Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Cancer ; 150(4): 678-687, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-34741527

RESUMEN

Gynecological cancers are in the top 10 of most common cancers in women. Survival and outcome are strongly related to the stage at diagnosis. Therefore, early diagnosis is essential in reducing morbidity and mortality. The high mortality rate of gynecological cancers can mainly be attributed to ovarian cancer (OC). OC is commonly diagnosed at an advanced stage due to a lack of proper screening tools allowing early detection. Endometrial cancer (EC) on the contrary, is mostly diagnosed at an early stage and has, in general, better outcomes. The incidence of nonendometrioid EC has increased in the last decade, displaying a shared tumor biology with OC and consequently significantly worse outcome. New approaches allowing detection of gynecological cancers in an early stage are therefore desired. Recent studies on cancer biology have shown the relevance of altered glycosylation in the occurrence and progression of cancer. The aberrant expression of sialic acid, a specific carbohydrate terminating glycoproteins and glycolipids on the cell-surface, is frequently correlated with malignancy. We aimed to determine the current understanding of sialic acid function in different gynecological cancers to identify the gaps in knowledge and its potential use for new diagnostic and therapeutic avenues. Therefore we performed a review on current literature focusing on studies where sialylation was linked to gynecological cancers. The identified studies showed elevated levels of sialic acid in serum, tissue and sialylated antigens in most patients with gynecological cancers, underlining its potential for diagnosis.


Asunto(s)
Neoplasias de los Genitales Femeninos/etiología , Ácidos Siálicos/fisiología , Animales , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Neoplasias de los Genitales Femeninos/diagnóstico , Neoplasias de los Genitales Femeninos/terapia , Glicosilación , Humanos , Ácidos Siálicos/análisis , Sialiltransferasas/fisiología , beta-Galactosida alfa-2,3-Sialiltransferasa
2.
J Biol Chem ; 296: 100034, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33148698

RESUMEN

ST6Gal-I, an enzyme upregulated in numerous malignancies, adds α2-6-linked sialic acids to select membrane receptors, thereby modulating receptor signaling and cell phenotype. In this study, we investigated ST6Gal-I's role in epithelial to mesenchymal transition (EMT) using the Suit2 pancreatic cancer cell line, which has low endogenous ST6Gal-I and limited metastatic potential, along with two metastatic Suit2-derived subclones, S2-013 and S2-LM7AA, which have upregulated ST6Gal-I. RNA-Seq results suggested that the metastatic subclones had greater activation of EMT-related gene networks than parental Suit2 cells, and forced overexpression of ST6Gal-I in the Suit2 line was sufficient to activate EMT pathways. Accordingly, we evaluated expression of EMT markers and cell invasiveness (a key phenotypic feature of EMT) in Suit2 cells with or without ST6Gal-I overexpression, as well as S2-013 and S2-LM7AA cells with or without ST6Gal-I knockdown. Cells with high ST6Gal-I expression displayed enrichment in mesenchymal markers (N-cadherin, slug, snail, fibronectin) and cell invasiveness, relative to ST6Gal-I-low cells. Contrarily, epithelial markers (E-cadherin, occludin) were suppressed in ST6Gal-I-high cells. To gain mechanistic insight into ST6Gal-I's role in EMT, we examined the activity of epidermal growth factor receptor (EGFR), a known EMT driver. ST6Gal-I-high cells had greater α2-6 sialylation and activation of EGFR than ST6Gal-I-low cells. The EGFR inhibitor, erlotinib, neutralized ST6Gal-I-dependent differences in EGFR activation, mesenchymal marker expression, and invasiveness in Suit2 and S2-LM7AA, but not S2-013, lines. Collectively, these results advance our understanding of ST6Gal-I's tumor-promoting function by highlighting a role for ST6Gal-I in EMT, which may be mediated, at least in part, by α2-6-sialylated EGFR.


Asunto(s)
Transición Epitelial-Mesenquimal/fisiología , Neoplasias Pancreáticas/patología , Sialiltransferasas/fisiología , Biomarcadores/metabolismo , Línea Celular Tumoral , Receptores ErbB/metabolismo , Humanos , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias Pancreáticas/enzimología , beta-D-Galactósido alfa 2-6-Sialiltransferasa
3.
Front Immunol ; 11: 617, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32391003

RESUMEN

Humoral immunity is an effective but metabolically expensive defense mechanism. It is unclear whether systemic cues exist to communicate the dynamic need for antigen presentation and immunoglobulin production. Here, we report a novel role for the liver-produced, acute phase reactant ST6GAL1 in IgG production. B cell expression of ST6GAL1, a sialyltransferase mediating the attachment of α2,6-linked sialic acids on N-glycans, is classically implicated in the dysregulated B cell development and immunoglobulin levels of St6gal1-deficient mice. However, the blood-borne pool of ST6GAL1, upregulated during systemic inflammation, can also extrinsically modify leukocyte cell surfaces. We show that B cell independent, extracellular ST6GAL1 enhances B cell IgG production and increases blood IgG titers. B cells of mice lacking the hepatocyte specific St6gal1 promoter have reduced sialylation of cell surface CD22 and CD45 and produce less IgG upon stimulation. Sialylation of B cells by extracellular ST6GAL1 boosts expression of IgM, IgD, and CD86, proliferation, and IgG production in vitro. In vivo, elevation of blood ST6GAL1 enhances B cell development and systemic IgG in a CD22-dependent manner. Our data point to a function of an extracellular glycosyltransferase in promoting humoral immunity. Manipulation of systemic ST6GAL1 may represent an effective therapeutic approach for humoral insufficiency.


Asunto(s)
Linfocitos B/inmunología , Inmunoglobulina G/biosíntesis , Sialiltransferasas/fisiología , Animales , Inmunidad Humoral , Ratones , Ratones Endogámicos C57BL , Ácido N-Acetilneuramínico/metabolismo , Lectina 2 Similar a Ig de Unión al Ácido Siálico/fisiología , Sialiltransferasas/sangre , beta-D-Galactósido alfa 2-6-Sialiltransferasa
4.
J Chin Med Assoc ; 83(4): 337-344, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31904658

RESUMEN

Sialylation (the covalent addition of sialic acid to the terminal end of glycoproteins or glycans), tightly regulated cell- and microenvironment-specific process and orchestrated by sialyltransferases and sialidases (neuraminidases) family, is one of the posttranslational modifications, which plays an important biological role in the maintenance of normal physiology and involves many pathological dysfunctions. Glycans have roles in all the cancer hallmarks, referring to capabilities acquired during all steps of cancer development to initiate malignant transformation (a driver of a malignant genotype), enable cancer cells to survive, proliferate, and metastasize (a consequence of a malignant phenotype), which includes sustaining proliferative signaling, evading growth suppressor, resisting cell apoptosis, enabling replicative immortality, inducing angiogenesis, reprogramming of energy metabolism, evading tumor destruction, accumulating inflammatory microenvironment, and activating invasion and accelerating metastases. Regarding the important role of altered sialylation of cancers, further knowledge about the initiation and the consequences of altered sialylation pattern in tumor cells is needed, because all may offer a better chance for developing novel therapeutic strategy. In this review, we would like to update alteration of sialylation in ovarian cancers.


Asunto(s)
Neoplasias Ováricas/metabolismo , Ácidos Siálicos/metabolismo , Biomarcadores de Tumor , Proteínas Sanguíneas/metabolismo , Femenino , Humanos , Neuraminidasa/fisiología , Antígeno Sialil Lewis X/análisis , Sialiltransferasas/fisiología
5.
Osteoarthritis Cartilage ; 27(2): 314-325, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30471358

RESUMEN

OBJECTIVE: Gangliosides, ubiquitously existing membrane components that modulate transmembrane signaling and mediate cell-to-cell and cell-to-matrix interactions, are key molecules of inflammatory and neurological disorders. However, the functions of gangliosides in the cartilage degradation process remain unclear. We investigated the functional role of gangliosides in cartilage metabolism related to osteoarthritis (OA) pathogenesis. DESIGN: We generated knockout (KO) mice by targeting the ß1, 4-N-acetylgalactosaminyltransferase (GalNAcT) gene, which encodes an enzyme of major gangliosides synthesis, and the GD3 synthase (GD3S) gene, which encodes an enzyme of partial gangliosides synthesis. In vivo OA and in vitro cartilage degradation models were used to evaluate the effect of gangliosides on the cartilage degradation process. RESULTS: The GalNAcT and GD3S KO mice developed and grew normally; nevertheless, OA changes in these mice were enhanced with aging. The GalNAcT KO mice showed significantly enhanced OA progression compared to GD3S mice in vivo. Both GalNAcT and GD3S KO mice showed severe IL-1α-induced cartilage degradation ex vivo. Phosphorylation of MAPKs was enhanced in both GalNAcT and GD3S KOs after IL-1α stimulation. Gangliosides modulated by GalNAcT or GD3S rescued an increase of MMP-13 induced by IL-1α in mice lacking GalNAcT or GD3S after exogenous replenishment in vitro. CONCLUSION: These data show that the deletion of gangliosides in mice enhanced OA development. Moreover, the gangliosides modulated by GalNAcT are important for cartilage metabolism, suggesting that GalNAcT is a potential target molecule for the development of novel OA treatments.


Asunto(s)
Artritis Experimental/metabolismo , Cartílago Articular/metabolismo , Gangliósidos/fisiología , Osteoartritis/metabolismo , Envejecimiento/fisiología , Animales , Artritis Experimental/patología , Cartílago Articular/efectos de los fármacos , Cartílago Articular/patología , Células Cultivadas , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Progresión de la Enfermedad , Gangliósidos/deficiencia , Gangliósidos/farmacología , Eliminación de Gen , Crecimiento/genética , Interleucina-1alfa/antagonistas & inhibidores , Interleucina-1alfa/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Metaloproteinasa 13 de la Matriz/biosíntesis , Ratones Noqueados , N-Acetilgalactosaminiltransferasas/deficiencia , N-Acetilgalactosaminiltransferasas/genética , N-Acetilgalactosaminiltransferasas/fisiología , Óxido Nítrico/metabolismo , Osteoartritis/patología , Sialiltransferasas/deficiencia , Sialiltransferasas/genética , Sialiltransferasas/fisiología , Técnicas de Cultivo de Tejidos , Regulación hacia Arriba/fisiología , Polipéptido N-Acetilgalactosaminiltransferasa
6.
J Pathol ; 247(5): 629-640, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30582157

RESUMEN

This review aims to provide an overview of recent developments regarding the roles of MMPs in tumour invasion and metastasis. Much of the mortality burden belonging to cancer relates to its ability to invade adjacent tissue and form metastases at distant sites. This would not be possible without remodelling of the ECM, a process which is enabled by the functions of MMPs. Recent studies provide a better understanding of the importance of the biophysical nature of the ECM, how this influences cancer cell motility, and how MMPs act to modify matrix stiffness. The regulation of MMPs and the role of immune cell generated MMPs has also become better understood. All of this provides a framework for the therapeutic targeting of MMPs and recent advances in the development of selective MMPs inhibitors are also reviewed. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Metaloproteinasas de la Matriz/fisiología , Neoplasias/patología , Antígenos CD/fisiología , Matriz Extracelular/patología , Humanos , Sistema Inmunológico/fisiología , Inhibidores de la Metaloproteinasa de la Matriz/uso terapéutico , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias/inmunología , Receptores Acoplados a Proteínas G/fisiología , Sialiltransferasas/fisiología , Terminología como Asunto , Trombospondinas/fisiología , Factor de Crecimiento Transformador beta/fisiología , beta-D-Galactósido alfa 2-6-Sialiltransferasa
7.
Sci Rep ; 7(1): 14428, 2017 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-29089525

RESUMEN

Human ß-galactoside α-2,6-sialyltransferase I (ST6Gal I) catalyses the synthesis of sialylated glycoconjugates. Overexpression of ST6Gal I is observed in many cancers, where it promotes metastasis through altered cell surface sialylation. A wide range of sialyltransferase inhibitors have been developed, with analogues structurally similar to the transition state exhibiting the highest inhibitory activity. To improve synthetic accessibility and pharmacokinetics of previously reported inhibitors, the replacement of the charged phosphodiester linker with a potential neutral isostere such as a carbamate or a 1,2,3-triazole has been investigated. Extensive molecular dynamics simulations have demonstrated that compounds with the alternate linkers could maintain key interactions with the human ST6Gal I active site, demonstrating the potential of a carbamate or a 1,2,3-triazole as a phosphodiester isostere. Free energy perturbation calculations provided energetic evidence suggesting that the carbamate and 1,2,3-triazole were slightly more favourable than the phosphodiester. Further exploration with free energy component, quasi-harmonic and cluster analysis suggested that there is an enthalpy-entropy compensation accounting for the replacement of the flexible charged phosphodiester with a neutral and rigid isostere. Overall, these simulations provide a strong rationale for the use of a carbamate or 1,2,3-triazole as a phosphodiester isostere in the development of novel inhibitors of human ST6Gal I.


Asunto(s)
Antígenos CD/fisiología , Sialiltransferasas/antagonistas & inhibidores , Sialiltransferasas/fisiología , Carbamatos/metabolismo , Dominio Catalítico , Línea Celular , Entropía , Humanos , Simulación de Dinámica Molecular , Sialiltransferasas/química , Sialiltransferasas/metabolismo , Termodinámica , Triazoles/metabolismo
8.
Cancer Sci ; 108(2): 267-276, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27906498

RESUMEN

Aberrant sialylation profiles on the cell surface have been recognized for their potential diagnostic value in identifying the regulation of tumor properties in several cancers, including hepatocellular carcinoma (HCC). Recently, increasing evidence has suggested that the deregulation of microRNA (miRNA) is a common feature in human cancers. In this study, we found obvious upregulation of sialyltransferase ST3GAL6 both in HCC cell lines and in tissue samples. The altered expression of ST3GAL6 was found to correlate with cell proliferation, migration, and invasion ability in HCC. Further investigation showed that miR-26a negatively regulated ST3GAL6, inducing the suppression of cell proliferation, migration, and invasion in vitro. Moreover, we identified the protein kinase B/mammalian target of rapamycin (Akt/mTOR) pathway as the target of ST3GAL6 based on Western blot analysis. Analysis of a xenograft mouse model showed that miR-26a significantly reduced tumor growth by suppressing activation of the Akt/mTOR pathway by directly targeting ST3GAL6. In conclusion, these data indicate that ST3GAL6 promotes cell growth, migration, and invasion and mediates the effect of miR-26a through the Akt/mTOR signaling pathway in HCC.


Asunto(s)
Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , MicroARNs/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sialiltransferasas/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/secundario , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Humanos , Neoplasias Hepáticas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Transducción de Señal , Ensayo de Tumor de Célula Madre , Regulación hacia Arriba , beta-Galactosida alfa-2,3-Sialiltransferasa
9.
ASN Neuro ; 8(6)2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27879349

RESUMEN

Much confusion surrounds the physiological function of the cellular prion protein (PrPC). It is, however, anticipated that knowledge of its function will shed light on its contribution to neurodegenerative diseases and suggest ways to interfere with the cellular toxicity central to them. Consequently, efforts to elucidate its function have been all but exhaustive. Building on earlier work that uncovered the evolutionary descent of the prion founder gene from an ancestral ZIP zinc transporter, we recently investigated a possible role of PrPC in a morphogenetic program referred to as epithelial-to-mesenchymal transition (EMT). By capitalizing on PrPC knockout cell clones in a mammalian cell model of EMT and using a comparative proteomics discovery strategy, neural cell adhesion molecule-1 emerged as a protein whose upregulation during EMT was perturbed in PrPC knockout cells. Follow-up work led us to observe that PrPC regulates the polysialylation of the neural cell adhesion molecule NCAM1 in cells undergoing morphogenetic reprogramming. In addition to governing cellular migration, polysialylation modulates several other cellular plasticity programs PrPC has been phenotypically linked to. These include neurogenesis in the subventricular zone, controlled mossy fiber sprouting and trimming in the hippocampal formation, hematopoietic stem cell renewal, myelin repair and maintenance, integrity of the circadian rhythm, and glutamatergic signaling. This review revisits this body of literature and attempts to present it in light of this novel contextual framework. When approached in this manner, a coherent model of PrPC acting as a regulator of polysialylation during specific cell and tissue morphogenesis events comes into focus.


Asunto(s)
Antígeno CD56/metabolismo , Proteínas Priónicas/metabolismo , Sialiltransferasas/fisiología , Animales , Antígeno CD56/genética , Humanos , Neurogénesis , Proteínas Priónicas/genética , Mapas de Interacción de Proteínas , Ácidos Siálicos/metabolismo , Transducción de Señal
10.
Oncol Rep ; 36(6): 3317-3324, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27779707

RESUMEN

Changes in the carbohydrate structure on the surface of tumor cells is an important feature of cancer metastasis. The specific role of sialic acids in the glycoconjugate terminal has not yet been clearly elucidated in these processes. Previously, we reported that α2,3-sialic acid residues in breast cancer are associated with metastatic potential. The α2,3-sialyltransferase ST3Gal III, which adds α2,3-sialic acids to glycoproteins, is overexpressed in various tumors, and enzyme activity is correlated with tumor metastasis, yet its mechanistic role has not been fully evaluated. In the present study, we aimed to investigate the influence of ST3Gal III on key steps in the process of breast cancer metastasis. ST3Gal III-overexpressing and ST3Gal III-silenced breast cancer MDA-MB-231 cell lines were generated. They showed an increase or decrease in the tumor-associated antigen sialyl-Lewis X (SLeX). The E-selectin binding capacity of the transfectants was proportional to cell surface SLeX levels. Cell migration and invasion were positively correlated with ST3Gal III levels. Moreover, ST3Gal III expression modulated the protein expression of invasion-related molecules, including ß1 integrin, matrix metalloproteinase (MMP)-2, MMP-9 and cyclooxygenase-2, which may account for the mechanism involved in the effects of ST3Gal III on breast cancer invasiveness. In conclusion, our findings in these novel models of ST3Gal III expression revealed a critical requirement for ST3Gal III in several steps of breast cancer metastasis. ST3Gal III modulates breast cancer cell adhesion and invasion by altering the expression of invasion-related molecules. This study provides novel insights into the mechanisms underlying metastasis and suggests a new target for the effective drug treatment of breast cancer metastasis.


Asunto(s)
Neoplasias de la Mama/enzimología , Regulación Neoplásica de la Expresión Génica , Sialiltransferasas/fisiología , Neoplasias de la Mama/genética , Adhesión Celular , Línea Celular Tumoral , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Selectina E/metabolismo , Femenino , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Integrina beta1/genética , Integrina beta1/metabolismo , Antígeno Lewis X/metabolismo , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Invasividad Neoplásica , beta-Galactosida alfa-2,3-Sialiltransferasa
11.
Proc Natl Acad Sci U S A ; 113(20): 5592-7, 2016 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-27143722

RESUMEN

The cancer stem cells (CSCs) of glioblastoma multiforme (GBM), a grade IV astrocytoma, have been enriched by the expressed marker CD133. However, recent studies have shown that CD133(-) cells also possess tumor-initiating potential. By analysis of gangliosides on various cells, we show that ganglioside D3 (GD3) is overexpressed on eight neurospheres and tumor cells; in combination with CD133, the sorted cells exhibit a higher expression of stemness genes and self-renewal potential; and as few as six cells will form neurospheres and 20-30 cells will grow tumor in mice. Furthermore, GD3 synthase (GD3S) is increased in neurospheres and human GBM tissues, but not in normal brain tissues, and suppression of GD3S results in decreased GBM stem cell (GSC)-associated properties. In addition, a GD3 antibody is shown to induce complement-dependent cytotoxicity against cells expressing GD3 and inhibition of GBM tumor growth in vivo. Our results demonstrate that GD3 and GD3S are highly expressed in GSCs, play a key role in glioblastoma tumorigenicity, and are potential therapeutic targets against GBM.


Asunto(s)
Neoplasias Encefálicas/patología , Gangliósidos/fisiología , Glioblastoma/patología , Células Madre Neoplásicas/química , Sialiltransferasas/fisiología , Antígeno AC133/análisis , Animales , Línea Celular Tumoral , Gangliósido G(M1)/análisis , Gangliósidos/análisis , Glioblastoma/química , Glioblastoma/etiología , Humanos , Ratones , Proteínas Proto-Oncogénicas c-met/metabolismo , Sialiltransferasas/análisis
12.
Curr Top Microbiol Immunol ; 396: 131-48, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26814887

RESUMEN

The lipooligosaccharide (LOS) of Histophilus somni is a multifaceted molecule that provides critical protection to the bacterium against host defenses, may act as an adhesin, and like similar molecules of gram-negative bacteria, is an endotoxin that signals through toll-like receptor 4 and NF-κB to cause inflammation. The lipid A component is responsible for the endotoxic and apoptotic activity of the LOS. The H. somni LOS lacks O-side chains typically characteristic of gram-negative bacteria that have lipopolysaccharide, but has a complex, microheterogeneous outer core. The LOS of disease isolates is capable of undergoing structural and antigenic phase variation of its outer core due to slip-strand mispairing of glycosyltransferase genes that contain repetitive sequences of DNA base pairs. Such variation enables the bacteria to evade bactericidal antibodies made to oligosaccharide antigens. In addition, the LOS can be decorated with phase-variable phosphorylcholine (ChoP), which binds to platelet-activating factor receptor on host cells, thereby aiding in colonization of the upper respiratory tract. However, ChoP is likely not expressed when the bacteria are in systemic sites because ChoP also binds to C-reactive protein, resulting in activation of host complement and promoting bactericidal activity. The structure of some LOS outer core chains is identical to oligosaccharides on host glycosphingolipids of red blood cells, other cells, and merconium (lacto-N-neotetraose, lacto-N-biose, N-acetyllactosamine, etc.). Furthermore, terminal galactose residues on LOS and elsewhere are decorated with sialic acid, which blocks antibody binding, activation of complement, phagocytosis, and intracellular killing. Therefore, antigenic mimicry of host antigens is an important defense mechanism provided by the oligosaccharide component of the LOS to avoid innate and adaptive host defense mechanisms. However, some strains of H. somni isolated from the bovine genital tract, particularly the normal bovine prepuce, are incapable of LOS phase variation, sialylation of the LOS, and expression of ChoP. At least 1 such strain has been shown to be avirulent, underscoring the importance of the LOS as a virulence factor, although this strain is deficient in other factors as well. The structure and arrangement of the inner core glycoses (heptose and 3-deoxy-D-manno-2-octulosnic acid) is remarkably similar to the inner core oligosaccharide on some strains of Neisseria spp., and mutants that contain a truncated LOS oligosaccharide are considerably more serum-sensitive than the parent strain. Therefore, the LOS is a critical component that enables H. somni to resist host defenses and cause disease.


Asunto(s)
Lipopolisacáridos/fisiología , Pasteurellaceae/patogenicidad , Factores de Virulencia/fisiología , Lipopolisacáridos/química , Lipopolisacáridos/aislamiento & purificación , Ácido N-Acetilneuramínico/metabolismo , Sialiltransferasas/fisiología
13.
Brain Struct Funct ; 221(3): 1591-605, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25596866

RESUMEN

The neural cell adhesion molecule NCAM and its association with the polysialic acid (PSA) are believed to contribute to brain structural plasticity that underlies memory formation. Indeed, the attachment of long chains of PSA to the glycoprotein NCAM down-regulates its adhesive properties by altering cell-cell interactions. In the brain, the biosynthesis of PSA is catalyzed by two polysialyltransferases, which are differentially regulated during lifespan. One of them, ST8SiaIV (PST), is predominantly expressed during adulthood whereas the other one, ST8SiaII (STX), dominates during embryonic and post-natal development. To understand the role played by ST8SiaIV during learning and memory and its underlying hippocampal plasticity, we used knockout mice deleted for the enzyme ST8SiaIV (PST-ko mice). At adult age, PST-ko mice show a drastic reduction of PSA-NCAM expression in the hippocampus and intact hippocampal adult neurogenesis. We found that these mice display impaired long-term but not short-term memory in both, spatial and non-spatial behavioral tasks. Remarkably, memory deficits of PST-ko mice were abolished by exposure to environmental enrichment that was also associated with an increased number of PSA-NCAM expressing new neurons in the dentate gyrus of these mice. Whether the presence of a larger pool of immature, likely plastic, new neurons favored the rescue of long-term memory in PST-ko mice remains to be determined. Our findings add new evidence to the role played by PSA in memory consolidation. They also suggest that PSA synthesized by PST critically controls the tempo of new neurons maturation in the adult hippocampus.


Asunto(s)
Ambiente Controlado , Hipocampo/enzimología , Memoria/fisiología , Moléculas de Adhesión de Célula Nerviosa/fisiología , Sialiltransferasas/fisiología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Neurogénesis , Plasticidad Neuronal , Sialiltransferasas/genética , Memoria Espacial/fisiología
14.
Circ Res ; 114(6): 976-81, 2014 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-24425712

RESUMEN

RATIONALE: Sialylation by α2,3-sialyltransferases has been shown to be a crucial glycosylation step in the generation of functional selectin ligands. Recent evidence suggests that sialylation also affects the binding of chemokines to their corresponding receptor. OBJECTIVE: Because the chemokine receptors for Ccl5 and Ccl2 are important in atherogenic recruitment of neutrophils and monocytes, we here investigated the role of α2,3-sialyltransferase IV (ST3Gal-IV) in Ccl5- and Ccl2-mediated myeloid cell arrest and further studied its relevance in a mouse model of atherosclerosis. METHODS AND RESULTS: St3Gal4-deficient myeloid cells showed a reduced binding of Ccl5 and an impaired Ccl5-triggered integrin activation. Correspondingly, Ccl5-induced arrest on tumor necrosis factor-α-stimulated endothelium was almost completely abrogated, as observed in flow chamber adhesion assays and during ex vivo perfusion or intravital microscopy of carotid arteries. Moreover, Ccl5-triggered neutrophil and monocyte extravasation into the peritoneal cavity was severely reduced in St3Gal4(-/-) mice. In contrast, St3Gal4 deficiency did not significantly affect Ccl2 binding and only marginally decreased Ccl2-induced flow arrest of myeloid cells. In agreement with the crucial role of leukocyte accumulation in atherogenesis, and the importance of Ccl5 chemokine receptors mediating myeloid cell recruitment to atherosclerotic vessels, St3Gal4 deficiency drastically reduced the size, stage, and inflammatory cell content of atherosclerotic lesions in Apoe(-/-) mice on high-fat diet. CONCLUSIONS: In summary, these findings identify ST3Gal-IV as a promising target to reduce inflammatory leukocyte recruitment and arrest.


Asunto(s)
Aterosclerosis/enzimología , Quimiocina CCL5/fisiología , Rodamiento de Leucocito/fisiología , Células Mieloides/patología , Sialiltransferasas/deficiencia , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/patología , Aterosclerosis/prevención & control , Quimiocina CCL2/metabolismo , Grasas de la Dieta/toxicidad , Femenino , Inflamación , Molécula 1 de Adhesión Intercelular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ácido N-Acetilneuramínico/metabolismo , Neuraminidasa/farmacología , Procesamiento Proteico-Postraduccional , Sialiltransferasas/genética , Sialiltransferasas/fisiología , Factor de Necrosis Tumoral alfa/farmacología , Molécula 1 de Adhesión Celular Vascular/metabolismo , beta-Galactosida alfa-2,3-Sialiltransferasa
15.
Mini Rev Med Chem ; 14(14): 1132-8, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25643611

RESUMEN

Molecules that link innate and adaptive immunity and regulate immune response in health and disease are now the focus of many studies. This review discusses CD74 and its ligand macrophage migration inhibitory factor (MIF), and their central position in linking these two components of the immune response by controlling survival of macrophages and B cells.


Asunto(s)
Presentación de Antígeno/fisiología , Antígenos CD/fisiología , Linfocitos B/metabolismo , Supervivencia Celular/fisiología , Homeostasis/fisiología , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Sialiltransferasas/fisiología , Transducción de Señal/fisiología , Animales , Linfocitos B/inmunología , Humanos , Factores Inhibidores de la Migración de Macrófagos/inmunología
16.
Biochem J ; 449(1): 241-51, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23050851

RESUMEN

TGF-ß (transforming growth factor-ß)-induced EMT (epithelial-mesenchymal transition) induces the proliferation and migration of the HLE (human lens epithelial) cells. Ganglioside GM3, simple sialic-acid-containing glycosphingolipids on mammalian cell membranes, regulates various pathological phenomena such as insulin resistance and tumour progression. However, the relationship between ganglioside GM3 and TGF-ß-induced EMT in the HLE B-3 cells is poorly understood. In the present study we demonstrated that ganglioside GM3 was involved in TGF-ß1-induced EMT in HLE B-3 cells. Our results indicated that the expression of ganglioside GM3 and GM3 synthase mRNA were significantly increased in TGF-ß1-induced HLE B-3 cells. Reporter gene analysis also demonstrated that transcriptional activation of the GM3 synthase gene was regulated by Sp1 (specificity protein 1) in HLE B-3 cells upon TGF-ß1 stimulation. Interestingly, the inhibition of ganglioside GM3 expression by d-PDMP [d-threo-1-phenyl-2-decanoylamino-3-morpholino-1-propanol] and GM3 synthase shRNA (short hairpin RNA) resulted significantly in the suppression of cell migration and EMT-related signalling in HLE B-3 cells stimulated by TGF-ß. Furthermore, exogenous treatment of ganglioside GM3 rescued the expression of EMT molecules and cell migration suppressed by the depletion of ganglioside GM3 in TGF-ß1-induced HLE B-3 cells. We also found that ganglioside GM3 interacted with TGFßRs (TGF-ß receptors) in TGF-ß1-induced HLE B-3 cells. Taken together, these results suggest that ganglioside GM3 induced by TGF-ß1 regulates EMT by potential interaction with TGFßRs.


Asunto(s)
Movimiento Celular/fisiología , Transición Epitelial-Mesenquimal/fisiología , Cristalino/citología , Cristalino/metabolismo , Sialiltransferasas/química , Factor de Crecimiento Transformador beta1/fisiología , Secuencia de Bases , Línea Celular , Células Epiteliales/metabolismo , Humanos , Mesodermo/metabolismo , Datos de Secuencia Molecular , Sialiltransferasas/fisiología , Factor de Crecimiento Transformador beta1/química
17.
J Immunol ; 189(7): 3631-40, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22914050

RESUMEN

NADPH oxidase is a multisubunit complex that assembles during phagocytosis to generate reactive oxygen species. Several components of this complex have been implicated in chronic granulomatous disease and Crohn's disease, highlighting the importance of reactive oxygen species in regulating host immune response. In this study, we use genetically deficient mice to elucidate how p40(phox), one subunit of the NADPH oxidase complex, functions during intestinal inflammation. We show that p40(phox) deficiency enhances inflammation in both dextran sulfate sodium-induced and innate immune-mediated murine colitis models. This inflammation is characterized by severe colonic tissue injury, increased proinflammatory cytokines, and increased neutrophil recruitment. We demonstrate that neutrophils are essential during the recovery phase of intestinal inflammation and that p40(phox) expression is necessary for this restitution. Lastly, using an integrative bioinformatic approach, we show that p40(phox) deficiency leads to upregulation of chemokine receptor 1 and downregulation of enzymes involved in glycan modifications, including fucosyltransferases and sialyltransferases, during inflammation. We propose that p40(phox) deficiency enhances intestinal inflammation through the dysregulation of these two pathways in neutrophils.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Infiltración Neutrófila/inmunología , Neutrófilos/inmunología , Neutrófilos/patología , Fosfoproteínas/fisiología , Animales , Colitis/inducido químicamente , Colitis/enzimología , Colitis/inmunología , Sulfato de Dextran , Modelos Animales de Enfermedad , Fucosiltransferasas/antagonistas & inhibidores , Fucosiltransferasas/fisiología , Mucosa Intestinal/enzimología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/fisiología , Infiltración Neutrófila/genética , Neutrófilos/enzimología , Fosfoproteínas/deficiencia , Fosfoproteínas/genética , Sialiltransferasas/antagonistas & inhibidores , Sialiltransferasas/fisiología
18.
Blood ; 120(5): 1015-26, 2012 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-22700726

RESUMEN

Binding of selectins to their glycan ligands is a prerequisite for successful leukocyte trafficking. During synthesis and transport through the secretory pathway, selectin ligands are constructed with the participation of one or more sialyltransferases of the ST3Gal subfamily. Previous studies established that ST3Gal-IV only partially contributes to selectin ligand formation, indicating that other ST3Gal-sialyltransferases are involved. By generating and analyzing St3gal6-null mice and St3gal4/St3gal6 double-deficient mice, in the present study, we found that binding of E- and P-selectin to neutrophils and L-selectin binding to lymph node high endothelial venules is reduced in the absence of ST3Gal-VI and to a greater extent in double-deficient mice. In an ex vivo flow chamber assay, P- and E-selectin-dependent leukocyte rolling was mildly reduced in St3gal6-null mice and more severely in double-deficient mice. In inflamed cremaster muscle venules of St3gal6-null mice, we found impaired P-selectin-dependent, but not E-selectin-dependent leukocyte rolling, whereas in double-deficient mice, E-selectin-dependent rolling was almost completely absent. Furthermore, neutrophil recruitment into the inflamed peritoneal cavity and lymphocyte homing to secondary lymphoid organs were impaired in St3gal6-null mice and more severely in double-deficient mice. The results of the present study demonstrate the coordinated participation of both ST3Gal-VI and ST3Gal-IV in the synthesis of functional selectin ligands.


Asunto(s)
Selectinas/biosíntesis , Sialiltransferasas/fisiología , Animales , Capilares/metabolismo , Capilares/fisiología , Selectina E/metabolismo , Regulación Enzimológica de la Expresión Génica/fisiología , Hemostasis/genética , Rodamiento de Leucocito/genética , Ligandos , Ratones , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Selectina-P/metabolismo , Unión Proteica , Flujo Sanguíneo Regional/genética , Flujo Sanguíneo Regional/fisiología , Selectinas/metabolismo , Sialiltransferasas/genética , Sialiltransferasas/metabolismo , Distribución Tisular , beta-Galactosida alfa-2,3-Sialiltransferasa
19.
Clin Exp Metastasis ; 29(3): 229-38, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22228572

RESUMEN

ST6GalNAcI is a sialyltransferase responsible for the synthesis of sialyl Tn (sTn) antigen which is expressed in a variety of adenocarcinomas including gastric cancer especially in advanced cases, but the roles of ST6GalNAcI and sTn in cancer progression are largely unknown. We generated sTn-expressing human gastric cancer cells by ectopic expression of ST6GalNAcI to evaluate metastatic ability of these cells and prognostic effect of ST6GalNAcI and sTn in a mouse model, and identified sTn carrier proteins to gain insight into the function of ST6GalNAcI and sTn in gastric cancer progression. A green fluorescent protein-tagged human gastric cancer cell line was transfected with ST6GalNAcI to produce sTn-expressing cells, which were transplanted into nude mice. STn-positive gastric cancer cells showed higher intraperitoneal metastatic ability in comparison with sTn-negative control, resulting in shortened survival time of the mice, which was mitigated by anti-sTn antibody administration. Then, sTn-carrying proteins were immunoprecipitated from culture supernatants and lysates of these cells, and identified MUC1 and CD44 as major sTn carriers. It was confirmed that MUC1 carries sTn also in human advanced gastric cancer tissues. Identification of sTn carrier proteins will help understand mechanisms of metastatic phenotype acquisition of gastric cancer cells by ST6GalNAcI and sTn.


Asunto(s)
Antígenos de Carbohidratos Asociados a Tumores/fisiología , Metástasis de la Neoplasia , Sialiltransferasas/fisiología , Neoplasias Gástricas/patología , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Receptores de Hialuranos/fisiología , Masculino , Ratones , Mucina-1/fisiología , Trasplante de Neoplasias , Sialiltransferasas/antagonistas & inhibidores , Transfección , Trasplante Heterólogo
20.
Ann N Y Acad Sci ; 1253: 201-5, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22257379

RESUMEN

The ability of leukocytes to navigate through the different body compartments is an essential component for functioning immune defense and surveillance systems. In order to exit the blood circulation, leukocytes follow distinct recruitment steps, including capture of free-flowing leukocytes to, and rolling along, the vessel wall; firm leukocyte arrest on the endothelial lining; and postarrest modifications (spreading and crawling), which prepare the leukocyte for transmigration through the vascular wall. Post-translational glycosylation (including sialylation) has been known for many years to be functionally relevant for selectin ligands and, hence, selectin-mediated capture and rolling. Recently, sialylation by the α2-3 sialyltransferase ST3Gal-IV was identified to significantly influence chemokine-triggered firm leukocyte arrest, expanding the role of α2-3 sialylation from leukocyte rolling to subsequent chemokine-triggered leukocyte arrest. These findings make ST3Gal-IV an interesting drug target for modulating leukocyte trafficking in human disorders, including autoimmune diseases and cancer.


Asunto(s)
Leucocitos/fisiología , Sialiltransferasas/fisiología , Animales , Movimiento Celular/fisiología , Quimiocinas/fisiología , Glicosilación , Humanos , Rodamiento de Leucocito/fisiología , Ligandos , Ratones , Modelos Biológicos , Selectinas/fisiología , beta-Galactosida alfa-2,3-Sialiltransferasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...