Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros











Intervalo de año de publicación
1.
Glycobiology ; 31(11): 1444-1463, 2021 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-34350945

RESUMEN

Bladder cancer is the ninth most frequently diagnosed cancer worldwide, and there is a need to develop new biomarkers for staging and prognosis of this disease. Here we report that cell lines derived from low-grade and high-grade bladder cancers exhibit major differences in expression of glycans in surface glycoproteins. We analyzed protein glycosylation in three low-grade bladder cancer cell lines RT4 (grade-1-2), 5637 (grade-2), and SW780 (grade-1), and three high-grade bladder cancer cell lines J82COT (grade-3), T24 (grade-3) and TCCSUP (grade-4), with primary bladder epithelial cells, A/T/N, serving as a normal bladder cell control. Using a variety of approaches including flow cytometry, immunofluorescence, glycomics and gene expression analysis, we observed that the low-grade bladder cancer cell lines RT4, 5637 and SW780 express high levels of the fucosylated Lewis-X antigen (Lex, CD15) (Galß1-4(Fucα1-3)GlcNAcß1-R), while normal bladder epithelial A/T/N cells lack Lex expression. T24 and TCCSUP cells also lack Lex, whereas J82COT cells express low levels of Lex. Glycomics analyses revealed other major differences in fucosylation and sialylation of N-glycans between these cell types. O-glycans are highly differentiated, as RT4 cells synthesize core 2-based O-glycans that are lacking in the T24 cells. These differences in glycan expression correlated with differences in RNA expression levels of their cognate glycosyltransferases, including α1-3/4-fucosyltransferase genes. These major differences in glycan structures and gene expression profiles between low- and high-grade bladder cancer cells suggest that glycans and glycosyltransferases are candidate biomarkers for grading bladder cancers.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Fucosiltransferasas/metabolismo , Neoplasias de la Vejiga Urinaria/metabolismo , Biomarcadores de Tumor/genética , Células Cultivadas , Fucosiltransferasas/genética , Glicosilación , Humanos , Neoplasias de la Vejiga Urinaria/patología
2.
J Immunol ; 205(7): 1920-1932, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32868410

RESUMEN

Sialyl Lewis X (sLeX) regulates T cell trafficking from the vasculature into skin and sites of inflammation, thereby playing a critical role in immunity. In healthy persons, only a small proportion of human blood T cells express sLeX, and their function is not fully defined. Using a combination of biochemical and functional studies, we find that human blood sLeX+CD4+T cells comprise a subpopulation expressing high levels of Th2 and Th17 cytokines, chemokine receptors CCR4 and CCR6, and the transcription factors GATA-3 and RORγT. Additionally, sLeX+CD4+T cells exclusively contain the regulatory T cell population (CD127lowCD25high and FOXP3+) and characteristically display immune-suppressive molecules, including the coinhibitor receptors PD-1 and CTLA-4. Among CD8+T cells, sLeX expression distinguishes a subset displaying low expression of cytotoxic effector molecules, perforin and granzyme ß, with reduced degranulation and CD57 expression and, consistently, marginal cytolytic capacity after TCR engagement. Furthermore, sLeX+CD8+T cells present a pattern of features consistent with Th cell-like phenotype, including release of pertinent Tc2 cytokines and elevated expression of CD40L. Together, these findings reveal that sLeX display is associated with unique functional specialization of both CD4+ and CD8+T cells and indicate that circulating T cells that are primed to migrate to lesional sites at onset of inflammation are not poised for cytotoxic function.


Asunto(s)
Antígeno Sialil Lewis X/metabolismo , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Tolerancia Central , Citotoxicidad Inmunológica , Factor de Transcripción GATA3/metabolismo , Regulación de la Expresión Génica , Humanos , Tolerancia Inmunológica , Memoria Inmunológica , Activación de Linfocitos , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Tolerancia Periférica , Receptor de Muerte Celular Programada 1/metabolismo , Antígeno Sialil Lewis X/genética
3.
J Biol Chem ; 294(48): 18465-18474, 2019 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-31628196

RESUMEN

Tissue colonization (homing) by blood-borne cells critically hinges on the ability of the cells to adhere to vascular endothelium with sufficient strength to overcome prevailing hemodynamic shear stress. These adhesive interactions are most effectively engendered via binding of the endothelial lectin E-selectin (CD62E) to its cognate ligand, sialyl Lewis-X (sLe X ), displayed on circulating cells. Although chimeric antigen receptor (CAR) T-cell immunotherapy holds promise for treatment of various hematologic and non-hematologic malignancies, there is essentially no information regarding the efficiency of CAR T-cell homing. Accordingly, we performed integrated biochemical studies and adhesion assays to examine the capacity of human CAR T-cells to engage E-selectin. Our data indicate that CAR T-cells do not express sLe X and do not bind E-selectin. However, enforced sLe X display can be achieved on human CAR T-cells by surface fucosylation, with resultant robust E-selectin binding under hemodynamic shear. Importantly, following intravascular administration into mice, fucosylated human CAR-T cells infiltrate marrow with 10-fold higher efficiency than do unfucosylated cells. Collectively, these findings indicate that custom installation of sLe X programs tissue colonization of vascularly administered human CAR T-cells, offering a readily translatable strategy to augment tissue delivery, thereby lowering the pertinent cell dosing and attendant cell production burden, for CAR T-cell immunotherapy applications.


Asunto(s)
Selectina E/metabolismo , Glicoproteínas/metabolismo , Receptores Quiméricos de Antígenos/metabolismo , Antígeno Sialil Lewis X/metabolismo , Linfocitos T/metabolismo , Animales , Adhesión Celular , Línea Celular Tumoral , Células Cultivadas , Fucosa/metabolismo , Glicosilación , Humanos , Inmunoterapia Adoptiva/métodos , Ligandos , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Unión Proteica , Ingeniería de Proteínas/métodos
4.
Nat Cell Biol ; 21(5): 627-639, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30988423

RESUMEN

How disseminated tumour cells engage specific stromal components in distant organs for survival and outgrowth is a critical but poorly understood step of the metastatic cascade. Previous studies have demonstrated the importance of the epithelial-mesenchymal transition in promoting the cancer stem cell properties needed for metastasis initiation, whereas the reverse process of mesenchymal-epithelial transition is required for metastatic outgrowth. Here we report that this paradoxical requirement for the simultaneous induction of both mesenchymal-epithelial transition and cancer stem cell traits in disseminated tumour cells is provided by bone vascular niche E-selectin, whose direct binding to cancer cells promotes bone metastasis by inducing mesenchymal-epithelial transition and activating Wnt signalling. E-selectin binding activity mediated by the α1-3 fucosyltransferases Fut3/Fut6 and Glg1 are instrumental to the formation of bone metastasis. These findings provide unique insights into the functional role of E-selectin as a component of the vascular niche critical for metastatic colonization in bone.


Asunto(s)
Neoplasias Óseas/genética , Selectina E/genética , Fucosiltransferasas/genética , Metástasis de la Neoplasia/genética , Neoplasias/genética , Animales , Neoplasias Óseas/patología , Neoplasias Óseas/secundario , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Transición Epitelial-Mesenquimal/genética , Humanos , Ratones , Metástasis de la Neoplasia/patología , Neoplasias/patología , Células Madre Neoplásicas/patología , Receptores de Factores de Crecimiento de Fibroblastos/genética , Sialoglicoproteínas/genética , Transducción de Señal/genética , Nicho de Células Madre/genética , Activación Transcripcional/genética , Vía de Señalización Wnt/genética , Ensayos Antitumor por Modelo de Xenoinjerto
5.
J Biol Chem ; 293(19): 7300-7314, 2018 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-29593094

RESUMEN

In humans, six α(1,3)-fucosyltransferases (α(1,3)-FTs: FT3/FT4/FT5/FT6/FT7/FT9) reportedly fucosylate terminal lactosaminyl glycans yielding Lewis-X (LeX; CD15) and/or sialyl Lewis-X (sLeX; CD15s), structures that play key functions in cell migration, development, and immunity. Prior studies analyzing α(1,3)-FT specificities utilized either purified and/or recombinant enzymes to modify synthetic substrates under nonphysiological reaction conditions or molecular biology approaches wherein α(1,3)-FTs were expressed in mammalian cell lines, notably excluding investigations using primary human cells. Accordingly, although significant insights into α(1,3)-FT catalytic properties have been obtained, uncertainty persists regarding their human LeX/sLeX biosynthetic range across various glycoconjugates. Here, we undertook a comprehensive evaluation of the lactosaminyl product specificities of intracellularly expressed α(1,3)-FTs using a clinically relevant primary human cell type, mesenchymal stem cells. Cells were transfected with modified mRNA encoding each human α(1,3)-FT, and the resultant α(1,3)-fucosylated lactosaminyl glycoconjugates were analyzed using a combination of flow cytometry and MS. The data show that biosynthesis of sLeX is driven by FTs-3, -5, -6, and -7, with FT6 and FT7 having highest potency. FT4 and FT9 dominantly biosynthesize LeX, and, among all FTs, FT6 holds a unique capacity in creating sLeX and LeX determinants across protein and lipid glycoconjugates. Surprisingly, FT4 does not generate sLeX on glycolipids, and neither FT4, FT6, nor FT9 synthesizes the internally fucosylated sialyllactosamine VIM-2 (CD65s). These results unveil the relevant human lactosaminyl glycans created by human α(1,3)-FTs, providing novel insights on how these isoenzymes stereoselectively shape biosynthesis of vital glycoconjugates, thereby biochemically programming human cell migration and tuning human immunologic and developmental processes.


Asunto(s)
Fucosiltransferasas/metabolismo , Isoenzimas/metabolismo , Antígeno Lewis X/metabolismo , Células Madre Mesenquimatosas/enzimología , Amino Azúcares/metabolismo , Citometría de Flujo , Fucosiltransferasas/genética , Glicoconjugados/metabolismo , Glicómica , Humanos , Isoenzimas/genética , Antígeno Lewis X/genética , Espectrometría de Masas , Células Madre Mesenquimatosas/inmunología , ARN Mensajero/genética , Antígeno Sialil Lewis X
6.
Sci Rep ; 6: 30392, 2016 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-27458028

RESUMEN

There is often interest in dissecting the relative contributions of the N-glycans, O-glycans and glycosphingolipids (GSLs) in regulating complex biological traits like cell signaling, adhesion, development and metastasis. To address this, we developed a CRISPR-Cas9 toolkit to selectively truncate each of these commonly expressed glycan-types. Here, O-glycan biosynthesis was truncated by knocking-out Core 1 ß3Gal-T Specific Molecular Chaperone (COSMC), N-glycans by targeting the ß1,2 GlcNAc-transferase (MGAT1) and GSLs by deleting UDP-glucose ceramide glucosyltransferase (UGCG). These reagents were applied to reveal the glycoconjugates regulating human myeloid cell adhesion to selectins under physiological shear-flow observed during inflammation. These functional studies show that leukocyte rolling on P- and L-selectin is ablated in cells lacking O-glycans, with N-glycan truncation also increasing cell rolling velocity on L-selectin. All three glycan families contributed to E-selectin dependent cell adhesion with N-glycans contributing to all aspects of the leukocyte adhesion cascade, O-glycans only being important during initial recruitment, and GSLs stabilizing slow cell rolling and the transition to firm arrest. Overall, the genome editing tools developed here may be broadly applied in studies of cellular glycosylation.


Asunto(s)
Adhesión Celular , Glicoesfingolípidos/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Leucocitos/metabolismo , Polisacáridos/metabolismo , Sistemas CRISPR-Cas , Línea Celular Tumoral , Glucosiltransferasas/genética , Glucosiltransferasas/metabolismo , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Leucocitos/fisiología , N-Acetilglucosaminiltransferasas/genética , N-Acetilglucosaminiltransferasas/metabolismo , Selectinas/metabolismo
7.
Blood ; 125(4): 687-96, 2015 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-25498912

RESUMEN

The precise glycosyltransferase enzymes that mediate selectin-ligand biosynthesis in human leukocytes are unknown. This knowledge is important because selectin-mediated cell tethering and rolling is a critical component of both normal immune response and various vascular disorders. We evaluated the role of 3 α(2,3)sialyltransferases, ST3Gal-3, -4, and -6, which act on the type II N-Acetyllactosamine structure (Galß1,4GlcNAc) to create sialyl Lewis-X (sLe(X)) and related sialofucosylated glycans on human leukocytes of myeloid lineage. These genes were either silenced using lentiviral short hairpin RNA (shRNA) or functionally ablated using the clustered regularly interspaced short palindromic repeat/Cas9 technology. The results show that ST3Gal-4, but not ST3Gal-3 or -6, is the major sialyltransferase regulating the biosynthesis of E-, P-, and L-selectin ligands in humans. Reduction in ST3Gal-4 activity lowered cell-surface HECA-452 epitope expression by 75% to 95%. Glycomics profiling of knockouts demonstrate an almost complete loss of the sLe(X) epitope on both leukocyte N- and O-glycans. In cell-adhesion studies, ST3Gal-4 knockdown/knockout cells displayed 90% to 100% reduction in tethering and rolling density on all selectins. ST3Gal-4 silencing in neutrophils derived from human CD34(+) hematopoietic stem cells also resulted in 80% to 90% reduction in cell adhesion to all selectins. Overall, a single sialyltransferase regulates selectin-ligand biosynthesis in human leukocytes, unlike mice where multiple enzymes contribute to this function.


Asunto(s)
Selectina E/biosíntesis , Selectina L/biosíntesis , Neutrófilos/metabolismo , Selectina-P/biosíntesis , Sialiltransferasas/biosíntesis , Animales , Células CHO , Adhesión Celular/fisiología , Cricetinae , Cricetulus , Selectina E/genética , Silenciador del Gen , Glicómica , Células HL-60 , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Selectina L/genética , Rodamiento de Leucocito/fisiología , Ratones , Neutrófilos/citología , Selectina-P/genética , Sialiltransferasas/genética , beta-Galactosida alfa-2,3-Sialiltransferasa
8.
Am J Physiol Cell Physiol ; 302(7): C968-78, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22205391

RESUMEN

Lipoxins are formed by leukocytes during cell-cell interactions with epithelial or endothelial cells. Native lipoxin A(4) (LXA(4)) binds to the G protein-coupled lipoxin receptors formyl peptide receptor 2 (FPR2)/ALX and CysLT1. Furthermore, LXA(4) inhibits recruitment of neutrophils, by attenuating chemotaxis, adhesion, and transmigration across vascular endothelial cells. LXA(4) thus appears to serve as an endogenous "stop signal" for immune cell-mediated tissue injury (Serhan CN; Annu Rev Immunol 25: 101-137, 2007). The role of LXA(4) has not been addressed in salivary epithelium, and little is known about its effects on vascular endothelium. Here, we determined that interleukin-1ß (IL-1ß) and tumor necrosis factor-α (TNF-α) receptor activation in vascular endothelium and salivary epithelium upregulated the expression of adhesion molecules that facilitates the binding of immune cells. We hypothesize that the activation of the ALX/FPR2 and/or CysLT1 receptors by LXA(4) decreases this cytokine-mediated upregulation of cell adhesion molecules that enhance lymphocyte binding to both the vascular endothelium and salivary epithelium. In agreement with this hypothesis, we observed that nanomolar concentrations of LXA(4) blocked IL-1ß- and TNF-α-mediated upregulation of E-selectin and intercellular cell adhesion molecule-1 (ICAM-1) on human umbilical vein endothelial cells (HUVECs). Binding of Jurkat cells to stimulated HUVECs was abrogated by LXA(4). Furthermore, LXA(4) preincubation with human submandibular gland cell line (HSG) also blocked TNF-α-mediated upregulation of vascular cell adhesion molecule-1 (VCAM-1) in these cells, and it reduced lymphocyte adhesion. These findings suggest that ALX/FPR2 and/or CysLT1 receptor activation in endothelial and epithelial cells blocks cytokine-induced adhesion molecule expression and consequent binding of lymphocytes, a critical event in the pathogenesis of Sjögren's syndrome (SS).


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Comunicación Celular/fisiología , Endotelio Vascular/efectos de los fármacos , Lipoxinas/farmacología , Glándula Submandibular/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Adhesión Celular/genética , Adhesión Celular/fisiología , Comunicación Celular/genética , Células Cultivadas , Selectina E/genética , Selectina E/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Células Jurkat , Receptores de Formil Péptido/genética , Receptores de Formil Péptido/metabolismo , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/metabolismo , Receptores de Leucotrienos/genética , Receptores de Leucotrienos/metabolismo , Receptores de Lipoxina/genética , Receptores de Lipoxina/metabolismo , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Glándula Submandibular/citología , Glándula Submandibular/inmunología , Glándula Submandibular/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA