Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
1.
Angiogenesis ; 2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-39222273

RESUMO

The permeability of blood vessels plays a crucial role in the spread of cancer cells, facilitating their metastasis at distant sites. Small extracellular vesicles (sEVs) are known to contribute to the metastasis of various cancers by crossing the blood vessel wall. However, the role of abnormal glycoconjugates on sEVs in tumor blood vessels remains unclear. Our study found elevated levels of fucosyltransferase VII (FUT7) and its product sialyl Lewis X (sLeX) in muscle-invasive bladder cancer (BLCA), with high levels of sLeX promoting the growth and invasion of BLCA cells. Further investigation revealed that sLeX was enriched in sEVs derived from BLCA. sLeX-decorated sEVs increased blood vessel permeability by disrupting the tight junctions of human umbilical vein endothelial cells (HUVECs). Using the glycoproteomics approach, we identified integrin α3 (ITGA3) as a sLeX-bearing glycoprotein in BLCA cells and their sEVs. Mechanically, sLeX modification stabilized ITGA3 by preventing its degradation in lysosomes. sEVs carrying sLeX-modified ITGA3 can be effectively internalized by HUVECs, leading to a decrease in the expression of tight junction protein. Conversely, silencing ITGA3 in sLeX-decorated sEVs restored tight junction proteins and reduced blood vessel permeability by inhibiting the MAPK pathway. Moreover, sLeX-modification of ITGA3 at Asn 265 in HUVECs promoted occludin dephosphorylation at Ser/Thr residues, followed by inducing its importin α1-mediated nuclear translocation, which resulted in the disruption of tight junctions. Our findings suggest a potential strategy for disrupting the formation of a metastatic microenvironment and preventing the spread of malignant bladder cancer.

2.
Bioorg Med Chem ; 98: 117553, 2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-38128297

RESUMO

Neutrophil binding to vascular P- and E-selectin is the rate-limiting step in the recruitment of immune cells to sites of inflammation. Many diseases, including sickle cell anemia, post-myocardial infarction reperfusion injury, and acute respiratory distress syndrome are characterized by dysregulated inflammation. We have recently reported sialyl Lewisx analogues as potent antagonists of P- and E-selectin and demonstrated their in vivo immunosuppressive activity. A key component of these molecules is a tartrate diester that serves as an acyclic tether to orient the fucoside and the galactoside moiety in the required gauche conformation for optimal binding. The next stage of our study involved attaching an extended carbon chain onto one of the esters. This chain could be utilized to tether other pharmacophores, lipids, and contrast agents in the context of enhancing pharmacological applications through the sialyl Lewisx / receptor-mediated mechanism. Herein, we report our preliminary studies to generate a small library of tartrate based sialyl Lewisx analogues bearing extended carbon chains. Anionic charged chemical entities are attached to take advantage of proximal charged amino acids in the carbohydrate recognition domain of the selectin receptors. Starting with a common azido intermediate, synthesized using copper-catalyzed Huisgen 1,3-dipolar cycloadditions, these molecules demonstrate E- and P-selectin binding properties.


Assuntos
Selectina E , Selectina-P , Humanos , Selectina-P/metabolismo , Selectina E/metabolismo , Tartaratos , Antígeno Sialil Lewis X , Oligossacarídeos/química , Sítios de Ligação , Carbono , Inflamação , Adesão Celular
3.
J Virol ; 96(19): e0134422, 2022 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-36125302

RESUMO

Subtype H7 avian influenza A viruses (IAVs) are enzootic in wild aquatic birds and have caused sporadic spillovers into domestic poultry and humans. Here, we determined the distribution of fucosylated α2,3 sialoglycan (i.e., sialyl Lewis X [SLeX]) in chickens and five common dabbling duck species and the association between SLeX and cell/tissue/host tropisms of H7 IAVs. Receptor binding analyses showed that H7 IAVs bind to both α2,3-linked (SA2,3Gal) and α2,6-linked sialic acids (SA2,6Gal), but with a higher preference for SLeX; H7 IAVs replicated more efficiently in SLeX-overexpressed than SLeX-deficient MDCK cells. While chickens and all tested dabbling ducks expressed abundant SA2,3Gal and SA2,6Gal, SLeX was detected in both respiratory and gastrointestinal tissues of chickens and mallard ducks and in only the respiratory tissues of gadwall, green-wing teal, and northern shoveler but not in wood ducks. Viral-tissue binding assays showed that H7 IAVs bind to chicken colon crypt cells that express SLeX but fewer bind to mallard colon crypt cells, which do not express SLeX; H7 IAVs bind efficiently to epithelial cells of all tissues expressing SA2,3Gal. High viral replication was identified in both chickens and mallards infected with an H7 virus, regardless of SLeX expression, and viruses were detected in all cells to the same degree as viruses detected in the viral-tissue binding assays. In summary, this study suggests that SLeX facilitates infection of H7 viruses, but other types of SA2,3Gal glycan receptors shape the tissue/host tropisms of H7 IAVs. IMPORTANCE In addition to causing outbreaks in domestic poultry, subtype H7 IAVs can cause sporadic spillover infections in lower mammals and humans. In this study, we showed that SLeX expression varies among wild dabbling ducks. Although it facilitated virus binding and affected infection of H7 IAV in cells, SLeX expression is not the only determinant of viral replication at either the tissue or host level. This study suggested that access to heterologous SA2,3Gal glycan receptors, including fucosylated α2,3-linked sialoglycans, shape tissue and host tropism of H7 IAVs in aquatic wild birds.


Assuntos
Vírus da Influenza A , Influenza Aviária , Antígeno Sialil Lewis X , Tropismo Viral , Animais , Animais Selvagens/virologia , Galinhas/virologia , Cães , Patos/virologia , Vírus da Influenza A/patogenicidade , Vírus da Influenza A/fisiologia , Células Madin Darby de Rim Canino , Polissacarídeos , Ácidos Siálicos , Antígeno Sialil Lewis X/metabolismo
4.
Int J Mol Sci ; 22(18)2021 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-34576124

RESUMO

Asthma is an allergic disease that causes severe infiltration of leukocytes into the lungs. Leukocyte infiltration is mediated by the binding of sialyl Lewis X (sLex) glycans present on the leukocytes to E-and P-selectins present on the endothelial cells at the sites of inflammation. Here, we found that mouse eosinophils express sLex glycans, and their infiltration into the lungs and proliferation in the bone marrow were significantly suppressed by an anti-sLex monoclonal antibody (mAb) F2 in a murine model of ovalbumin-induced asthma. The percentage of eosinophils in the bronchoalveolar lavage fluid and bone marrow and serum IgE levels decreased significantly in the F2-administered mice. Levels of T helper type 2 (Th2) cytokines and chemokines, involved in IgE class switching and eosinophil proliferation and recruitment, were also decreased in the F2-administered mice. An ex vivo cell rolling assay revealed that sLex glycans mediate the rolling of mouse eosinophils on P-selectin-expressing cells. These results indicate that the mAb F2 exerts therapeutic effects in a murine model of allergen-induced asthma, suggesting that sLex carbohydrate antigen could serve as a novel therapeutic target for allergic asthma.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Asma/tratamento farmacológico , Hipersensibilidade/tratamento farmacológico , Antígeno Sialil Lewis X/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Asma/complicações , Medula Óssea/patologia , Diferenciação Celular , Modelos Animais de Doenças , Eosinófilos/imunologia , Feminino , Hipersensibilidade/complicações , Imunidade , Pulmão/imunologia , Pulmão/patologia , Camundongos Endogâmicos C57BL , Modelos Biológicos , Selectina-P/metabolismo , Ligação Proteica
5.
J Cell Mol Med ; 24(21): 12789-12798, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32985079

RESUMO

In this study, a new water and alkaline-soluble polysaccharide (ALP), with an average molecular weight of 6.63 × 104  Da, was successfully purified from the rhizomes of Atractylodes lancea. GC analysis demonstrated that ALP was a kind of glucan. The effect of the ALP on the interaction between E-selectin and sialyl Lewis X (sLex ) was examined in human osteosarcoma U-2 OS cells. It was obvious that the expression of sLex antigen on the surface of U-2 OS cells was visible under fluorescence microscopy. The addition of ALP (0.5, 1 and 2 mg/mL) resulted in a marked inhibition on the adhesion, migration and invasion of U-2 OS cells to human umbilical vein endothelial cells (HUVECs), which was achieved by the decreased sLex expression on U-2 OS cells. Additionally, the induction of apoptosis can be observed in U-2 OS cells following ALP treatment using TUNEL staining and Annexin V-FITC/PI double-staining analysis on flow cytometry. In conclusion, these results indicated that ALP exerted anti-metastatic activity towards osteosarcoma cells via inhibition of sLex /E-selectin binding, which suggested that ALP could be a potent agent for human osteosarcoma intervention.


Assuntos
Atractylodes/química , Selectina E/metabolismo , Osteossarcoma/patologia , Polissacarídeos/farmacologia , Antígeno Sialil Lewis X/metabolismo , Apoptose/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Movimento Celular/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Monossacarídeos/análise , Metástase Neoplásica , Polissacarídeos/isolamento & purificação , Ligação Proteica/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia
6.
J Biol Chem ; 294(48): 18465-18474, 2019 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-31628196

RESUMO

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.


Assuntos
Selectina E/metabolismo , Glicoproteínas/metabolismo , Receptores de Antígenos Quiméricos/metabolismo , Antígeno Sialil Lewis X/metabolismo , Linfócitos T/metabolismo , Animais , Adesão Celular , Linhagem Celular Tumoral , Células Cultivadas , Fucose/metabolismo , Glicosilação , Humanos , Imunoterapia Adotiva/métodos , Ligantes , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Ligação Proteica , Engenharia de Proteínas/métodos
7.
Glycoconj J ; 37(2): 277-291, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32062824

RESUMO

In the past decades, the roles of carbohydrates in living organisms and their potential use in many fields have been extensively investigated. Sialyl Lewis x (sLex), a member of body carbohydrate, is an inherent blood-type tetrasaccharide on the surface of different cells, the lymphocyte, neutrophil, some T cells, multiple tumor cells and so on. SLex is a common ligand of the three selectins, L-selectin, E-selectin and P-selectin, and plays important roles in multiple physiological phenomenas by interacting with selectins. Under normal physiological conditions, sLex can affect the immune process and fertilization process. Lower expression of sLex could cause leukocyte adhesion defects (LAD) II. Overexpression of sLex on the other hand has been linked to several cancers including melanoma, breast, pancreatic, liver, lung, head and neck, ovarian, bladder carcinomas and some blood disease including Hodgkin disease, some B cell chronic lymphocytic leukemias, acute lymphoblastic leukemias, and most acute nonlymphocytic leukemias. This paper mainly reviews the physiological functions and pathological effects of sLex and its applications in disease diagnosis, drug delivery, gene transfer and medical molecular imaging. We aim to help researchers and other readers quickly grasp the physiological and pathological roles and its medical applications of sLex, and give some suggestions for research directions.


Assuntos
Neoplasias/metabolismo , Selectinas/metabolismo , Antígeno Sialil Lewis X/metabolismo , Animais , Biomarcadores Tumorais/metabolismo , Portadores de Fármacos/metabolismo , Humanos , Neoplasias/diagnóstico , Neoplasias/tratamento farmacológico
8.
Int J Mol Sci ; 21(21)2020 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-33167483

RESUMO

Metastasis is the main cause of death among colorectal cancer (CRC) patients. E-selectin and its carbohydrate ligands, including sialyl Lewis X (sLeX) antigen, are key players in the binding of circulating tumor cells to the endothelium, which is one of the major events leading to organ invasion. Nevertheless, the identity of the glycoprotein scaffolds presenting these glycans in CRC remains unclear. In this study, we firstly have characterized the glycoengineered cell line SW620 transfected with the fucosyltransferase 6 (FUT6) coding for the α1,3-fucosyltransferase 6 (FUT6), which is the main enzyme responsible for the synthesis of sLeX in CRC. The SW620FUT6 cell line expressed high levels of sLeX antigen and E-selectin ligands. Moreover, it displayed increased migration ability. E-selectin ligand glycoproteins were isolated from the SW620FUT6 cell line, identified by mass spectrometry, and validated by flow cytometry and Western blot (WB). The most prominent E-selectin ligand we identified was the neural cell adhesion molecule L1 (L1CAM). Previous studies have shown association of L1CAM with metastasis in cancer, thus the novel role as E-selectin counter-receptor contributes to understand the molecular mechanism involving L1CAM in metastasis formation.


Assuntos
Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Selectina E/metabolismo , Molécula L1 de Adesão de Célula Nervosa/fisiologia , Adesão Celular/genética , Neoplasias do Colo/genética , Humanos , Imunoprecipitação , Ligantes , Molécula L1 de Adesão de Célula Nervosa/genética , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Ligação Proteica/genética , Transfecção , Células Tumorais Cultivadas
9.
Molecules ; 25(12)2020 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-32575485

RESUMO

Selectins belong to a group of adhesion molecules that fulfill an essential role in immune and inflammatory responses and tissue healing. Selectins are glycoproteins that decode the information carried by glycan structures, and non-covalent interactions of selectins with these glycan structures mediate biological processes. The sialylated and fucosylated tetrasaccharide sLex is an essential glycan recognized by selectins. Several glycosyltransferases are responsible for the biosynthesis of the sLex tetrasaccharide. Selectins are involved in a sequence of interactions of circulated leukocytes with endothelial cells in the blood called the adhesion cascade. Recently, it has become evident that cancer cells utilize a similar adhesion cascade to promote metastases. However, like Dr. Jekyll and Mr. Hyde's two faces, selectins also contribute to tissue destruction during some infections and inflammatory diseases. The most prominent function of selectins is associated with the initial stage of the leukocyte adhesion cascade, in which selectin binding enables tethering and rolling. The first adhesive event occurs through specific non-covalent interactions between selectins and their ligands, with glycans functioning as an interface between leukocytes or cancer cells and the endothelium. Targeting these interactions remains a principal strategy aimed at developing new therapies for the treatment of immune and inflammatory disorders and cancer. In this review, we will survey the significant contributions to and the current status of the understanding of the structure of selectins and the role of selectins in various biological processes. The potential of selectins and their ligands as therapeutic targets in chronic and acute inflammatory diseases and cancer will also be discussed. We will emphasize the structural characteristic of selectins and the catalytic mechanisms of glycosyltransferases involved in the biosynthesis of glycan recognition determinants. Furthermore, recent achievements in the synthesis of selectin inhibitors will be reviewed with a focus on the various strategies used for the development of glycosyltransferase inhibitors, including substrate analog inhibitors and transition state analog inhibitors, which are based on knowledge of the catalytic mechanism.


Assuntos
Adesão Celular , Migração e Rolagem de Leucócitos , Leucócitos/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Selectinas/metabolismo , Animais , Humanos , Inflamação/metabolismo , Inflamação/patologia , Leucócitos/patologia , Neoplasias/patologia
10.
J Biol Chem ; 293(19): 7300-7314, 2018 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-29593094

RESUMO

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.


Assuntos
Fucosiltransferases/metabolismo , Isoenzimas/metabolismo , Antígenos CD15/metabolismo , Células-Tronco Mesenquimais/enzimologia , Amino Açúcares/metabolismo , Citometria de Fluxo , Fucosiltransferases/genética , Glicoconjugados/metabolismo , Glicômica , Humanos , Isoenzimas/genética , Antígenos CD15/genética , Espectrometria de Massas , Células-Tronco Mesenquimais/imunologia , RNA Mensageiro/genética , Antígeno Sialil Lewis X
11.
J Virol ; 92(18)2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-29997206

RESUMO

An outbreak of respiratory disease caused by the equine-origin influenza A(H3N8) virus was first detected in dogs in 2004 and since then has been enzootic among dogs. Currently, the molecular mechanisms underlying host adaption of this virus from horses to dogs is unknown. Here, we have applied quantitative binding, growth kinetics, and immunofluorescence analyses to elucidate these mechanisms. Our findings suggest that a substitution of W222L in the hemagglutinin of the equine-origin A(H3N8) virus facilitated its host adaption to dogs. This mutation increased binding avidity of the virus specifically to receptor glycans with N-glycolylneuraminic acid (Neu5Gc) and sialyl Lewis X (SLeX) motifs. We have demonstrated these motifs are abundantly located in the submucosal glands of dog trachea. Our findings also suggest that in addition to the type of glycosidic linkage (e.g., α2,3-linkage or α2,6-linkage), the type of sialic acid (Neu5Gc or 5-N-acetyl neuraminic acid) and the glycan substructure (e.g., SLeX) also play an important role in host tropism of influenza A viruses.IMPORTANCE Influenza A viruses (IAVs) cause a significant burden on human and animal health, and mechanisms for interspecies transmission of IAVs are far from being understood. Findings from this study suggest that an equine-origin A(H3N8) IAV with mutation W222L at its hemagglutinin increased binding to canine-specific receptors with sialyl Lewis X and Neu5Gc motifs and, thereby, may have facilitated viral adaption from horses to dogs. These findings suggest that in addition to the glycosidic linkage (e.g., α2,3-linked and α2,6-linked), the substructure in the receptor saccharides (e.g., sialyl Lewis X and Neu5Gc) could present an interspecies transmission barrier for IAVs and drive viral mutations to overcome such barriers.


Assuntos
Hemaglutininas/genética , Especificidade de Hospedeiro , Vírus da Influenza A Subtipo H3N8/genética , Mutação , Receptores Virais/genética , Animais , Sítios de Ligação , Cães , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Cavalos , Vírus da Influenza A Subtipo H3N8/crescimento & desenvolvimento , Vírus da Influenza A Subtipo H3N8/metabolismo , Cinética , Ácidos Neuramínicos/análise , Oligossacarídeos/análise , Infecções por Orthomyxoviridae/virologia , Ligação Proteica , Receptores Virais/metabolismo , Antígeno Sialil Lewis X , Traqueia/química , Traqueia/virologia , Tropismo Viral , Ligação Viral
12.
Mol Cell Biochem ; 450(1-2): 159-166, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29922946

RESUMO

Sialyl Lewis x (sLex) is a minimal recognition motif for ligands of P-selectin and plays an important role in tumor cell adhesion and migration. Thus, targeting sLex could be an effective method to prevent tumor metastasis. In this study, we aimed to identify a microRNA (miRNA) which is capable to suppress the expression of sLex. MicroRNAs which may target ST3GAL4 were predicted by the online tools. Colo 320 HSR human colon adenocarcinoma cells were employed. The transcriptional and translational levels of ST3GAL4 were evaluated by western blotting and Real-time quantitative polymerase chain reaction. Cell adhesion and spread were assessed with or without hsa-miR-370 treatment. It was shown that hsa-miR-370 inhibited the expression of sLex in colo-320 cells, which repressed the binding of P-selectin, and led to reduced cell attachment and spread. Our results found that P-selectin-induced elevations of p-p38 and p-PI3K levels were significantly inhibited by hsa-miR-370, indicating that repressed sLex level is able to reduce the P-selectin binding and therefore eliminating the P-selectin-induced activation of p38 and PI3K signaling. In conclusion, we found that hsa-miR-370 specifically inhibits the expression of sLex, represses cell adhesion and spreading in colo-320 cells. Our study provides a possible effective treatment against tumor invasion.


Assuntos
Adenocarcinoma/metabolismo , Neoplasias do Colo/metabolismo , Sistema de Sinalização das MAP Quinases , MicroRNAs/metabolismo , Proteínas de Neoplasias/metabolismo , Selectina-P/metabolismo , RNA Neoplásico/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/patologia , Adesão Celular , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Humanos , Antígenos CD15/genética , Antígenos CD15/metabolismo , MicroRNAs/genética , Proteínas de Neoplasias/genética , Selectina-P/genética , RNA Neoplásico/genética , Antígeno Sialil Lewis X
13.
Stem Cells ; 35(4): 1080-1092, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27888602

RESUMO

The clinical effectiveness of systemically administered human mesenchymal stem cells (hMSCs) depends on their capacity to engage vascular endothelium. hMSCs derived from bone marrow (BM-hMSCs) natively lack endothelial binding capacity, but express a CD44 glycovariant containing N-linked sialyllactosamines that can be α(1,3)-fucosylated using fucosyltransferase-VI (FTVI) to enforce sLeX decorations, thereby creating hematopoietic cell E-/L-selectin ligand (HCELL). HCELL expression programs potent shear-resistant adhesion of circulating cells to endothelial beds expressing E-selectin. An alternative source of hMSCs is adipose tissue (A-hMSCs), and we assessed whether A-hMSCs bind E-selectin and/or possess sialyllactosamine-decorated CD44 accessible to α(1,3)-fucosylation. Similar to BM-hMSCs, we found that A-hMSCs natively lack E-selectin ligands, but FTVI-mediated cell surface α(1,3)-fucosylation induces sLeX expression and robust E-selectin binding secondary to conversion of CD44 into HCELL. Moreover, treatment with the α(1,3)-fucosyltransferase-FTVII also generated expression of HCELL on both BM-hMSCs and A-hMSCs, with sLeX decorations created on N-linked glycans of the "standard" CD44 (CD44s) isoform. The finding that hMSCs from both source tissues each lack native E-selectin ligand expression prompted examination of the expression of glycosyltransferases that direct lactosaminyl glycan synthesis. These studies reveal that both types of hMSCs conspicuously lack transcripts encoding α(1,3)-fucosyltransferases, but equally express glycosyltransferases critical to creation of sialyllactosamines. Collectively, these data indicate that assembly of a sialyllactosaminyl-decorated CD44s glycovariant is a conserved feature of hMSCs derived from adipose tissue and marrow, thus identifying a CD44 glycosignature of these cells and supporting the applicability of cell surface α(1,3)-fucosylation in programming migration of systemically administered A-hMSCs to sites of tissue injury/inflammation. Stem Cells 2017;35:1080-1092.


Assuntos
Receptores de Hialuronatos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Tecido Adiposo/citologia , Células da Medula Óssea/citologia , Adesão Celular , Diferenciação Celular , Linhagem Celular , Selectina E/metabolismo , Fibronectinas/metabolismo , Regulação da Expressão Gênica , Glicoproteínas/metabolismo , Glicosilação , Glicosiltransferases/metabolismo , Humanos , Imunofenotipagem , Selectina L/metabolismo , Ligantes , Células-Tronco Mesenquimais/citologia , Neuraminidase/metabolismo , Polissacarídeos/metabolismo , Ligação Proteica
14.
Cytotherapy ; 20(9): 1110-1123, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30170815

RESUMO

BACKGROUND: The regenerative and immunomodulatory properties of human mesenchymal stromal cells (hMSCs) have raised great hope for their use in cell therapy. However, when intravenously infused, hMSCs fail to reach sites of tissue injury. Fucose addition in α(1,3)-linkage to terminal sialyllactosamines on CD44 creates the molecule known as hematopoietic cell E-/L-selectin ligand (HCELL), programming hMSC binding to E-selectin that is expressed on microvascular endothelial cells of bone marrow (BM), skin and at all sites of inflammation. Here we describe how this modification on BM-derived hMSCs (BM-hMSCs) can be adapted to good manufacturing practice (GMP) standards. METHODS: BM-hMSCs were expanded using xenogenic-free media and exofucosylated using α(1,3)-fucosyltransferases VI (FTVI) or VII (FTVII). Enforced fucosylation converted CD44 into HCELL, and HCELL formation was assessed using Western blot, flow cytometry and cell-binding assays. Untreated (unfucosylated), buffer-treated and exofucosylated BM-hMSCs were each analyzed for cell viability, immunophenotype and differentiation potential, and E-selectin binding stability was assessed at room temperature, at 4°C, and after cryopreservation. Cell product safety was evaluated using microbiological testing, karyotype analysis, and c-Myc messenger RNA (mRNA) expression, and potential effects on genetic reprogramming and in cell signaling were analyzed using gene expression microarrays and receptor tyrosine kinase (RTK) phosphorylation arrays. RESULTS: Our protocol efficiently generates HCELL on clinical-scale batches of BM-hMSCs. Exofucosylation yields stable HCELL expression for 48 h at 4°C, with retained expression after cell cryopreservation. Cell viability and identity are unaffected by exofucosylation, without changes in gene expression or RTK phosphorylation. DISCUSSION: The described exofucosylation protocol using xenogenic-free reagents enforces HCELL expression on hMSCs endowing potent E-selectin binding without affecting cell viability or native phenotype. This described protocol is readily scalable for GMP-compliant clinical production.


Assuntos
Biotecnologia/métodos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Biotecnologia/normas , Diferenciação Celular , Sobrevivência Celular , Células Cultivadas , Criopreservação , Selectina E/metabolismo , Células Endoteliais/metabolismo , Fucose/metabolismo , Fucosiltransferases/metabolismo , Glicosilação , Humanos , Receptores de Hialuronatos/metabolismo , Imunofenotipagem , Transcriptoma
15.
BMC Cancer ; 18(1): 495, 2018 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-29716546

RESUMO

BACKGROUND: The E-selectin ligands expressed by cancer cells mediate adhesion of circulating cancer cells to endothelial cells, as well as within tissue microenvironments important for tumor progression and metastasis. The identification of E-selectin ligands within cancer tissue could yield new biomarkers for patient stratification and aid in identifying novel therapeutic targets. The determinants of selectin ligands consist of sialylated tetrasaccharides, the sialyl Lewis X and A (sLeX and sLeA), displayed on protein or lipid scaffolds. Standardized procedures for immunohistochemistry make use of the antibodies against sLeX and/or sLeA. However, antibody binding does not define E-selectin binding activity. METHODS: In this study, we developed an immunohistochemical staining technique, using E-selectin-human Ig Fc chimera (E-Ig) to characterize the expression and localization of E-selectin binding sites on paraffin-embedded sections of different cancer tissue. RESULTS: E-Ig successfully stained cancer cells with high specificity. The E-Ig staining show high reactivity scores in colon and lung adenocarcinoma and moderate reactivity in triple negative breast cancer. Compared with reactivity of antibody against sLeX/A, the E-Ig staining presented higher specificity to cancer tissue with better defined borders and less background. CONCLUSIONS: The E-Ig staining technique allows the qualitative and semi-quantitative analysis of E-selectin binding activity on cancer cells. The development of accurate techniques for detection of selectin ligands may contribute to better diagnostic and better understanding of the molecular basis of tumor progression and metastasis.


Assuntos
Selectina E/metabolismo , Ligantes , Neoplasias/metabolismo , Biomarcadores , Selectina E/genética , Humanos , Imuno-Histoquímica , Neoplasias/genética , Neoplasias/patologia , Inclusão em Parafina
16.
Biochim Biophys Acta ; 1860(8): 1795-808, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26721331

RESUMO

BACKGROUND: Terminal α2-3 and α2-6 sialylation of glycans precludes further chain elongation, leading to the biosynthesis of cancer relevant epitopes such as sialyl-Lewis X (SLe(X)). SLe(X) overexpression is associated with tumor aggressive phenotype and patients' poor prognosis. METHODS: MKN45 gastric carcinoma cells transfected with the sialyltransferase ST3GAL4 were established as a model overexpressing sialylated terminal glycans. We have evaluated at the structural level the glycome and the sialoproteome of this gastric cancer cell line applying liquid chromatography and mass spectrometry. We further validated an identified target expression by proximity ligation assay in gastric tumors. RESULTS: Our results showed that ST3GAL4 overexpression leads to several glycosylation alterations, including reduced O-glycan extension and decreased bisected and increased branched N-glycans. A shift from α2-6 towards α2-3 linked sialylated N-glycans was also observed. Sialoproteomic analysis further identified 47 proteins with significantly increased sialylated N-glycans. These included integrins, insulin receptor, carcinoembryonic antigens and RON receptor tyrosine kinase, which are proteins known to be key players in malignancy. Further analysis of RON confirmed its modification with SLe(X) and the concomitant activation. SLe(X) and RON co-expression was validated in gastric tumors. CONCLUSION: The overexpression of ST3GAL4 interferes with the overall glycophenotype of cancer cells affecting a multitude of key proteins involved in malignancy. Aberrant glycosylation of the RON receptor was shown as an alternative mechanism of oncogenic activation. GENERAL SIGNIFICANCE: This study provides novel targets and points to an integrative tumor glycomic/proteomic-profiling for gastric cancer patients' stratification. This article is part of a Special Issue entitled "Glycans in personalised medicine" Guest Editor: Professor Gordan Lauc.


Assuntos
Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Antígenos CD15/biossíntese , Proteínas de Neoplasias/biossíntese , Polissacarídeos/biossíntese , Receptores Proteína Tirosina Quinases/metabolismo , Neoplasias Gástricas/metabolismo , Glicômica , Humanos , Antígenos CD15/genética , Proteínas de Neoplasias/genética , Polissacarídeos/genética , Receptores Proteína Tirosina Quinases/genética , Antígeno Sialil Lewis X , Sialiltransferases/biossíntese , Sialiltransferases/genética , Neoplasias Gástricas/genética , beta-Galactosídeo alfa-2,3-Sialiltransferase
17.
Cytokine ; 99: 91-98, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28756336

RESUMO

Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease that primarily affect the joints and inflammatory cell migration into inflamed articular sites contribute to this disease. Among the inflammatory cells, human mucosal-associated invariant T (MAIT) cells were recently recognized as critical cellular component with a pathological role in RA. However, their migratory characteristics are poorly understood. The aim of this study was to determine whether human MAIT cells preferentially traffick to inflamed synovial sites in rheumatoid arthritis patients and to elucidate the underlying mechanism. First, we found that TNFα and IL-1ß were elevated in synovial fluid (SF) of RA patients, which resulted in increased expression of E-selectin, ICAM-1 and V-CAM-1 on blood vessel endothelial cells. To understand whether TNFα and IL-1ß in the SF facilitated MAIT cell migration, we analyzed CD161+ TCRα7.2+ MAIT and other CD3+ T cells for differences in migratory capacity. Collectively, our results demonstrate that TNFα and IL-1ß in the SF facilitated MAIT cell migration dependent on expression of selectin ligand, sialyl LewisX (sLeX) and CCR6 on MAIT cells. We also showed that MAIT cells in the SF from RA patients equipped upregulated sLeX compared to the peripheral blood of RA patients and healthy persons, which suggest that TNFα and IL-1ß mediated expression of E-selectin preferentially attract sLeX mediated MAIT cell migration into the SF of RA patients.


Assuntos
Movimento Celular , Interleucina-1beta/farmacologia , Células T Invariantes Associadas à Mucosa/citologia , Líquido Sinovial/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Adulto , Idoso , Artrite Reumatoide/patologia , Movimento Celular/efeitos dos fármacos , Demografia , Selectina E/metabolismo , Feminino , Glicosilação , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Ligantes , Masculino , Pessoa de Meia-Idade , Receptores CCR6/metabolismo , Molécula 1 de Adesão de Célula Vascular/metabolismo
18.
Stem Cells ; 34(10): 2501-2511, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27335219

RESUMO

Human mesenchymal stem cells (MSCs) hold great promise in cellular therapeutics for skeletal diseases but lack expression of E-selectin ligands that direct homing of blood-borne cells to bone marrow. Previously, we described a method to engineer E-selectin ligands on the MSC surface by exofucosylating cells with fucosyltransferase VI (FTVI) and its donor sugar, GDP-Fucose, enforcing transient surface expression of the potent E-selectin ligand HCELL with resultant enhanced osteotropism of intravenously administered cells. Here, we sought to determine whether E-selectin ligands created via FTVI-exofucosylation are distinct in identity and function to those created by FTVI expressed intracellularly. To this end, we introduced synthetic modified mRNA encoding FTVI (FUT6-modRNA) into human MSCs. FTVI-exofucosylation (i.e., extracellular fucosylation) and FUT6-modRNA transfection (i.e., intracellular fucosylation) produced similar peak increases in cell surface E-selectin ligand levels, and shear-based functional assays showed comparable increases in tethering/rolling on human endothelial cells expressing E-selectin. However, biochemical analyses revealed that intracellular fucosylation induced expression of both intracellular and cell surface E-selectin ligands and also induced a more sustained expression of E-selectin ligands compared to extracellular fucosylation. Notably, live imaging studies to assess homing of human MSC to mouse calvarium revealed more osteotropism following intravenous administration of intracellularly-fucosylated cells compared to extracellularly-fucosylated cells. This study represents the first direct analysis of E-selectin ligand expression programmed on human MSCs by FTVI-mediated intracellular versus extracellular fucosylation. The observed differential biologic effects of FTVI activity in these two contexts may yield new strategies for improving the efficacy of human MSCs in clinical applications. Stem Cells 2016;34:2501-2511.


Assuntos
Osso e Ossos/citologia , Movimento Celular , Selectina E/metabolismo , Fucose/metabolismo , Células-Tronco Mesenquimais/citologia , Engenharia Metabólica/métodos , Animais , Medula Óssea/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Espaço Extracelular/metabolismo , Extravasamento de Materiais Terapêuticos e Diagnósticos/patologia , Fucosiltransferases/metabolismo , Glicoproteínas/metabolismo , Glicosilação , Humanos , Espaço Intracelular/metabolismo , Cinética , Ligantes , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Camundongos , Crânio/metabolismo , Transfecção , Transplante Heterólogo
19.
Future Oncol ; 13(2): 145-157, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27646625

RESUMO

AIM: Cervical intraepithelial neoplasia (CIN) is commonly divided into three grades. Guidelines increasingly recommend surgery only in CIN 3 lesions. We investigated markers to evaluate differences in CIN 2 and 3 lesions as well as possible predictors for regression/progression in CIN 2 lesions. MATERIALS & METHODS: Biopsies (n = 128) of healthy cervical tissue and CIN 1-3 were stained for Sialyl Lewis a, Sialyl Lewis x, Lewis y, Gal-3, Gal-7, STMN1 and p16. RESULTS: We observed significant differences between CIN 2 and 3 lesions for Sialyl Lewis a, Sialyl Lewis x, Gal-3, Gal-7, STMN1 and p16. Expression of Sialyl Lewis a was significantly higher in CIN 2 patients who progressed during follow-up. CONCLUSION: Significant differences in marker expression support the differentiation of CIN 2 and 3. Lewis a may help to predict progression/regression in CIN 2 patients.


Assuntos
Biomarcadores Tumorais , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Oligossacarídeos/metabolismo , Estatmina/metabolismo , Displasia do Colo do Útero/metabolismo , Displasia do Colo do Útero/patologia , Antígeno CA-19-9 , Estudos de Casos e Controles , Inibidor p16 de Quinase Dependente de Ciclina/genética , Progressão da Doença , Feminino , Expressão Gênica , Humanos , Imuno-Histoquímica , Gradação de Tumores , Antígeno Sialil Lewis X , Estatmina/genética , Displasia do Colo do Útero/genética
20.
Int J Mol Sci ; 18(7)2017 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-28698503

RESUMO

Expression of the cell adhesion molecule (CAM), Sialyl Lewis X (CD15s) correlates with cancer metastasis, while expression of E-selectin (CD62E) is stimulated by TNF-α. CD15s/CD62E interaction plays a key role in the homing process of circulating leukocytes. We investigated the heterophilic interaction of CD15s and CD62E in brain metastasis-related cancer cell adhesion. CD15s and CD62E were characterised in human brain endothelium (hCMEC/D3), primary non-small cell lung cancer (NSCLC) (COR-L105 and A549) and metastatic NSCLC (SEBTA-001 and NCI-H1299) using immunocytochemistry, Western blotting, flow cytometry and immunohistochemistry in human brain tissue sections. TNF-α (25 pg/mL) stimulated extracellular expression of CD62E while adhesion assays, under both static and physiological flow live-cell conditions, explored the effect of CD15s-mAb immunoblocking on adhesion of cancer cell-brain endothelium. CD15s was faintly expressed on hCMEC/D3, while high levels were observed on primary NSCLC cells with expression highest on metastatic NSCLC cells (p < 0.001). CD62E was highly expressed on hCMEC/D3 cells activated with TNF-α, with lower levels on primary and metastatic NSCLC cells. CD15s and CD62E were expressed on lung metastatic brain biopsies. CD15s/CD62E interaction was localised at adhesion sites of cancer cell-brain endothelium. CD15s immunoblocking significantly decreased cancer cell adhesion to brain endothelium under static and shear stress conditions (p < 0.001), highlighting the role of CD15s-CD62E interaction in brain metastasis.


Assuntos
Neoplasias Encefálicas/etiologia , Neoplasias Encefálicas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Selectina E/metabolismo , Células Endoteliais/metabolismo , Antígenos CD15/metabolismo , Neoplasias Pulmonares/complicações , Neoplasias Pulmonares/metabolismo , Adesão Celular/genética , Adesão Celular/fisiologia , Linhagem Celular , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Selectina E/genética , Humanos , Imuno-Histoquímica , Antígenos CD15/genética , Microscopia Confocal , Metástase Neoplásica/genética , Metástase Neoplásica/fisiopatologia , Resistência ao Cisalhamento/fisiologia , Antígeno Sialil Lewis X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA