Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
PLoS One ; 15(12): e0244762, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33378413

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) is associated with obesity and type 2 diabetes and is characterized by the accumulation of fat in the liver (steatosis). NAFLD can transition into non-alcoholic steatohepatitis (NASH), with liver cell injury, inflammation, and an increased risk of fibrosis. We previously found that injections of either 1866, a synthetic ligand for the lectin receptor CD209, or DANA, a sialidase inhibitor, can inhibit inflammation and fibrosis in multiple animal models. The methionine and choline-deficient (MCD) diet is a model of NASH which results in the rapid induction of liver steatosis and inflammation. In this report, we show that for C57BL/6 mice on a MCD diet, injections of both 1866 and DANA reversed MCD diet-induced decreases in white fat, decreases in adipocyte size, and white fat inflammation. However, these effects were not observed in type 2 diabetic db/db mice on a MCD diet. In db/db mice on a MCD diet, 1866 decreased liver steatosis, but these effects were not observed in C57BL/6 mice. There was no correlation between the ability of 1866 or DANA to affect steatosis and the effects of these compounds on the density of liver macrophage cells expressing CLEC4F, CD64, F4/80, or Mac2. Together these results indicate that 1866 and DANA modulate adipocyte size and adipose tissue macrophage populations, that 1866 could be useful for modulating steatosis, and that changes in the local density of 4 different liver macrophages cell types do not correlate with effects on liver steatosis.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Moléculas de Adhesión Celular/agonistas , Lectinas Tipo C/agonistas , Hígado/efectos de los fármacos , Macrófagos/efectos de los fármacos , Ácido N-Acetilneuramínico/análogos & derivados , Neuraminidasa/antagonistas & inhibidores , Receptores de Superficie Celular/agonistas , Tejido Adiposo/metabolismo , Animales , Deficiencia de Colina/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animales de Enfermedad , Hígado Graso/tratamiento farmacológico , Hígado Graso/metabolismo , Inflamación/metabolismo , Hígado/metabolismo , Macrófagos/metabolismo , Ratones , Ácido N-Acetilneuramínico/farmacología , Ácido N-Acetilneuramínico/uso terapéutico , Neuraminidasa/metabolismo , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/metabolismo
2.
Nanomedicine ; 24: 102053, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31344502

RESUMEN

Here, we report various therapeutic cargo-loadable DNA nanostructures that are shelled in polydopamine and noncovalently tethered with cancer cell-targeting DNA aptamers. Initial DNA nanostructure was formed by rolling-circle amplification and condensation with Mu peptides. This DNA nanostructure was loaded with an antisense oligonucleotide, a photosensitizer, or an anticancer chemotherapeutic drug. Each therapeutic agent-loaded DNA nanostructure was then shelled with polydopamine (PDA), and noncovalently decorated with a poly adenine-tailed nucleic acid aptamer (PA) specific for PTK7 receptor, resulting in PA-tethered and PDA-shelled DNA nanostructure (PA/PDN). PDA coating shell enabled photothermal therapy. In the cells overexpressing PTK7 receptor, photosensitizer-loaded PA/PDN showed greater photodynamic activity. Doxorubicin-loaded PA/PDN exerted higher anticancer activity than the other groups. Antisense oligonucleotide-loaded PA/PDN provided selective reduction of target proteins compared with other groups. Our results suggest that the PA-tethered and PDA-shelled DNA nanostructures could enable the specific receptor-targeted phototherapy, chemotherapy, and gene therapy against cancer cells.


Asunto(s)
Aptámeros de Nucleótidos , Terapia Genética , Hipertermia Inducida , Neoplasias , Fototerapia , Aptámeros de Nucleótidos/química , Aptámeros de Nucleótidos/farmacología , Moléculas de Adhesión Celular/agonistas , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Humanos , Nanoestructuras/química , Nanoestructuras/uso terapéutico , Proteínas de Neoplasias/agonistas , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/terapia , Proteínas Tirosina Quinasas Receptoras/agonistas , Proteínas Tirosina Quinasas Receptoras/metabolismo
3.
Immunology ; 156(1): 74-85, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30220083

RESUMEN

B7 family members and their receptors play a central role in the regulation of T-cell responses through T-cell co-stimulation and co-inhibition pathways that constitute attractive targets for the development of immunotherapeutic drugs. In this study, we report that VSIG-3/IGSF11 is a ligand of B7 family member VISTA/PD-1H and inhibits human T-cell functions through a novel VSIG-3/VISTA pathway. An extensive functional ELISA binding screening assay reveals that VSIG-3 binds to the new B7 family member VISTA but does not interact with other known members of the B7 family. Under the same experimental conditions, we did not observe any significant interaction between VSIG-8 and VISTA. In addition, VSIG-3 inhibits human T-cell proliferation in the presence of T-cell receptor signaling. Furthermore, VSIG-3 significantly reduces cytokine and chemokine production by human T cells including IFN-γ, IL-2, IL-17, CCL5/Rantes, CCL3/MIP-1α, and CXCL11/I-TAC. Anti-VISTA neutralization antibodies attenuate the binding of VSIG-3 and VISTA, as well as VSIG-3-induced T-cell inhibition. Hence, we have identified a novel ligand for VISTA that is able to inhibit human T-cell proliferation and cytokine production. This unique VSIG-3/VISTA co-inhibitory pathway may provide new strategies for the treatment of human cancers, autoimmune disorders, infection, and transplant rejection and may aid in the design of better vaccines.


Asunto(s)
Antígenos B7/agonistas , Moléculas de Adhesión Celular/agonistas , Linfocitos T/inmunología , Anticuerpos Neutralizantes/farmacología , Antígenos B7/genética , Moléculas de Adhesión Celular/genética , Proliferación Celular , Células Cultivadas , Citocinas/metabolismo , Humanos , Tolerancia Inmunológica , Inmunoglobulinas/genética , Inmunoterapia/tendencias , Ligandos , Activación de Linfocitos , ARN Interferente Pequeño/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal
4.
EBioMedicine ; 29: 112-127, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29433983

RESUMEN

Epidermal growth factor receptor (EGFR)-mutation-positive non-smallcell lung cancer (NSCLC) is incurable, despite high rates of response to EGFR tyrosine kinase inhibitors (TKIs). We investigated receptor tyrosine kinases (RTKs), Src family kinases and focal adhesion kinase (FAK) as genetic modifiers of innate resistance in EGFR-mutation-positive NSCLC. We performed gene expression analysis in two cohorts (Cohort 1 and Cohort 2) of EGFR-mutation-positive NSCLC patients treated with EGFR TKI. We evaluated the efficacy of gefitinib or osimertinib with the Src/FAK/Janus kinase 2 (JAK2) inhibitor, TPX0005 in vitro and in vivo. In Cohort 1, CUB domain-containing protein-1 (CDCP1) was an independent negative prognostic factor for progression-free survival (hazard ratio of 1.79, p=0.0407) and overall survival (hazard ratio of 2.23, p=0.0192). A two-gene model based on AXL and CDCP1 expression was strongly associated with the clinical outcome to EGFR TKIs, in both cohorts of patients. Our preclinical experiments revealed that several RTKs and non-RTKs, were up-regulated at baseline or after treatment with gefitinib or osimertinib. TPX-0005 plus EGFR TKI suppressed expression and activation of RTKs and downstream signaling intermediates. Co-expression of CDCP1 and AXL is often observed in EGFR-mutation-positive tumors, limiting the efficacy of EGFR TKIs. Co-treatment with EGFR TKI and TPX-0005 warrants testing.


Asunto(s)
Antígenos CD/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Moléculas de Adhesión Celular/genética , Receptores ErbB/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidad , Mutación , Proteínas de Neoplasias/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antígenos de Neoplasias , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Moléculas de Adhesión Celular/agonistas , Supervivencia Celular , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos , Activación Enzimática , Receptores ErbB/antagonistas & inhibidores , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/patología , Masculino , Ratones , Persona de Mediana Edad , Modelos Biológicos , Proteínas de Neoplasias/agonistas , Proteómica/métodos , Proteínas Proto-Oncogénicas/agonistas , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas Tirosina Quinasas Receptoras/agonistas , Análisis de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto , Tirosina Quinasa del Receptor Axl
5.
Mol Med Rep ; 17(3): 4049-4056, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29286115

RESUMEN

Numerous microRNAs (miRNAs) are aberrantly expressed in glioma, and implicated in glioma occurrence and development. Therefore, the development of miRNAs as potential therapeutic targets for the treatment of patients with glioma has been proposed. miR­379 has been shown to be aberrantly expressed in the progression of malignant tumours. However, the expression, biological functions and mechanism of miR­379 in glioma are yet to be fully understood. Hence, the present study aimed to detect miR­379 expression, investigate its functional relevance and explore its associated molecular mechanism in glioma. In this study, miR­379 expression was significantly downregulated in glioma tissues and cell lines. Enforced miR­379 expression markedly suppressed the cell proliferation and invasion of glioma. Metadherin (MTDH) was identified as a direct target of miR­379 in glioma. The miR­379 expression and MTDH mRNA levels exhibited an inverse association in glioma tissues. The restoration of the MTDH expression partially rescued the inhibitory effects of miR­379 overexpression on glioma cell proliferation and invasion, and the upregulation of miR­379 inhibited the activation of phosphatase and tensin homolog (PTEN)/AKT serine/threonine kinase (AKT) signaling pathway. Overall, these findings demonstrated that miR­379 may play tumour­suppressing roles in glioma through downregulation of MTDH and regulation of the PTEN/AKT signaling pathway, suggesting that miR­379 might be a possible target for the treatment of patients with this malignancy.


Asunto(s)
Neoplasias Encefálicas/genética , Moléculas de Adhesión Celular/genética , Regulación Neoplásica de la Expresión Génica , Glioma/genética , MicroARNs/genética , Fosfohidrolasa PTEN/genética , Proteínas Proto-Oncogénicas c-akt/genética , Antagomirs/genética , Antagomirs/metabolismo , Secuencia de Bases , Sitios de Unión , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/cirugía , Moléculas de Adhesión Celular/agonistas , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Ensayos de Migración Celular , Movimiento Celular , Proliferación Celular , Genes Reporteros , Glioma/metabolismo , Glioma/patología , Glioma/cirugía , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Proteínas de la Membrana , MicroARNs/agonistas , MicroARNs/antagonistas & inhibidores , MicroARNs/metabolismo , Oligorribonucleótidos/genética , Oligorribonucleótidos/metabolismo , Fosfohidrolasa PTEN/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN , Transducción de Señal
6.
J Biol Regul Homeost Agents ; 31(1): 77-80, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28337873

RESUMEN

Cyclosporine-A induces gingival overgrowth with disturbance in the homeostasis of cells and connective tissue proteins. Human gingival fibroblasts were cultured with cyclosporine A, and the expression of two vital endoplasmic stress markers and two prime matrix proteins (connective tissue growth factor (CTGF and periostin) were assessed by RT-PCR. We found that expression of Glucose-Regulated Protein 78 (GRP78/BIP) and CCAAT/enhancer binding protein, C/EBP homologous protein (CHOP) were significantly increased, along with CTGF and periostin, suggesting a role for these factors in gingival overgrowth.


Asunto(s)
Moléculas de Adhesión Celular/genética , Factor de Crecimiento del Tejido Conjuntivo/genética , Ciclosporina/efectos adversos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Proteínas de Choque Térmico/genética , Inmunosupresores/efectos adversos , Factor de Transcripción CHOP/genética , Biomarcadores/metabolismo , Moléculas de Adhesión Celular/agonistas , Moléculas de Adhesión Celular/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/agonistas , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/genética , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Encía/citología , Encía/efectos de los fármacos , Encía/metabolismo , Proteínas de Choque Térmico/agonistas , Proteínas de Choque Térmico/metabolismo , Humanos , Cultivo Primario de Células , Transducción de Señal , Factor de Transcripción CHOP/agonistas , Factor de Transcripción CHOP/metabolismo
7.
FEBS J ; 282(20): 3971-85, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26236947

RESUMEN

Recent studies have highlighted the involvement of metadherin (MTDH), an oncogenic protein, in promoting cancer progression, metastasis and chemoresistance in many cancers including mammary carcinomas. However, the molecular regulation of MTDH is still not completely understood. In this study we document that AMP activated protein kinase (AMPK) activation-induced anti-proliferative effects are, in part, mediated by inhibiting MTDH expression in MDA-MB-231 and BT-549 triple negative breast cancer (TNBC) cells. 5-Aminoimidazole-4-carboxamide ribonucleotide (AICAR), an AMPK activator, caused growth arrest, inhibition of migration and invasion of TNBC cells. Intriguingly, AICAR or metformin treatment resulted in significant downregulation of MTDH expression via inhibiting c-Myc expression. In contrast, treatment of cells with compound C, an inhibitor of AMPK, increased both c-Myc and MTDH expressions in TNBC cells. Also, AMPK activation caused increased glycogen synthase kinase 3ß (GSK3ß) activity by inhibiting the inactive phosphorylation at Ser9, on the one hand, and activation of sirtuin1 (SIRT1) by inhibiting Ser47 phosphorylation, as evidenced by deacetylation of p53, on the other hand. Moreover, AMPK-induced GSK3ß and SIRT1 activities were found to be responsible for inhibiting c-Myc-mediated upregulation of MTDH, as LiCl (an inhibitor of GSK3ß) and EX-527 (an inhibitor of SIRT1) reversed AICAR-mediated downregulation of c-Myc and MTDH expressions. Similar results were observed with siSIRT1 treatment. Furthermore, AICAR and EX-527 treatments caused increased cell death under MTDH-depleted conditions. Finally, we uncovered a novel regulation of MTDH expression and showed that AMPK activation by inducing GSK3ß and SIRT1 downregulates MTDH expression via inhibiting c-Myc in TNBC cells.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Carcinoma/metabolismo , Moléculas de Adhesión Celular/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica , Glucógeno Sintasa Quinasa 3/metabolismo , Sirtuina 1/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/química , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/enzimología , Adenocarcinoma/metabolismo , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/metabolismo , Carcinoma/tratamiento farmacológico , Carcinoma/enzimología , Moléculas de Adhesión Celular/agonistas , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/química , Glucógeno Sintasa Quinasa 3 beta , Humanos , Hipoglucemiantes/farmacología , Proteínas de la Membrana , Proteínas de Neoplasias/agonistas , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Interferencia de ARN , Proteínas de Unión al ARN , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/química , Sirtuina 1/genética , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/enzimología
9.
Int Immunol ; 25(4): 221-33, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23291968

RESUMEN

Dendritic cells (DCs) are antigen-presenting cells efficient in capturing pathogens, and processing their antigenic determinants for presentation to antigen-specific T cells to induce robust immune responses. Their location at peripheral tissues and the expression of pattern-recognition receptors, among them DC-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN), facilitates the capture of pathogens before spreading. However, some pathogens have developed strategies to escape the immune system. One of the most successful is HIV-1, which targets DC-SIGN for transport to the lymph node where the virus infects CD4(+) T cells. Contact of HIV-1 with DC-SIGN is thus the first event in the pathogenic cascade and, therefore, it is the primary target point for therapies aimed at HIV infection prevention. DC-SIGN recognizes specific glycans on HIV-1 and this interaction can be blocked by competitive inhibition through glycans. Although the affinity of glycans is relatively low, multivalency may increase avidity and the strength to compete with HIV-1 virions. We have designed multivalent dendrimeric compounds based on Lewis-type antigens that bind DC-SIGN with high selectivity and avidity and that effectively block gp120 binding to DC-SIGN and, consequently, HIV transmission to CD4(+) T cells. Binding to DC-SIGN and gp120 inhibition was higher on glycodendrimers with larger molecular diameter, indicating that the geometry of the compounds is an important factor determining their functionality. Our compounds elicited DC-SIGN internalization, a property of the receptor upon triggering, but did not affect the maturation status of DCs. Thus, Le(X) glycodendrimers could be incorporated into topic prophylactic approaches for the prevention of HIV-1 transmission.


Asunto(s)
Moléculas de Adhesión Celular/inmunología , Dendrímeros/química , Células Dendríticas/inmunología , Infecciones por VIH/prevención & control , Infecciones por VIH/transmisión , VIH-1/inmunología , Lectinas Tipo C/inmunología , Antígenos del Grupo Sanguíneo de Lewis/química , Polisacáridos/farmacología , Receptores de Superficie Celular/inmunología , Unión Competitiva , Moléculas de Adhesión Celular/agonistas , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Células Dendríticas/efectos de los fármacos , Células Dendríticas/virología , Proteína gp120 de Envoltorio del VIH/metabolismo , Humanos , Evasión Inmune , Lectinas Tipo C/agonistas , Terapia Molecular Dirigida , Polisacáridos/agonistas , Polisacáridos/síntesis química , Receptores de Superficie Celular/agonistas , Relación Estructura-Actividad , Internalización del Virus/efectos de los fármacos
10.
ACS Chem Biol ; 7(9): 1603-8, 2012 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-22747463

RESUMEN

An understanding of the biological roles of lectins will be advanced by ligands that can inhibit or even recruit lectin function. To this end, glycomimetics, noncarbohydrate ligands that function analogously to endogenous carbohydrates, are being sought. The advantage of having such ligands is illustrated by the many roles of the protein DC-SIGN. DC-SIGN is a C-type lectin displayed on dendritic cells, where it binds to mannosides and fucosides to mediate interactions with other host cells or bacterial or viral pathogens. DC-SIGN engagement can modulate host immune responses (e.g., suppress autoimmunity) or benefit pathogens (e.g., promote HIV dissemination). DC-SIGN can bind to glycoconjugates, internalize glycosylated cargo for antigen processing, and transduce signals. DC-SIGN ligands can serve as inhibitors as well as probes of the lectin's function, so they are especially valuable for elucidating and controlling DC-SIGN's roles in immunity. We previously reported a small molecule that embodies key features of the carbohydrates that bind DC-SIGN. Here, we demonstrate that this noncarbohydrate ligand acts as a true glycomimetic. Using NMR HSQC experiments, we found that the compound mimics saccharide ligands: It occupies the same carbohydrate-binding site and interacts with the same amino acid residues on DC-SIGN. The glycomimetic also is functional. It had been shown previously to antagonize DC-SIGN function, but here we use it to generate DC-SIGN agonists. Specifically, appending this glycomimetic to a protein scaffold affords a conjugate that elicits key cellular signaling responses. Thus, the glycomimetic can give rise to functional glycoprotein surrogates that elicit lectin-mediated signaling.


Asunto(s)
Carbohidratos/química , Moléculas de Adhesión Celular/agonistas , Moléculas de Adhesión Celular/antagonistas & inhibidores , Glicoproteínas/química , Lectinas Tipo C/agonistas , Lectinas Tipo C/antagonistas & inhibidores , Receptores de Superficie Celular/agonistas , Receptores de Superficie Celular/antagonistas & inhibidores , Sitios de Unión , Moléculas de Adhesión Celular/metabolismo , Línea Celular , Humanos , Lectinas Tipo C/metabolismo , Ligandos , Receptores de Superficie Celular/metabolismo , Transducción de Señal/efectos de los fármacos
11.
BMB Rep ; 45(3): 159-64, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22449702

RESUMEN

CD99 is known to be involved in the regulation of cell-cell adhesion. However, it remains unclear whether CD99 controls cell-extracellular matrix adhesion. In this study, the effects of CD99 activation on cell-extracellular matrix adhesion were investigated. It was found that engagement of CD99 with the stimulating antibody YG32 downregulated the adhesion of MCF-7 cells to fibronectin, laminin and collagen IV in a dose-dependent manner. The CD99 effect on cell-ECM adhesion was inhibited by overexpression of the dominant negative form of CD99 or CD99 siRNA transfection. Treatment of cells with Mn(2+) or by ß(1) integrin-stimulating antibody restored the inhibitory effect of CD99 on cell-ECM adhesion. Cross-linking CD99 inactivated ß(1) integrin through conformational change. CD99 activation caused dephosphorylation at Tyr-397 in FAK, which was restored by the ß(1) stimulating antibody. Taken together, these results provide the first evidence that CD99 inhibits cell-extracellular matrix adhesion by suppressing ß(1) integrin affinity. [BMB reports 2012; 45(3): 159-164].


Asunto(s)
Antígenos CD/metabolismo , Moléculas de Adhesión Celular/agonistas , Moléculas de Adhesión Celular/metabolismo , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Integrina beta1/metabolismo , Antígeno 12E7 , Adhesión Celular/efectos de los fármacos , Humanos , Células MCF-7 , Células Tumorales Cultivadas
12.
Immunology ; 128(1 Suppl): e816-25, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19740343

RESUMEN

Interleukin-27 (IL-27) is a new IL-12-related heterodimeric cytokine comprising a novel p28 molecule and the Epstein-Barr-virus-induced gene 3 (EBI3) molecules. It augments initiation of T helper type 1-mediated immunity by enhancing the proliferation and cytokine production of T cells. In this study, we examined whether a secreted form of IL-27 subunits would inhibit IL-27-mediated immunological responses. COS-7 cells transduced with the mouse (m) p28 gene secreted a monomeric mp28 protein; however, those transduced with the mEBI3 gene did not detect a mEBI3 protein in the culture supernatants. The secreted mp28 prevented the IL-27-mediated signal transduction and activator of transcription 1 phosphorylation and subsequently inhibited the IL-27-mediated intercellular adhesion molecule-1 induction and interferon-gamma production in CD4(+) T cells. We generated mp28-expressing murine carcinoma Colon 26 cells and inoculated a mixture of the mp28- and mIL-27-expressing Colon 26 cells into syngeneic BALB/c mice. Simultaneous production of mp28 and mIL-27 from Colon 26 cells suppressed IL-27-mediated anti-tumour effects in the mice. We examined the p28-mediated immune suppression by inoculating mp28-expressing myoblasts into allogeneic mice. Forced production of mp28 suppressed the allogeneic cytotoxic T-lymphocyte induction and subsequently retarded the graft rejection. Furthermore, production of both mp28 and mp40, which inhibits the functions of IL-12 and IL-23, prolonged the graft survival longer than the grafts expressing either mp28 or mp40. We propose that p28 can be a regulatory subunit for IL-27-mediated cellular immune responses and a possible therapeutic agent to suppress unfavourable immune responses.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Supervivencia de Injerto/inmunología , Interleucina-17/inmunología , Neoplasias/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Células COS , Moléculas de Adhesión Celular/agonistas , Moléculas de Adhesión Celular/inmunología , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Chlorocebus aethiops , Femenino , Interferón gamma/biosíntesis , Interferón gamma/efectos de los fármacos , Interferón gamma/inmunología , Interleucina-17/genética , Interleucina-17/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Antígenos de Histocompatibilidad Menor , Mioblastos/inmunología , Mioblastos/metabolismo , Fosforilación , Subunidades de Proteína/genética , Subunidades de Proteína/inmunología , Subunidades de Proteína/farmacología , Receptores de Citocinas/genética , Receptores de Citocinas/inmunología , Receptores de Interleucina-12/agonistas , Receptores de Interleucina-12/inmunología , Receptores de Interleucina-12/metabolismo , Factor de Transcripción STAT1/agonistas , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT1/metabolismo , Linfocitos T Citotóxicos/metabolismo , Células TH1/inmunología , Células TH1/metabolismo , Transducción Genética
13.
Expert Opin Emerg Drugs ; 10(2): 299-310, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15934868

RESUMEN

The development of specific targeted therapies, such as anti-TNF-alpha treatment, for chronic inflammatory disorders such as rheumatoid arthritis, has significantly improved treatment, although not all patients respond. Targeting cellular adhesion molecules and chemokines/chemokine receptors as regulators of the extravasation and migration of leukocytes may provide a novel approach for the treatment of these diseases. Moreover, the possibility of developing small-molecule antagonists offers an excellent method for the oral delivery of compounds with a short half-life.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Moléculas de Adhesión Celular/metabolismo , Quimiocinas/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Receptores de Quimiocina/metabolismo , Animales , Antirreumáticos/administración & dosificación , Antirreumáticos/metabolismo , Artritis Reumatoide/metabolismo , Moléculas de Adhesión Celular/agonistas , Moléculas de Adhesión Celular/antagonistas & inhibidores , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Quimiocinas/agonistas , Quimiocinas/antagonistas & inhibidores , Ensayos Clínicos como Asunto/estadística & datos numéricos , Humanos , Receptores de Quimiocina/agonistas , Receptores de Quimiocina/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA