation of newborn Il10(-/-);St3gal4(-/-) mice with 3SL increased colitis severity. Conversely, fostering of newborn Il10(-/-) mice to lactating St3gal4(-/-) mothers reduced colitis severity. 3SL directly stimulated mesenteric lymph node CD11c(+) dendritic cells and induced production of cytokines required for expansion of TH1 and TH17 T cells. The stimulatory effect of 3SL was attenuated in Tlr4-deficient CD11c(+) cells, demonstrating that 3SL induces inflammation through Toll-like receptor 4 (TLR4) signaling. Thus, 3SL directly modulates mucosal immunity, which increases susceptibility to colitis.


Assuntos
Antígeno CD11c/imunologia , Intestinos/imunologia , Lactose/análogos & derivados , Ácidos Siálicos/imunologia , Receptor 4 Toll-Like/imunologia , Animais , Animais Recém-Nascidos , Bactérias/classificação , Bactérias/genética , Bactérias/imunologia , Antígeno CD11c/metabolismo , Células Cultivadas , Colite/genética , Colite/imunologia , Colite/metabolismo , Citocinas/genética , Citocinas/imunologia , Citocinas/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Feminino , Citometria de Fluxo , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/patologia , Expressão Gênica/imunologia , Imunidade nas Mucosas/genética , Imunidade nas Mucosas/imunologia , Interleucina-10/deficiência , Interleucina-10/genética , Interleucina-10/imunologia , Mucosa Intestinal/metabolismo , Intestinos/citologia , Lactação/imunologia , Lactose/imunologia , Lactose/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Leite/química , Leite/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ácidos Siálicos/metabolismo , Sialiltransferases/deficiência , Sialiltransferases/genética , Sialiltransferases/imunologia , Receptor 4 Toll-Like/metabolismo , beta-Galactosídeo alfa-2,3-Sialiltransferase
12.
J Virol ; 88(2): 838-45, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24173233

RESUMO

Collagens, the most abundant proteins in animals, also occur in some recently described nucleocytoplasmic large DNA viruses such as Mimiviridae, which replicate in amoebae. To clarify the impact of viral collagens on the immune response of animals exposed to Mimiviridae, we have investigated the localization of collagens in Acanthamoeba polyphaga mimivirus particles and the response of mice to immunization with mimivirus particles. Using protein biotinylation, we have first shown that viral collagen encoded by open reading frame L71 is present at the surface of mimivirus particles. Exposure to mimivirus collagens elicited the production of anti-collagen antibodies in DBA/1 mice immunized intradermally with mimivirus protein extracts. This antibody response also targeted mouse collagen type II and was accompanied by T-cell reactivity to collagen and joint inflammation, as observed in collagen-induced arthritis following immunization of mice with bovine collagen type II. The broad distribution of nucleocytoplasmic large DNA viruses in the environment suggests that humans are constantly exposed to such large virus particles. A survey of blood sera from healthy human subjects and from rheumatoid arthritis patients indeed demonstrated that 30% of healthy-subject and 36% of rheumatoid arthritis sera recognized the major mimivirus capsid protein L425. Moreover, whereas 6% of healthy-subject sera recognized the mimivirus collagen protein L71, 22% of rheumatoid arthritis sera were positive for mimivirus L71. Accordingly, our study shows that environmental exposure to mimivirus represents a risk factor in triggering autoimmunity to collagens.


Assuntos
Artrite/virologia , Colágeno/imunologia , Mimiviridae/imunologia , Proteínas Virais/imunologia , Animais , Anticorpos Antivirais/imunologia , Artrite/imunologia , Bovinos , Colágeno/genética , Modelos Animais de Doenças , Humanos , Imunização , Camundongos , Camundongos Endogâmicos DBA , Mimiviridae/genética , Proteínas Virais/genética
13.
Glycobiology ; 24(8): 703-14, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24794008

RESUMO

Acanthamoeba polyphaga mimivirus is a member of the giant nucleocytoplasmic large DNA viruses, infecting various Acanthamoeba spp. The genomes of giant viruses encode components previously thought to be exclusive to cellular life, such as proteins involved in nucleic acid and protein synthesis. Recent work on enzymes involved in carbohydrate biosynthesis and metabolism show that instead of utilizing host cell resources, Mimivirus produces its own glycosylation machinery. To obtain a more detailed view of glycosylation in Mimivirus, we developed a periodate oxidation-based method to selectively enrich Mimivirus surface glycoproteins. O-Glycosylation in Mimivirus glycoproteins was identified by permethylation and matrix-assisted laser desorption/ionization-mass spectrometry analyses of beta-eliminated glycans. We sequenced 26 previously undescribed O-glycans, most of which contain glucose as their reducing end saccharide. These data will facilitate future studies on the functional significance of glycosylation in Mimivirus.


Assuntos
Glicoproteínas/metabolismo , Mimiviridae/metabolismo , Proteínas Virais/metabolismo , Glicoproteínas/biossíntese , Glicoproteínas/química , Glicosilação , Oxirredução , Proteínas Virais/biossíntese , Proteínas Virais/química
14.
Br J Nutr ; 111(9): 1602-10, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24411010

RESUMO

The oligosaccharides 2-fucosyllactose and 3-fucosyllactose are major constituents of human breast milk but are not found in mouse milk. Milk oligosaccharides have a prebiotic action, thus affecting the colonisation of the infant intestine by microbiota. To determine the specific effect of fucosyllactose exposure on intestinal microbiota in mice, in the present study, we orally supplemented newborn mice with pure 2-fucosyllactose and 3-fucosyllactose. Exposure to 2-fucosyllactose and 3-fucosyllactose increased the levels of bacteria of the Porphyromonadaceae family in the intestinal gut, more precisely members of the genus Barnesiella as analysed by 16S pyrosequencing. The ability of Barnesiella to utilise fucosyllactose as energy source was confirmed in bacterial cultures. Whereas B. intestinihominis and B. viscericola did not grow on fucose alone, they proliferated in the presence of 2-fucosyllactose and 3-fucosyllactose following the secretion of linkage-specific fucosidase enzymes that liberated lactose. The change in the composition of intestinal microbiota mediated by fucosyllactose supplementation affected the susceptibility of mice to dextran sulphate sodium-induced colitis, as indicated by increased resistance of mice subjected to 2-fucosyllactose supplementation for 6 weeks. The present study underlines the ability of specific milk oligosaccharides to change the composition of intestinal microbiota and thereby to shape an intestinal milieu resilient to inflammatory diseases.


Assuntos
Bacteroidetes/metabolismo , Colite/prevenção & controle , Mucosa Intestinal/microbiologia , Intestinos/microbiologia , Oligossacarídeos/uso terapêutico , Prebióticos , Trissacarídeos/uso terapêutico , Animais , Animais Recém-Nascidos , Proteínas de Bactérias/metabolismo , Bacteroidetes/classificação , Bacteroidetes/crescimento & desenvolvimento , Bacteroidetes/isolamento & purificação , Colite/imunologia , Colite/patologia , Modelos Animais de Doenças , Resistência à Doença , Fezes/química , Fezes/microbiologia , Feminino , Mucosa Intestinal/crescimento & desenvolvimento , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Intestinos/crescimento & desenvolvimento , Intestinos/imunologia , Intestinos/patologia , Lactose/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Leite Humano/química , Tipagem Molecular , Oligossacarídeos/administração & dosagem , Oligossacarídeos/efeitos adversos , Oligossacarídeos/metabolismo , Prebióticos/efeitos adversos , Caracteres Sexuais , Trissacarídeos/administração & dosagem , Trissacarídeos/efeitos adversos , Trissacarídeos/metabolismo , alfa-L-Fucosidase/metabolismo
15.
Appl Microbiol Biotechnol ; 98(10): 4445-55, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24362857

RESUMO

Collagen is the most abundant protein in the human body and thereby a structural protein of considerable biotechnological interest. The complex maturation process of collagen, including essential post-translational modifications such as prolyl and lysyl hydroxylation, has precluded large-scale production of recombinant collagen featuring the biophysical properties of endogenous collagen. The characterization of new prolyl and lysyl hydroxylase genes encoded by the giant virus mimivirus reveals a method for production of hydroxylated collagen. The coexpression of a human collagen type III construct together with mimivirus prolyl and lysyl hydroxylases in Escherichia coli yielded up to 90 mg of hydroxylated collagen per liter culture. The respective levels of prolyl and lysyl hydroxylation reaching 25 % and 26 % were similar to the hydroxylation levels of native human collagen type III. The distribution of hydroxyproline and hydroxylysine along recombinant collagen was also similar to that of native collagen as determined by mass spectrometric analysis of tryptic peptides. The triple helix signature of recombinant hydroxylated collagen was confirmed by circular dichroism, which also showed that hydroxylation increased the thermal stability of the recombinant collagen construct. Recombinant hydroxylated collagen produced in E. coli supported the growth of human umbilical endothelial cells, underlining the biocompatibility of the recombinant protein as extracellular matrix. The high yield of recombinant protein expression and the extensive level of prolyl and lysyl hydroxylation achieved indicate that recombinant hydroxylated collagen can be produced at large scale for biomaterials engineering in the context of biomedical applications.


Assuntos
Colágeno Tipo III/metabolismo , Escherichia coli/metabolismo , Engenharia Metabólica/métodos , Processamento de Proteína Pós-Traducional , Colágeno Tipo III/genética , Estabilidade Enzimática , Escherichia coli/genética , Humanos , Hidroxilação , Espectrometria de Massas , Mimiviridae/enzimologia , Mimiviridae/genética , Dados de Sequência Molecular , Pró-Colágeno-Lisina 2-Oxoglutarato 5-Dioxigenase/genética , Pró-Colágeno-Lisina 2-Oxoglutarato 5-Dioxigenase/metabolismo , Prolil Hidroxilases/genética , Prolil Hidroxilases/metabolismo , Conformação Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Análise de Sequência de DNA , Temperatura
16.
Biochim Biophys Acta ; 1820(9): 1306-17, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22343051

RESUMO

BACKGROUND: Diseases of glycosylation are rare inherited disorders, which are often referred to as congenital disorders of glycosylation (CDG). Several types of CDG have been described in the last decades, encompassing defects of nucleotide-sugar biosynthesis, nucleotide-sugar transporters, glycosyltransferases and vesicular transport. Although clinically heterogeneous, most types of CDG are associated with neurological impairments ranging from severe psychomotor retardation to moderate intellectual disabilities. CDG are mainly caused by defects of N-glycosylation, owing to the simple detection of under-glycosylated serum transferrin by isoelectric focusing. SCOPE OF REVIEW: In the last years, several disorders of O-glycosylation, glycolipid and glycosaminoglycan biosynthesis have been described, which are known by trivial names not directly associated with the family of CDG. The present review outlines 64 gene defects affecting glycan biosynthesis and modifications, thereby underlining the complexity of glycosylation pathways and pointing to unexpected phenotypes and functional redundancies in the control of glycoconjugate biosynthesis. MAJOR CONCLUSIONS: The increasing application of whole-genome sequencing techniques unravels new defects of glycosylation, which are associated to moderate forms of mental disabilities. GENERAL SIGNIFICANCE: The knowledge gathered through the investigation of CDG increases the understanding of the functions associated to protein glycosylation in humans. This article is part of a Special Issue entitled Glycoproteomics.


Assuntos
Erros Inatos do Metabolismo dos Carboidratos/classificação , Erros Inatos do Metabolismo dos Carboidratos/genética , Defeitos Congênitos da Glicosilação/genética , Animais , Erros Inatos do Metabolismo dos Carboidratos/epidemiologia , Sequência de Carboidratos , Defeitos Congênitos da Glicosilação/classificação , Defeitos Congênitos da Glicosilação/epidemiologia , Glicosilação , Humanos , Modelos Biológicos , Dados de Sequência Molecular
17.
Am J Clin Nutr ; 117(1): 64-72, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36789945

RESUMO

BACKGROUND: Whether prebiotic human milk oligosaccharides (HMO), such as 2'-fucosyllactose (2'-FL) and lacto-N-neotetraose (LNnT), enhance iron absorption in infants is unknown. Moreover, whether maternal HMO profile affects absorption of iron fortificants or the effects of prebiotic galacto-oligosaccharides (GOS) and/or HMO on iron absorption is uncertain. OBJECTIVES: The aim of this study was to test whether consumption of 3.0 g GOS or HMO enhances iron absorption from iron-fortified maize porridge in partially breastfed Kenyan infants and whether maternal HMO profile modulates these effects. METHODS: In a randomized, prospective crossover study, 55 infants (aged 8-12 mo) were fed test meals fortified with 1 of the following: 1) 5.0 mg iron as 54Fe-labeled ferrous fumarate (FeFum); 2) 5.0 mg iron as 58FeFum and 3.0 g GOS (FeFum+GOS); and 3) 5.0 mg iron as 57FeFum and 2.0 g 2'-FL and 1.0 g LNnT (FeFum+HMO). Fractional iron absorption (FIA) was assessed by erythrocyte incorporation of iron isotopes. HMO profiles were determined by capillary gel electrophoresis with laser-induced florescence detection. Data were analyzed with mixed-effect models, and iron dialyzability was measured in vitro. RESULTS: Of the 55 infants included, 49 were fed as instructed. FIA from the FeFum+GOS group [median (IQR) 22.2% (16.5%-25.9%)] was higher than that from the FeFum group [12.5% (9.5%-20.9%)] (P = 0.005). FIA from the FeFum+HMO group was 13.3% (7.1%-24.4%) and did not differ from the FeFum group (P = 0.923). Maternal HMO profile did not predict FIA or modulate the effects of GOS or HMO on FIA. Iron dialyzability ratios at pH 2 of FeFum+GOS to FeFum and FeFum+HMO to FeFum were 2.1 and 0.9 (P = 0.001 and P = 0.322), respectively. CONCLUSIONS: In Kenyan infants consuming FeFum-fortified maize porridge, co-provision of 3.0 g GOS increased FIA by 78%, whereas co-provision of 3.0 g HMO did not affect FIA. Variations in maternal HMO profile, including secretor and Lewis phenotype, did not predict FIA. These data argue against a physiologic role for 2'-FL and LNnT in facilitating iron absorption in infancy. The study was registered at clinicaltrials.gov as NCT04163406 (https://clinicaltrials.gov/ct2/show/NCT04163406).


Assuntos
Ferro , Leite Humano , Feminino , Humanos , Lactente , Leite Humano/metabolismo , Quênia , Estudos Cross-Over , Estudos Prospectivos , Oligossacarídeos/farmacologia , Oligossacarídeos/metabolismo , Prebióticos
18.
J Biol Chem ; 286(51): 43701-43709, 2011 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-22045808

RESUMO

Collagens, the most abundant proteins in animals, are modified by hydroxylation of proline and lysine residues and by glycosylation of hydroxylysine. Dedicated prolyl hydroxylase, lysyl hydroxylase, and collagen glycosyltransferase enzymes localized in the endoplasmic reticulum mediate these modifications prior to the formation of the collagen triple helix. Whereas collagen-like proteins have been described in some fungi, bacteria, and viruses, the post-translational machinery modifying collagens has never been described outside of animals. We demonstrate that the L230 open reading frame of the giant virus Acanthamoeba polyphaga mimivirus encodes an enzyme that has distinct lysyl hydroxylase and collagen glycosyltransferase domains. We show that mimivirus L230 is capable of hydroxylating lysine and glycosylating the resulting hydroxylysine residues in a native mimivirus collagen acceptor substrate. Whereas in animals from sponges to humans the transfer of galactose to hydroxylysine in collagen is conserved, the mimivirus L230 enzyme transfers glucose to hydroxylysine, thereby defining a novel type of collagen glycosylation in nature. The presence of hydroxylysine in mimivirus proteins was confirmed by amino acid analysis of mimivirus recovered from A. polyphaga cultures. This work shows for the first time that collagen post-translational modifications are not confined to the domains of life. The utilization of glucose instead of the galactose found throughout animals as well as a bifunctional enzyme rather than two separate enzymes may represent a parallel evolutionary track in collagen biology. These results suggest that giant viruses may have contributed to the evolution of collagen biology.


Assuntos
Colágeno/metabolismo , Glicosiltransferases/química , Mimiviridae/metabolismo , Pró-Colágeno-Lisina 2-Oxoglutarato 5-Dioxigenase/química , Acanthamoeba/metabolismo , Sequência de Aminoácidos , Animais , Baculoviridae/metabolismo , Clonagem Molecular , Colágeno/química , Vetores Genéticos , Glicosilação , Humanos , Hidroxilisina/química , Dados de Sequência Molecular , Fases de Leitura Aberta , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos
19.
J Biol Chem ; 286(8): 6085-91, 2011 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-21183681

RESUMO

The majority of congenital disorders of glycosylation (CDG) are caused by defects of dolichol (Dol)-linked oligosaccharide assembly, which lead to under-occupancy of N-glycosylation sites. Most mutations encountered in CDG are hypomorphic, thus leaving residual activity to the affected biosynthetic enzymes. We hypothesized that increased cellular levels of Dol-linked substrates might compensate for the low biosynthetic activity and thereby improve the output of protein N-glycosylation in CDG. To this end, we investigated the potential of the squalene synthase inhibitor zaragozic acid A to redirect the flow of the polyisoprene pathway toward Dol by lowering cholesterol biosynthesis. The addition of zaragozic acid A to CDG fibroblasts with a Dol-P-Man synthase defect led to the formation of longer Dol-P species and to increased Dol-P-Man levels. This treatment was shown to decrease the pathologic accumulation of incomplete Dol pyrophosphate-GlcNAc(2)Man(5) in Dol-P-Man synthase-deficient fibroblasts. Zaragozic acid A treatment also decreased the amount of truncated protein N-linked oligosaccharides in these CDG fibroblasts. The increased cellular levels of Dol-P-Man and possibly the decreased cholesterol levels in zaragozic acid A-treated cells also led to increased availability of the glycosylphosphatidylinositol anchor as shown by the elevated cell-surface expression of the CD59 protein. This study shows that manipulation of the cellular Dol pool, as achieved by zaragozic acid A addition, may represent a valuable approach to improve N-linked glycosylation in CDG cells.


Assuntos
Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Defeitos Congênitos da Glicosilação/metabolismo , Dolicóis/metabolismo , Inibidores Enzimáticos/farmacologia , Farnesil-Difosfato Farnesiltransferase/antagonistas & inibidores , Oligossacarídeos/biossíntese , Ácidos Tricarboxílicos/farmacologia , Antígenos CD59/biossíntese , Antígenos CD59/genética , Células Cultivadas , Colesterol/biossíntese , Colesterol/genética , Defeitos Congênitos da Glicosilação/genética , Dolicóis/genética , Farnesil-Difosfato Farnesiltransferase/genética , Farnesil-Difosfato Farnesiltransferase/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Glicosilação/efeitos dos fármacos , Humanos , Manosiltransferases/genética , Manosiltransferases/metabolismo , Oligossacarídeos/genética
20.
Front Immunol ; 13: 797759, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35222379

RESUMO

Altered surface glycosylation is a major hallmark of tumor cells associated with aggressive phenotype and poor prognosis. By recognizing specific carbohydrate motifs, lectins can be applied to distinguish tumor from healthy cells based on the expression of glycosylation-dependent markers. Through their ability to bind to specific carbohydrates, lectins induce cell agglutination and cross-link surface glycoproteins, thereby mediating mitogenic and death-inducing effects in various cell types. The carbohydrate-selective cytotoxic effect of lectins also enables their possible application in therapies targeting cancer cells. To clarify the intracellular pathways mediating cell death induced by a group of plant and fungal lectins, we investigated mouse adenocarcinoma MC-38 cells harboring inactive genes involved in apoptosis, necroptosis and pyroptosis. Treatment of MC-38 cells with wheat germ agglutinin, Maackia amurensis lectin I, and Aleuria aurantia lectin induced multiple cell death pathways through reactions that relied on the autophagy machinery without depending on caspase activation. Furthermore, inhibition of de novo protein synthesis by cycloheximide strongly decreased the cytotoxic response, indicating that the lectins investigated induced cell death via effector molecules that are not expressed under normal circumstances and supporting the non-apoptotic nature of cell death. The broad cytotoxic response to lectins can be beneficial for the development of combination therapies targeting tumor cells. Given that tumors acquire resistance to various cytotoxic treatments because of mutations in cell death pathways, compounds inducing broad cytotoxic responses, such as lectins, represent potent sensitizers to promote tumor cell killing.


Assuntos
Adenocarcinoma , Antineoplásicos , Animais , Antineoplásicos/farmacologia , Apoptose , Carboidratos , Glicosilação , Lectinas/metabolismo , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA