Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
1.
Reprod Biol Endocrinol ; 19(1): 14, 2021 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-33494759

RESUMO

In recent years, the discovery of ovarian germ stem cells (OGSCs) has provided a new research direction for the treatment of female infertility. The ovarian microenvironment affects the proliferation and differentiation of OGSCs, and immune cells and related cytokines are important components of the microenvironment. However, whether improving the ovarian microenvironment can regulate the proliferation of OGSCs and remodel ovarian function has not been reported. In this study, we chelated chito-oligosaccharide (COS) with fluorescein isothiocyanate (FITC) to track the distribution of COS in the body. COS was given to mice through the best route of administration, and the changes in ovarian and immune function were detected using assays of organ index, follicle counting, serum estrogen (E2) and anti-Mullerian hormone (AMH) levels, and the expression of IL-2 and TNF-α in the ovaries. We found that COS significantly increased the organ index of the ovary and immune organs, reduced the rate of follicular atresia, increased the levels of E2 and AMH hormones, and increased the protein expression of IL-2 and TNF-α in the ovary. Then, COS and OGSCs were co-cultured to observe the combination of COS and OGSCs, and measure the survival rate of OGSCs. With increasing time, the fluorescence intensity of cells gradually increased, and the cytokines IL-2 and TNF-α significantly promoted the proliferation of OGSCs. In conclusion, COS could significantly improve the ovarian and immune function of chemotherapy model mice, and improve the survival rate of OGSCs, which provided a preliminary blueprint for further exploring the mechanism of COS in protecting ovarian function.


Assuntos
Oligossacarídeos/fisiologia , Ovário/fisiologia , Insuficiência Ovariana Primária/patologia , Células-Tronco/fisiologia , Animais , Antineoplásicos/efeitos adversos , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Quitosana/farmacocinética , Modelos Animais de Doenças , Feminino , Células Germinativas/metabolismo , Células Germinativas/patologia , Células Germinativas/fisiologia , Camundongos , Oligossacarídeos/farmacocinética , Folículo Ovariano/metabolismo , Folículo Ovariano/fisiologia , Ovário/metabolismo , Ovário/patologia , Insuficiência Ovariana Primária/induzido quimicamente , Recuperação de Função Fisiológica , Células-Tronco/metabolismo , Células-Tronco/patologia
2.
J Immunol ; 204(12): 3283-3295, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32434942

RESUMO

Neisseria gonorrhoeae deploys a unique immune evasion strategy wherein the lacto-N-neotetraose termini of lipooligosaccharide (LOS) are "capped" by a surface LOS sialyltransferase (Lst), using extracellular host-derived CMP-sialic acid (CMP-Neu5Ac in humans). LOS sialylation enhances complement resistance by recruiting factor H (FH; alternative complement pathway inhibitor) and also by limiting classical pathway activation. Sialylated LOS also engages inhibitory Siglecs on host leukocytes, dampening innate immunity. Previously, we showed that analogues of CMP-sialic acids (CMP-nonulosonates [CMP-NulOs]), such as CMP-Leg5,7Ac2 and CMP-Neu5Ac9N3, are also substrates for Lst. Incorporation of Leg5,7Ac2 and Neu5Ac9N3 into LOS results in N. gonorrhoeae being fully serum sensitive. Importantly, intravaginal administration of CMP-Leg5,7Ac2 attenuated N. gonorrhoeae colonization of mouse vaginas. In this study, we characterize and develop additional candidate therapeutic CMP-NulOs. CMP-ketodeoxynonulosonate (CMP-Kdn) and CMP-Kdn7N3, but not CMP-Neu4,5Ac2, were substrates for Lst, further elucidating gonococcal Lst specificity. Lacto-N-neotetraose LOS capped with Kdn and Kdn7N3 bound FH to levels ∼60% of that seen with Neu5Ac and enabled gonococci to resist low (3.3%) but not higher (10%) concentrations of human complement. CMP-Kdn, CMP-Neu5Ac9N3, and CMP-Leg5,7Ac2 administered intravaginally (10 µg/d) to N. gonorrhoeae-colonized mice were equally efficacious. Of the three CMP-NulOs above, CMP-Leg5,7Ac2 was the most pH and temperature stable. In addition, Leg5,7Ac2-fed human cells did not display this NulO on their surface. Moreover, CMP-Leg5,7Ac2 was efficacious against several multidrug-resistant gonococci in mice with a humanized sialome (Cmah-/- mice) or humanized complement system (FH/C4b-binding protein transgenic mice). CMP-Leg5,7Ac2 and CMP-Kdn remain viable leads as topical preventive/therapeutic agents against the global threat of multidrug-resistant N. gonorrhoeae.


Assuntos
Ácido N-Acetilneuramínico do Monofosfato de Citidina/farmacologia , Monofosfato de Citidina/análogos & derivados , Monofosfato de Citidina/fisiologia , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Gonorreia/tratamento farmacológico , Neisseria gonorrhoeae/efeitos dos fármacos , Ácidos Neuramínicos/farmacologia , Ácidos Siálicos/farmacologia , Animais , Linhagem Celular Tumoral , Fator H do Complemento/metabolismo , Proteínas do Sistema Complemento/farmacologia , Monofosfato de Citidina/farmacologia , Feminino , Gonorreia/metabolismo , Gonorreia/microbiologia , Humanos , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Oligossacarídeos/fisiologia , Sialiltransferases/farmacologia
3.
Rev. chil. nutr ; 46(5): 633-643, oct. 2019.
Artigo em Espanhol | LILACS | ID: biblio-1042705

RESUMO

Durante los primeros meses de vida, los oligosacáridos de la leche materna (HMOs) aportados por la leche materna participan en procesos asociados con la maduración de tejidos y sistemas del tubo digestivo, modulan algunos de sus procesos metabólicos y ejercen efectos prebióticos y antimicrobianos. Otros efectos estudiados son su contribución a la instalación, desarrollo y estimulación de la microbiota residente con predomino de Bifidobacterium y Bacteroides, con efectos protectores frente a posibles colonizaciones o patologías por enteropatógenos (bacterianas, virus o parásitarias) que pueden actuar nivel local en el tubo digestivo, pero también pueden influir a nivel sistémico. Los HMOs modularían el desarrollo de la inmunidad innata y adaptativa, y probablemente previenen el desarrollo de fenómenos de atopia/alergia. Una patología propia de la etapa neonatal de los prematuros es la enterocolitis necrosante y algunos HMOs podrían disminuir el riesgo de su manifestación. Las actividades de los oligosacáridos de la leche materna contribuyen a la adaptación del lactante a los desafíos que plantea su entorno incluyendo la prevención de algunas patologías en edades posteriores, como es el caso de la diabetes tipo 1 y la obesidad.


During the first months of life, breast milk oligosaccharides (HMOs) stimulate development of the gastrointestinal tract in newborns and young infants; they modulate its metabolism and transport capabilities. Additionally, they exert prebiotic and antimicrobial activities and contribute to the development of the resident intestinal microbiota with a predominance of Bifidobacterium and Bacteroides and protect from colonization and infections by enteropathogens (bacteria, virus or parasites). It is highly probable that their activities extend beyond infancy and persist into adult life. HMOs stimulate the development of the innate and adaptive immune systems and decrease the risk of atopy/allergy. Their intake has been associated with a degree of protection against as necrotizing enterocolitis among premature infants. HMOs contribute to the long term adaptation and protection of newborn infants to unfavorable conditions of their environment and in this way may contribute to protect breastfed infants from type 1 diabetes and obesity.


Assuntos
Oligossacarídeos/fisiologia , Microbioma Gastrointestinal , Leite Humano , Oligossacarídeos/imunologia
4.
Carbohydr Polym ; 186: 402-410, 2018 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-29456003

RESUMO

Swallow root pectic oligosaccharide fraction (SRO1) from swallow root pectic polysaccharide (SRPP) possessed a molecular size of 831 Da. Structural analysis revealed that it is a rhamnogalacturonan I type, bearing arabinogalactan side chain with ß-d-(1→4) galactose along with α-l-Araf (1→5)-α-l-Araf (1→3) structure on α-d-GalA-OAc-(1→2)-α-l-Rha-(1→4)- linear backbone. ß-d (1→4) linked galactose being the specific sugar for galectin-3, SRO1 had potentials in inhibiting galectin-3 mediated cancer progression. SRO1 inhibited galectin-3 mediated agglutination, in vitro, effectively with MIC of 1.08 µg/ mL and down regulated mRNA levels of galectin-3 (∼92%) along with its downstream key protein that inhibits apoptosis - survivin (∼78%) suggesting the capability of SRO1 in inhibiting galectin-3 mediated cancer promoting pathway. This is the first report, which highlights the inhibition of interplay of galectin-3 and survivin by a dietary pectic oligosaccharide.


Assuntos
Galectina 3/metabolismo , Proteínas Inibidoras de Apoptose/metabolismo , Oligossacarídeos/química , Oligossacarídeos/fisiologia , Pectinas/química , Proteínas Repressoras/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Camundongos , Survivina
5.
J Oral Pathol Med ; 46(9): 759-765, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28425129

RESUMO

BACKGROUND: Tumor-infiltrating lymphocytes (TILs) reportedly play a pivotal role in antitumor immunity against oral squamous cell carcinoma (OSCC); however, mechanisms governing TIL recruitment to OSCC tissues remain to be clarified. This study was undertaken to assess a potential association between TILs and high endothelial venule (HEV)-like vessels that express sialyl 6-sulfo Lewis X (LeX). METHODS: OSCC tissue sections (n=41) were subjected to immunohistochemistry for sialyl 6-sulfo LeX and CD34 to allow quantitation of HEV-like vessels. Triple immunohistochemistry for sialyl 6-sulfo LeX and either CD3 and CD20 or CD4 and CD8 was conducted to determine which lymphocyte subset is more closely associated with HEV-like vessels. RESULTS: HEV-like vessels expressing sialyl 6-sulfo LeX were detected in 27 of 41 (65.9%) OSCC cases, and these vessels were more frequently found in early disease (T1/T2 stages) compared with advanced (T3/T4) stages. The number of T cells attached to the inner surface of these HEV-like vessels was significantly greater than that of B cells, while the number of CD4+ helper T cells and CD8+ cytotoxic T cells did not differ significantly. Interestingly, sialyl 6-sulfo LeX was also expressed on the membrane of a fraction of OSCC cells, and CD8+ cytotoxic T cells were almost exclusively found attached to these carcinoma cells. CONCLUSIONS: Sialyl 6-sulfo LeX is displayed not only on HEV-like vessels but also on OSCC cells and may potentially function in antitumor immunity against OSCC.


Assuntos
Carcinoma de Células Escamosas/imunologia , Citotoxicidade Imunológica , Linfócitos do Interstício Tumoral/imunologia , Neoplasias Bucais/imunologia , Oligossacarídeos/fisiologia , Idoso , Carcinoma de Células Escamosas/patologia , Feminino , Humanos , Imuno-Histoquímica , Antígenos CD15/análogos & derivados , Masculino , Neoplasias Bucais/patologia , Antígeno Sialil Lewis X/análogos & derivados
6.
J Clin Gastroenterol ; 50 Suppl 2, Proceedings from the 8th Probiotics, Prebiotics & New Foods for Microbiota and Human Health meeting held in Rome, Italy on September 13-15, 2015: S131-S132, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27741156

RESUMO

Milk oligosaccharides have many associated bioactivities which can contribute to human health and offer protective properties to the host. Such bioactivities include anti-infective properties whereby oligosaccharides interact with bacterial cells and prevent adhesion to the host and subsequent colonization. Milk oligosaccharides have also been shown to alter the glycosylation of intestinal cells, leading to a reduction in pathogenic colonization. In addition, these sugars promote adhesion of commensal bacterial strains to host cells as well as possessing the ability to alter mucin expression in intestinal cells and improve barrier function. The ability of milk oligosaccharides to alter the transcriptome of both commensal bacterial strains and intestinal epithelial cells has also been revealed, indicating the potential of many cell types to detect the presence of milk oligosaccharides and respond accordingly at the genetic level. Interestingly, domestic animal milk may provide a bioactive source of oligosaccharides for formula supplementation with the aim of emulating the gold standard that is human milk. Overall, this review highlights the ability of milk oligosaccharides to promote health in a variety of ways, for example, through direct bacterial interactions, immunomodulatory activities, promotion of gut barrier function, and induction of protective transcriptional responses.


Assuntos
Interações Hospedeiro-Patógeno , Mucosa Intestinal/metabolismo , Leite/química , Oligossacarídeos/fisiologia , Animais , Células Epiteliais/metabolismo , Glicosilação , Humanos , Imunomodulação/fisiologia , Mucosa Intestinal/citologia , Mucosa Intestinal/microbiologia , Intestinos/citologia , Intestinos/microbiologia , Mucinas/metabolismo
7.
Early Hum Dev ; 91(11): 619-22, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26375354

RESUMO

Human milk oligosaccharides (HMOs) are a group of complex sugars that are highly abundant in human milk, but currently not present in infant formula. More than a hundred different HMOs have been identified so far. The amount and composition of HMOs are highly variable between women, and each structurally defined HMO might have a distinct functionality. HMOs are not digested by the infant and serve as metabolic substrates for select microbes, contributing to shape the infant gut microbiome. HMOs act as soluble decoy receptors that block the attachment of viral, bacterial or protozoan parasite pathogens to epithelial cell surface sugars, which may help prevent infectious diseases in the gut and also the respiratory and urinary tracts. HMOs are also antimicrobials that act as bacteriostatic or bacteriocidal agents. In addition, HMOs alter host epithelial and immune cell responses with potential benefits for the neonate. The article reviews current knowledge as well as future challenges and opportunities related to the functional biology of HMOs.


Assuntos
Leite Humano/química , Oligossacarídeos/fisiologia , Anti-Infecciosos/química , Anti-Infecciosos/metabolismo , Feminino , Fucose/metabolismo , Glicosilação , Humanos , Lactente , Fórmulas Infantis/química , Recém-Nascido/imunologia , Antígenos do Grupo Sanguíneo de Lewis/metabolismo , Leite Humano/metabolismo , Oligossacarídeos/metabolismo
8.
Scand J Immunol ; 80(1): 12-21, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24684681

RESUMO

Human multipotent mesenchymal stromal/stem cells (MSCs) have been shown to exert immunomodulatory properties that have great potential in therapies for various inflammatory and autoimmune disorders. However, intravenous delivery of these cells is followed by massive cell entrapment in the lungs and insufficient homing to target tissues or organs. In targeting to tissues, MSCs and other therapeutic cells employ similar mechanisms as leucocytes, including a cascade of rolling and adhesion steps mediated by selectins, integrins and their ligands. However, the mechanisms of MSCs homing are not well understood. We discovered that P-selectin (CD62P) binds to umbilical cord blood (UCB)-derived MSCs independently of the previously known sialyl Lewis x (sLex)-containing ligands such as P-selectin glycoprotein ligand-1 (PSGL-1, CD162). By biochemical assays, we identified galectin-1 as a novel ligand for P-selectin. Galectin-1 has previously been shown to be a key mediator of the immunosuppressive effects of human MSCs. We conclude that this novel interaction is likely to play a major role in the immunomodulatory targeting of human UCB-derived MSCs.


Assuntos
Sangue Fetal/citologia , Galectina 1/fisiologia , Células-Tronco Mesenquimais/fisiologia , Selectina-P/fisiologia , Humanos , Glicoproteínas de Membrana/fisiologia , Oligossacarídeos/fisiologia , Antígeno Sialil Lewis X
9.
Biochem J ; 457(1): 79-87, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24099577

RESUMO

We have shown previously that the pro-inflammatory cytokine TNF (tumour necrosis factor) could drive sLe(x) (sialyl-Lewis(x)) biosynthesis through the up-regulation of the BX transcript isoform of the ST3GAL4 (ST3 ß-galactoside α-2,3-sialyltransferase 4) sialyltransferase gene in lung epithelial cells and human bronchial mucosa. In the present study, we show that the TNF-induced up-regulation of the ST3GAL4 BX transcript is mediated by MSK1/2 (mitogen- and stress-activated kinase 1/2) through the ERK (extracellular-signal-regulated kinase) and p38 MAPK (mitogen-activated protein kinase) pathways, and increases sLe(x) expression on high-molecular-mass glycoproteins in inflamed airway epithelium. We also show that the TNF-induced sLe(x) expression increases the adhesion of the Pseudomonas aeruginosa PAO1 and PAK strains to lung epithelial cells in a FliD-dependent manner. These results suggest that ERK and p38 MAPK, and the downstream kinase MSK1/2, should be considered as potential targets to hamper inflammation, bronchial mucin glycosylation changes and P. aeruginosa binding in the lung of patients suffering from lung diseases such as chronic bronchitis or cystic fibrosis.


Assuntos
Aderência Bacteriana/efeitos dos fármacos , Pseudomonas aeruginosa/efeitos dos fármacos , Mucosa Respiratória/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas 90-kDa/fisiologia , Sialiltransferases/genética , Fator de Necrose Tumoral alfa/farmacologia , Proteínas de Bactérias/fisiologia , Brônquios/efeitos dos fármacos , Brônquios/enzimologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Oligossacarídeos/fisiologia , Pseudomonas aeruginosa/fisiologia , Mucosa Respiratória/metabolismo , Antígeno Sialil Lewis X , Sialiltransferases/metabolismo , Células Tumorais Cultivadas , Regulação para Cima/efeitos dos fármacos , beta-Galactosídeo alfa-2,3-Sialiltransferase
10.
Gastroenterology ; 142(5): 1172-82, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22333949

RESUMO

BACKGROUND & AIMS: Oligosaccharide modifications induce various functional changes in immune cells. The galactose-deficient fraction of fucosylated IgG oligosaccharides is increased, whereas that of ß-1,4-galactosyltransferase I (B4GalTI) is reduced, in patients with Crohn's disease. We investigated the role of oligosaccharide modification in the pathophysiology of colitis using B4galt1-deficient mice. METHODS: Colitis severity was compared between B4galt1(+/-) and B4galt1(+/+) mice. B cells isolated from B4galt1(+/-) and B4galt1(+/+) mice were adoptively transferred to recombination activating gene 2(-/-) mice, in which colitis was induced by administration of CD4(+)CD62L(+) T cells. Cell-surface glycan profiles were determined by lectin microarray analysis. Cytokine production was determined in a coculture of various types of cells isolated from either B4galt1(+/-) or B4galt1(+/+) mice. RESULTS: Colitis induction by dextran sodium sulfate or trinitrobenzene sulfonic acid was significantly reduced in B4galt1(+/-) mice, which had galactose deficiency in IgG oligosaccharides (similar to patients with Crohn's disease) compared with B4galt1(+/+) mice. Amelioration of colitis was associated with increased production of interleukin-10 by macrophages in B4galt1(+/-) mice. Colitis induction in recombination activating gene 2(-/-) mice by administration of CD4(+)CD62L(+) T cells was reduced by cotransfer of B cells isolated from B4galt1(+/-), but not from B4galt1(+/+) mice. Lectin microarray analysis revealed increased expression of polylactosamines on B4galt1(+/-) B cells and macrophages, compared with B4galt1(+/+) cells. The production of interleukin-10 from macrophages was induced via their direct interaction with B4galt1(+/-) B cells. CONCLUSIONS: Altered oligosaccharide structures on immune cells modulate mucosal inflammation. Oligosaccharides in immune cells might be a therapeutic target for inflammatory bowel diseases.


Assuntos
Colite/prevenção & controle , Galactosiltransferases/fisiologia , Oligossacarídeos/fisiologia , Animais , Antígeno CD11b/análise , Comunicação Celular , Colite/imunologia , Galectinas/fisiologia , Interleucina-10/biossíntese , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Oligossacarídeos/química
11.
Reproduction ; 142(5): 667-74, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21896636

RESUMO

Spermatogonia are a potential source of adult pluripotent stem cells and can be used for testis germ cell transplantation. Markers for the isolation of these cells are of great importance for biomedical applications. Primordial germ cells and prepubertal spermatogonia in many species can be identified by their binding of Dolichos biflorus agglutinin (DBA). This lectin binds to two different types of glycans, which are α-linked N-acetylgalactosamine (GalNac) and ß-linked GalNac, if this is part of the Sda or GM2 glycotopes. We used the MAB CT1, which is specific for the trisaccharides motif NeuAcα2-3(GalNAcß1-4)Galß1-, which is common to both Sda and GM2 glycotopes, to further define the glycosylation of DBA binding germ cells. In porcine embryos, CT1 bound to migratory germ cells and gonocytes. CT1/DBA double staining showed that the mesonephros was CT1 negative but contained DBA-positive cells. Gonocytes in the female gonad became CT1 negative, while male gonocytes remained CT1 positive. In immunohistological double staining of cattle, pig, horse and llama testis, DBA and CT1 staining was generally colocalised in a subpopulation of spermatogonia. These spermatogonia were mainly single, sometimes paired or formed chains of up to four cells. Our data show that the Sda/GM2 glycotope is present in developing germ cells and spermatogonia in several species. Owing to the narrower specificity of the CT1 antibody, compared with DBA, the former is likely to be a useful tool for labelling and isolation of these cells.


Assuntos
Gangliosídeo G(M2)/fisiologia , Células Germinativas/metabolismo , Oligossacarídeos/fisiologia , Polissacarídeos/fisiologia , Espermatogônias/classificação , Espermatogônias/metabolismo , Animais , Especificidade de Anticorpos , Biomarcadores/química , Biomarcadores/metabolismo , Camelídeos Americanos/metabolismo , Sequência de Carboidratos , Bovinos/metabolismo , Feminino , Gangliosídeo G(M2)/química , Gangliosídeo G(M2)/imunologia , Gangliosídeo G(M2)/metabolismo , Células Germinativas/classificação , Células Germinativas/citologia , Cavalos/metabolismo , Masculino , Dados de Sequência Molecular , Oligossacarídeos/química , Oligossacarídeos/imunologia , Oligossacarídeos/metabolismo , Polissacarídeos/química , Polissacarídeos/imunologia , Polissacarídeos/metabolismo , Ligação Proteica , Espermatogônias/citologia , Suínos/metabolismo
12.
PLoS One ; 6(1): e16281, 2011 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-21283832

RESUMO

Similar to mechanisms of recruitment of activated leukocytes to inflamed tissues, selectins mediate adhesion and extravasation of circulating cancer cells. Our objective was to determine whether sialyl Lewis X modified core 2 O-glycans (C2-O-sLe(X)) present on colon and hepatic carcinoma cells promote their adhesion and invasion. We examined membrane expression of C2-O-sLe(X), selectin binding, invasion of human colon and hepatic carcinoma cell lines, and mRNA levels of alpha-2,3 fucosyltransferase (FucT-III) and core 2 beta-1,6 N-acetylglucosaminyltransferase (C2GnT1) genes, necessary for C2-O-sLe(X) synthesis, by quantitative reverse-transcriptase (RT) PCR. Synthesis of core 2 branched O-glycans decorated by sLe(X) is dependent on C2GnT1 function and thus we determined enzyme activity of C2GnT1. The cell lines that expressed C2GnT1 and FucT-III mRNA by quantitative RT-PCR were highly positive for C2-O-sLe(X) by flow cytometry, and colon carcinoma cells possessed highly active C2GnT1 enzyme. Cells bound avidly to E-selection but not to P- and L-selectin. Gene knock-down of C2GnT1 in colon and hepatic carcinoma cells using short hairpin RNAs (shRNA) resulted in a 40-90% decrease in C2-O-sLe(X) and a 30-50% decrease in E-selectin binding compared to control cells. Invasion of hepatic and colon carcinoma cells containing C2GnT1 shRNA was significantly reduced compared to control cells in Matrigel assays and C2GnT1 activity was down-regulated in the latter cells. The sLe(X) epitope was predominantly distributed on core 2 O-glycans on colon and hepatic carcinoma cells. Our findings indicate that C2GnT1 gene expression and the resulting C2-O-sLe(X) carbohydrates produced mediate the adhesive and invasive behaviors of human carcinomas which may influence their metastatic potential.


Assuntos
Carcinoma Hepatocelular/patologia , Neoplasias do Colo/patologia , Selectina E/metabolismo , Invasividade Neoplásica/patologia , Oligossacarídeos/fisiologia , Carcinoma Hepatocelular/química , Adesão Celular , Linhagem Celular Tumoral , Neoplasias do Colo/química , Epitopos/análise , Glicoproteínas/análise , Glicoproteínas/fisiologia , Humanos , Ligantes , N-Acetilglucosaminiltransferases/genética , Oligossacarídeos/análise , RNA Mensageiro/análise
13.
Exp Biol Med (Maywood) ; 236(1): 84-90, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21239738

RESUMO

Oligosaccharides of hyaluronan (o-HA) can induce angiogenesis and the growth and tube formation of vascular endothelial cells (ECs) in particular. As the major o-HA receptor, CD44 has been implicated in EC function, but its role in mediating o-HA-induced EC proliferation and tube formation remains unclear. In this study, we investigated the role of CD44 in o-HA-induced proliferation and tube formation of human umbilical vein endothelial cells (HUVECs) and explored the molecular mechanisms underlying the angiogenesis process. A CD44 siRNA was delivered into HUVECs by electroporation and o-HA-induced proliferation and tube formation capacity of CD44-silenced or control HUVECs were assessed by methylthiazolyldiphenyl-tetrazolium bromide (MTT) and Matrigel assays. Furthermore, the changes in Src, focal adhesion kinase (FAK) and extracellular signal-regulated kinase1 and 2 (ERK1/2) phosphorylation, as well as the expression of c-jun and c-fos were examined by Western blot and realtime-polymerase chain reaction assays. Our results demonstrated that 10 µg/mL o-HA obviously induced the proliferation and tube formation in HUVECs, and stimulated the phosphorylation of Src, FAK and ERK1/2 and upregulation of c-jun and c-fos, which could be inhibited by CD44 silencing. Altogether our data suggest that CD44 functions to initiate tyrosine phosphorylation of Src, FAK and ERK1/2, and upregulates the expression of c-jun and c-fos, thus mediating o-HA-induced proliferation and tube formation in HUVECs.


Assuntos
Proliferação de Células/efeitos dos fármacos , Endotélio Vascular/crescimento & desenvolvimento , Receptores de Hialuronatos/fisiologia , Ácido Hialurônico/farmacologia , Oligossacarídeos/fisiologia , Transdução de Sinais/fisiologia , Western Blotting , Células Cultivadas , Relação Dose-Resposta a Droga , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiologia , Genes fos/efeitos dos fármacos , Genes fos/fisiologia , Genes jun/efeitos dos fármacos , Genes jun/fisiologia , Humanos , Receptores de Hialuronatos/biossíntese , Receptores de Hialuronatos/metabolismo , Oligossacarídeos/metabolismo , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Tirosina/metabolismo , Veias Umbilicais/citologia , Veias Umbilicais/metabolismo , Regulação para Cima/efeitos dos fármacos
14.
Mol Cell Biochem ; 350(1-2): 185-92, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21197561

RESUMO

The complex implantation process is initiated by the recognition and adhesion between the embryo and uterine endometrial epithelium. The expression and interactions between the adhesive molecules from both fetal and maternal sides are crucial for the successful implantation. In this study, we aimed to investigate the expression and adhesive function of sLeX on the trophoblasts and L-selectin on uterine epithelial cells mediated the adhesion at the fetal-maternal interface, and to further explore whether this adhesion system could induce endometrial apoptosis, using in vitro implantation model consisting of the human trophoblast cell line (JAR) and human uterine epithelial cell line (RL95-2). The results showed that sLeX was expressed on JAR cells by indirect immunofluorescence staining. After transfection of JAR cells with fucosyltransferase VII (FUT7) which is the key enzyme for sLeX synthesis, the expression of FUT7 and sLeX synthesis were increased, and the percent adhesion of trophoblast cells to RL95-2 cell monolayer was significantly increased (P < 0.01). L-selectin was strongly expressed but not E- and P-selectin on epithelial RL95-2 cells by RT-PCR, Western blot. Blocking L-selectin with specific antibody or heparin pretreatment in RL95-2 cells inhibited the adhesion of JAR cells to RL95-2 cell monolayer. Furthermore, regulating the expression of sLeX on JAR cells or blocking L-selectin on RL95-2 cells could activate the apoptosis of uterine epithelial cells. These results suggest the sLeX/L-selectin adhesion system at fetal-maternal interface not only mediates the adhesion of embryo to uterine epithelium, but also effectively induces the apoptosis in uterine epithelium. The study supplies a molecular basis for the elucidation of the initial recognition and adhesion during embryo implantation.


Assuntos
Implantação do Embrião/fisiologia , Selectina L/fisiologia , Oligossacarídeos/fisiologia , Anticorpos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Adesão Celular/efeitos dos fármacos , Adesão Celular/genética , Adesão Celular/fisiologia , Células Cultivadas , Implantação do Embrião/efeitos dos fármacos , Implantação do Embrião/genética , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/fisiologia , Feminino , Fucosiltransferases/genética , Fucosiltransferases/metabolismo , Humanos , Selectina L/metabolismo , Modelos Teóricos , Oligossacarídeos/antagonistas & inibidores , Oligossacarídeos/imunologia , Oligossacarídeos/farmacologia , Antígeno Sialil Lewis X , Transfecção , Trofoblastos/efeitos dos fármacos , Trofoblastos/metabolismo , Trofoblastos/fisiologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Útero/citologia , Útero/metabolismo , Útero/fisiologia
15.
Biochemistry ; 49(36): 7811-20, 2010 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-20695481

RESUMO

Carbohydrate structures with a 3'-sulfo betaGal linkage, such as 3'-sulfo-Le(x), can be synthesized by Gal:3-O-sulfotransferase-2 (Gal3ST-2) catalysis, but little is known about their roles in many biological processes. To investigate the role of Gal3ST-2 and its product 3'-sulfo-Le(x), we depleted Gal3ST-2 via siRNA and added exogenous Lewis-x trisaccharide 3'-sulfate sodium salt in human SMMC7721 hepatoma cells. After siRNA transfection, a striking morphological change in SMMC7721 hepatoma cells from polygon to shuttle shape and a significant decrease in the level of adhesion to sL-selectin, HUVEC, fibronectin, vitronectin, and fibrinogen were observed. The expression of integrin subunit alphaV was markedly downregulated, and 3'-sulfated subunit alphaV almost disappeared in the transfectants. The level of cell surface integrin alphaVbeta3 was reduced simultaneously, although total subunit beta3 underwent almost no change. After treatment with exogenous Lewis-x 3'-sulfate, cellular integrin subunit alphaV was upregulated and the level of cell surface integrin alphaVbeta3 was elevated. Interestingly, knockdown of Gal3ST-2 expression effectively inhibited cell proliferation, and the result was significantly correlated with the decrease in the levels of ILK, phosphorylated AKT, and ERK. On the other hand, treatment with Lewis-x trisaccharide 3'-sulfate sodium salt greatly upregulated the phosphorylation of AKT and ERK. Our results also indicated that downregulation of Gal3ST-2 via siRNA transfection was associated with the decrease in the level of expression of anti-apoptotic protein, Bcl-2, with a consequent decrease in the ratios for Bcl-2 to Bax. By exposure to Lewis-x trisaccharide 3'-sulfate sodium salt, the apoptotic response of cells was inhibited. Therefore, Gal3ST-2 and its product, 3'-sulfo-Le(x), were involved in regulation of integrin subunit alphaV and might be associated with cancer cell regulation.


Assuntos
Integrina alfaV/metabolismo , Oligossacarídeos/farmacologia , Sulfotransferases/antagonistas & inibidores , Linhagem Celular Tumoral , Proliferação de Células , Regulação para Baixo , Humanos , Integrina alfaV/química , Antígenos CD15/análogos & derivados , Oligossacarídeos/fisiologia , RNA Interferente Pequeno/metabolismo , Sulfotransferases/genética , Sulfotransferases/metabolismo , Transfecção , Trissacarídeos/química
16.
J Exp Med ; 206(13): 3061-72, 2009 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-20008521

RESUMO

Variation of surface antigen expression is a mechanism used by microbes to adapt to and persist within their host habitats. Helicobacter pylori, a persistent bacterial colonizer of the human stomach, can alter its surface Lewis (Le) antigen expression. We examined H. pylori colonization in mice to test the hypothesis that host phenotype selects for H. pylori (Le) phenotypes. When wild-type and Le(b)-expressing transgenic FVB/N mice were challenged with H. pylori strain HP1, expressing Le(x) and Le(y), we found that bacterial populations recovered after 8 mo from Le(b)-transgenic, but not wild-type, mice expressed Le(b). Changes in Le phenotype were linked to variation of a putative galactosyltransferase gene (beta-(1,3)galT); mutagenesis and complementation revealed its essential role in type I antigen expression. These studies indicate that H. pylori evolves to resemble the host's gastric Le phenotype, and reveal a bacterial genetic locus that is subject to host-driven selection pressure.


Assuntos
Helicobacter pylori/imunologia , Antígenos do Grupo Sanguíneo de Lewis/análise , Antígenos CD15/análise , Oligossacarídeos/fisiologia , Adesinas Bacterianas/análise , Animais , Anticorpos Antibacterianos/sangue , Citometria de Fluxo , Fucosiltransferases/genética , Galactosiltransferases/genética , Lipopolissacarídeos/toxicidade , Camundongos , Camundongos Transgênicos , Fenótipo
17.
Curr Med Chem ; 16(18): 2338-44, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19519394

RESUMO

Sulfated oligosaccharides display an important role in biological processes. They bind proteins through interactions mediated by highly specific sequences (heparin - antithrombin, heparan sulfate - growth factors / herpes simplex virus) or by electrostatic interaction between sulfate groups and cationic sites of proteins. Sulfated oligosaccharides are involved in biological events as protein localisation at cell surfaces, the control of proteolysis, the modulation of the angiogenesis and metastasis of tumours, the oligomerisation of cell growth factors. Sulfated residues have been recently found in glycoproteins, as GlcNAc or Mannose in N-glycosidic chains of different sources: gp120 of HIV, the envelope glycoprotein of influenza virus, the cysteine protease of Trypanosoma cruzi. This paper reviews recent findings concerning the implication of sulfated sugars in carbohydrate - protein interactions, from glycosaminoglycans to glycosidic chains in proteins, and their potential application as new targets for drugs/vaccines/diagnosis developments will be discussed. New approaches for their detection and analysis (ESI-MS, MALDI, Molecular Imprinting) are presented.


Assuntos
Anticorpos Monoclonais/farmacologia , Desenho de Fármacos , Oligossacarídeos/química , Ésteres do Ácido Sulfúrico/química , Animais , Anticorpos Monoclonais/imunologia , Humanos , Impressão Molecular , Oligossacarídeos/imunologia , Oligossacarídeos/metabolismo , Oligossacarídeos/farmacologia , Oligossacarídeos/fisiologia , Ligação Proteica , Espectrometria de Massas por Ionização por Electrospray , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Relação Estrutura-Atividade , Ésteres do Ácido Sulfúrico/imunologia , Ésteres do Ácido Sulfúrico/metabolismo , Ésteres do Ácido Sulfúrico/farmacologia
19.
Regul Pept ; 148(1-3): 62-7, 2008 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-18433898

RESUMO

Thyrostimulin is a heterodimeric hormone composed of GPA2 and GPB5, and shares the thyroid-stimulating hormone receptor (TSHR). Thyrostimulin has three N-linked oligosaccharide chains, two in GPA2 and one in GPB5. The roles of these N-linked oligosaccharides in secretion, heterodimer formation and signal transduction were analyzed. Recombinant GPA2s lacking either of the two oligosaccharides were obtained from conditioned medium, whereas dual site-disrupted GPA2 and the GPB5 mutant were not expressed in either the conditioned medium or cell lysate. The binding between GPA2 and GPB5 was weaker than that between TSH subunits GPA1 and TSH beta. Neither of the oligosaccharides in GPA2 had significant effects on heterodimerization. Disruption of either of the oligosaccharides in GPA2 significantly decreased receptor activation, suggesting their critical role in receptor activation.


Assuntos
Glicoproteínas/metabolismo , Oligossacarídeos/metabolismo , Sequência de Aminoácidos , Western Blotting , Linhagem Celular , AMP Cíclico/metabolismo , Dimerização , Glicoproteínas/química , Glicoproteínas/genética , Humanos , Immunoblotting , Modelos Biológicos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Oligossacarídeos/genética , Oligossacarídeos/fisiologia , Ligação Proteica , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA