Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cancer Res Commun ; 3(3): 347-360, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36875156

RESUMO

Cancer immunotherapy aims to activate the immune system. Some immunotherapeutic agents can be loaded in carrier cells for delivering to the tumors. However, a challenge with cell-based therapies is the selection of the appropriate cells to produce effective clinical outcomes. We hypothesize that therapies based on cells presenting a natural low proinflammatory profile ("silent cells") in the peripheral blood would result in better antitumor responses by increasing their homing to the tumor site. We studied our hypothesis in an immunotherapy model consisting of mesenchymal stromal cells (MSCs) carrying oncolytic adenoviruses for the treatment of immunocompetent mice. Toll-like receptor signaling-deficient cells (TLR4, TLR9, or MyD88 knockout) were used as "silent cells," while regular MSCs were used as control. Although in vitro migration was similar in regular and knockout carrier cells, in vivo tumor homing of silent cells was significantly higher after systemic administration. This better homing to the tumor site was highly related to the mild immune response triggered by these silent cells in peripheral blood. As a result, the use of silent cells significantly improved the antitumor efficacy of the treatment in comparison with the use of regular MSCs. While cancer immunotherapies generally aim to boost local immune responses in the tumor microenvironment, low systemic inflammation after systemic administration of the treatment may indeed enhance their tumor homing and improve the overall antitumor effect. These findings highlight the importance of selecting appropriate donor cells as therapeutic carriers in cell-based therapies for cancer treatment. Significance: Cells carrying drugs, virus, or other antitumor agents are commonly used for the treatment of cancer. This research shows that silent cells are excellent carriers for immunotherapies, improving tumor homing and enhancing the antitumor effect.


Assuntos
Antineoplásicos , Terapia Viral Oncolítica , Animais , Camundongos , Transdução de Sinais , Antineoplásicos/farmacologia , Imunoterapia , Receptores Toll-Like
2.
Cancer Res ; 81(8): 2142-2156, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33593822

RESUMO

The extraordinary plasticity of glioma cells allows them to contribute to different cellular compartments in tumor vessels, reinforcing the vascular architecture. It was recently revealed that targeting glioma-derived pericytes, which represent a big percentage of the mural cell population in aggressive tumors, increases the permeability of the vessels and improves the efficiency of chemotherapy. However, the molecular determinants of this transdifferentiation process have not been elucidated. Here we show that mutations in EGFR stimulate the capacity of glioma cells to function as pericytes in a BMX- (bone marrow and X-linked) and SOX9-dependent manner. Subsequent activation of platelet-derived growth factor receptor beta in the vessel walls of EGFR-mutant gliomas stabilized the vasculature and facilitated the recruitment of immune cells. These changes in the tumor microenvironment conferred a growth advantage to the tumors but also rendered them sensitive to pericyte-targeting molecules such as ibrutinib or sunitinib. In the absence of EGFR mutations, high-grade gliomas were enriched in blood vessels, but showed a highly disrupted blood-brain barrier due to the decreased BMX/SOX9 activation and pericyte coverage, which led to poor oxygenation, necrosis, and hypoxia. Overall, these findings identify EGFR mutations as key regulators of the glioma-to-pericyte transdifferentiation, highlighting the intricate relationship between the tumor cells and their vascular and immune milieu. Our results lay the foundations for a vascular-dependent stratification of gliomas and suggest different therapeutic vulnerabilities determined by the genetic status of EGFR. SIGNIFICANCE: This study identifies the EGFR-related mechanisms that govern the capacity of glioma cells to transdifferentiate into pericytes, regulating the vascular and immune phenotypes of the tumors. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/8/2142/F1.large.jpg.


Assuntos
Neoplasias Encefálicas/irrigação sanguínea , Transdiferenciação Celular , Microambiente Celular , Glioma/irrigação sanguínea , Mutação , Pericitos/fisiologia , Adenina/análogos & derivados , Adenina/farmacologia , Inibidores da Angiogênese/farmacologia , Animais , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Barreira Hematoencefálica/metabolismo , Medula Óssea , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Cromossomos Humanos X , Receptores ErbB/genética , Glioma/imunologia , Glioma/patologia , Humanos , Imunidade Celular , Isocitrato Desidrogenase/genética , Camundongos , Pericitos/efeitos dos fármacos , Pericitos/metabolismo , Piperidinas/farmacologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Fatores de Transcrição SOX9 , Sunitinibe/farmacologia , Hipóxia Tumoral , Microambiente Tumoral
3.
Cancer Gene Ther ; 28(1-2): 64-73, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32457488

RESUMO

There is increasing evidence about the use of oncolytic adenoviruses (Ads) as promising immunotherapy agents. We have previously demonstrated the clinical efficiency of mesenchymal stem cells (MSCs) infected with oncolytic Ads as an antitumoral immunotherapy (called Celyvir) in human and canine patients, using ICOVIR-5 or ICOCAV17 as human and canine oncolytic Ads, respectively. Considering the better clinical outcomes of canine patients, in this study we searched for differences in cellular responses of human and canine MSCs to Ad infection that may help understand the mechanisms leading to higher antitumor immune response. We found that infection of human and canine MSCs with ICOVIR-5 or ICOCAV17 did not activate the NF-κB pathway or the interferon regulatory factors IRF3 and IRF7. However, we observed differences in the profile of cytokines secretion, as infection of canine MSCs with ICOCAV17 resulted in lower secretion of several cytokines. Moreover, we showed that infection of human MSCs with ICOVIR-5 increased the phosphorylation of a number of proteins, including AKT and c-JUN. Finally, we demonstrated that differences in regulation of AKT and c-JUN in human and canine MSCs by ICOVIR-5 or ICOCAV17 are intrinsic to each virus. Our findings suggest that ICOCAV17 induces a more limited host response in canine MSCs, which may be related to a better clinical outcome. This result opens the possibility to develop new human oncolytic Ads with these specific properties. In addition, this improvement could be imitated by selecting specific human MSC on the basis of a limited host response after Ad infection.


Assuntos
Adenoviridae/imunologia , Células-Tronco Mesenquimais/metabolismo , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/imunologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Animais , Cães , Humanos , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/virologia , Proteínas Proto-Oncogênicas c-akt/imunologia , Proteínas Proto-Oncogênicas c-jun/imunologia
4.
Cancers (Basel) ; 12(11)2020 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-33147752

RESUMO

BACKGROUND: Gliomas remain refractory to all attempted treatments, including those using immune checkpoint inhibitors. The characterization of the tumor (immune) microenvironment has been recognized as an important challenge to explain this lack of response and to improve the therapy of glial tumors. METHODS: We designed a prospective analysis of the immune cells of gliomas by flow cytometry. Tumors with or without isocitrate dehydrogenase 1/2 (IDH1/2) mutations were included in the study. The genetic profile and the presence of different molecular and cellular features of the gliomas were analyzed in parallel. The findings were validated in syngeneic mouse models. RESULTS: We observed that few immune cells infiltrate mutant IDH1/2 gliomas whereas the immune content of IDH1/2 wild-type tumors was more heterogeneous. Some of them contained an important immune infiltrate, particularly enriched in myeloid cells with immunosuppressive features, but others were more similar to mutant IDH1/2 gliomas, with few immune cells and a less immunosuppressive profile. Notably, we observed a direct correlation between the percentage of leukocytes and the presence of vascular alterations, which were associated with a reduced expression of Tau, a microtubule-binding protein that controls the formation of tumor vessels in gliomas. Furthermore, overexpression of Tau was able to reduce the immune content in orthotopic allografts of GL261 cells, delaying tumor growth. CONCLUSIONS: We have confirmed the reduced infiltration of immune cells in IDH1/2 mutant gliomas. By contrast, in IDH1/2 wild-type gliomas, we have found a direct correlation between the presence of vascular alterations and the entrance of leukocytes into the tumors. Interestingly, high levels of Tau inversely correlated with the vascular and the immune content of gliomas. Altogether, our results could be exploited for the design of more successful clinical trials with immunomodulatory molecules.

5.
Mol Ther Oncolytics ; 18: 525-534, 2020 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-32995478

RESUMO

Oncolytic immunotherapy with competent viruses is an emerging approach in cancer treatment. The clinical safety of many types of oncolytic viruses (OVs) has been demonstrated. However, there is a lack of information about viral biodistribution in patients. The available data about oncolytic adenovirus biodistribution in human subjects treated intravenously consists of virus detection in body fluids, a few tumor biopsies, and a single report of patient necropsy samples. There is no information about adenoviral biodistribution in patients treated intravenously with cellular vehicles carrying an oncolytic adenovirus. We previously published reports regarding the efficacy and clinical safety of infusing mesenchymal stem cells (MSCs) infected with an OV in human and canine patients. In this study, we performed necropsies on 12 canine patients treated with dCelyvir, canine MSCs infected with ICOCAV17, a canine oncolytic adenovirus. The prevalence of microscopic lesions, especially chronic inflammatory responses in different organs, was higher than expected. Concomitantly, we found a positive immunoreaction to ICOCAV17 in analyzed samples. These findings support a possible role of the virus in development of histopathological alterations and ongoing systemic viral replication of ICOCAV17 in the period after therapy administration.

6.
Oxid Med Cell Longev ; 2019: 9719730, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31467641

RESUMO

Glioblastoma (GBM) is the most common and devastating primary brain tumor. The presence of cancer stem cells (CSCs) has been linked to their therapy resistance. Molecular and cellular components of the tumor microenvironment also play a fundamental role in the aggressiveness of these tumors. In particular, high levels of hypoxia and reactive oxygen species participate in several aspects of GBM biology. Moreover, GBM contains a large number of macrophages, which normally behave as immunosuppressive tumor-supportive cells. In fact, the presence of both, hypoxia and M2-like macrophages, correlates with malignancy and poor prognosis in gliomas. Antioxidant agents, as nutritional supplements, might have antitumor activity. Ocoxin® oral solution (OOS), in particular, has anti-inflammatory and antioxidant properties, as well as antitumor properties in several neoplasia, without known side effects. Here, we describe how OOS affects stem cell properties in certain GBMs, slowing down their tumor growth. In parallel, OOS has a direct effect on macrophage polarization in vitro and in vivo, inhibiting the protumoral features of M2 macrophages. Therefore, OOS could be a feasible candidate to be used in combination therapies during GBM treatment because it can target the highly resilient CSCs as well as their supportive immune microenvironment, without adding toxicity to conventional treatments.


Assuntos
Ácido Ascórbico/uso terapêutico , Glioblastoma/tratamento farmacológico , Macrófagos/metabolismo , Células-Tronco Neoplásicas/metabolismo , Extratos Vegetais/uso terapêutico , Vitamina B 12/uso terapêutico , Vitamina B 6/uso terapêutico , Animais , Ácido Ascórbico/farmacologia , Ácido Fólico , Glioblastoma/patologia , Humanos , Camundongos , Camundongos Nus , Ácido Pantotênico , Extratos Vegetais/farmacologia , Vitamina B 12/farmacologia , Vitamina B 6/farmacologia , Sulfato de Zinco
7.
Cancer Immunol Immunother ; 67(10): 1589-1602, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30066102

RESUMO

Oncolytic virotherapy uses oncolytic viruses that selectively replicate in cancer cells. The use of cellular vehicles with migration ability to tumors has been considered to increase their delivery to target sites. Following this approach, the antitumor efficacy of the treatment Celyvir (mesenchymal stem cells infected with the oncolytic adenovirus ICOVIR-5) has been demonstrated in patients with neuroblastoma. However, the better efficacy of syngeneic or allogeneic mesenchymal stem cells as cell carriers and the specific role of the immune system in this therapy are still unknown. In this study we use our virotherapy Celyvir with syngeneic and allogeneic mouse mesenchymal stem cells to determine their antitumor efficacy in a C57BL/6 murine adenocarcinoma model. Adoptive transfer of splenocytes from treated mice to new tumor-bearing mice followed by a secondary adoptive transfer to a third group was performed. Similar reduction of tumor growth and systemic activation of the innate and adaptive immune system was observed in groups treated with syngeneic or allogeneic mesenchymal stem cells loaded with ICOVIR-5. Moreover, a different pattern of infiltration was observed by immunofluorescence in Celyvir-treated groups. While non-treated tumors presented higher density of infiltrating immune cells in the periphery of the tumor, both syngeneic and allogeneic Celyvir-treated groups presented higher infiltration of CD45+ cells in the core of the tumor. Therefore, these results suggest that syngeneic and allogeneic Celyvir induce systemic activation of the immune system, similar antitumor effect and a higher intratumoral infiltration of leukocytes.


Assuntos
Adenocarcinoma/terapia , Carcinoma Pulmonar de Células não Pequenas/terapia , Neoplasias Pulmonares/terapia , Linfócitos do Interstício Tumoral/imunologia , Células-Tronco Mesenquimais/imunologia , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Adenocarcinoma/genética , Adenocarcinoma/imunologia , Animais , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/imunologia , Feminino , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Células Tumorais Cultivadas
8.
Cancer Res ; 78(17): 4891-4901, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29991502

RESUMO

Dogs with spontaneous tumors treated in veterinary hospitals offer an excellent opportunity for studying immunotherapies, including oncolytic viruses. Oncolytic viruses have advanced into the clinic as an intratumorally administered therapeutic; however, intravenous delivery has been hindered by neutralization in the blood. To circumvent this hurdle, mesenchymal stem cells have been used as a "Trojan horse." Here, we present the treatment of 27 canine patients with cancer with canine mesenchymal stem cells infected with ICOCAV17, a canine oncolytic adenovirus. No significant adverse effects were found. The response rate was 74%, with 14.8% showing complete responses, including total remissions of lung metastasis. We detected virus infection, stromal degeneration, and immune cell infiltration in tumor biopsies after 4 weeks of treatment. The increased presence of antiadenoviral antibodies in the peripheral blood of treated dogs did not appear to prevent the clinical benefit of this therapy. These data indicate that oncolytic viruses loaded in mesenchymal stem cells represent an effective cancer immunotherapy.Significance: The classical clinical limitations of antitumoral viroimmunotherapy can be overcome by use of mesenchymal stem cells.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/17/4891/F1.large.jpg Cancer Res; 78(17); 4891-901. ©2018 AACR.


Assuntos
Doenças do Cão/terapia , Células-Tronco Mesenquimais/metabolismo , Neoplasias/terapia , Terapia Viral Oncolítica , Animais , Doenças do Cão/imunologia , Doenças do Cão/virologia , Cães , Humanos , Imunoterapia , Neoplasias/imunologia , Neoplasias/veterinária , Neoplasias/virologia , Vírus Oncolíticos
9.
Oncotarget ; 8(28): 45415-45431, 2017 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-28525366

RESUMO

Oncolytic virotherapy represents a promising alternative for cancer treatment; however, viral delivery to the tumor represents a major challenge. Mesenchymal stem cells (MSCs) chemotax to tumors, and can serve as a viral delivery tool. Previously, we demonstrated antitumor therapeutic efficacy for mesenchymal stem cells (MSCs) infected with the oncolytic human adenovirus ICOVIR5 (Celyvir) for treatment of neuroblastoma patients. Given the lack of suitable immunocompetent preclinical models, the mechanism underlying Celyvir antitumor activity remains unknown. In this study, we used the syngeneic murine CMT64 cell line as a human adenovirus-semi-permissive tumor model and demonstrate the homing capacity of mouse Celyvir (mCelyvir) to CMT64 tumors. We found that the combined treatment of mCelyvir and intratumoral injections (i.t.) of ICOVIR5 was more effective than treatment with i.t. ICOVIR5 alone. Interestingly, the superior therapeutic effect of the combined therapy was associated with a higher tumor infiltration of CD8+ and CD4+ T cells. Our findings suggest that the use of MSCs as carriers of oncolytic adenovirus can improve the clinical efficacy of anti-cancer virotherapy, not only by driving the adenovirus to tumors, but also through their potential to recruit T cells.


Assuntos
Adenoviridae , Vetores Genéticos , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Terapia Viral Oncolítica , Vírus Oncolíticos , Adenoviridae/genética , Animais , Linhagem Celular Tumoral , Movimento Celular , Sobrevivência Celular/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Expressão Gênica , Genes Reporter , Terapia Genética , Vetores Genéticos/genética , Humanos , Imunoterapia , Mediadores da Inflamação/metabolismo , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Camundongos , Vírus Oncolíticos/genética , Transdução Genética , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Histochem Cell Biol ; 143(5): 517-29, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25417117

RESUMO

Our understanding about medullary compartment, its niches composition and formation is still limited. Previous studies using EphB2 and/or EphB3 knockout mice showed an abnormal thymic development that affects mainly to the epithelial component, including the cortex/medulla distribution, thymic epithelial cell (TEC) morphology and different epithelial-specific marker expression. We have already demonstrated that the lack of ephrinB1 and/or ephrinB2, either on thymocytes or on TECs, alters the cell intermingling processes necessary for thymus organization and affect cortical TEC subpopulations. In the present work, we have used the Cre-LoxP model to selectively delete ephrinB1 and/or ephrinB2 in thymocytes (EfnB1(thy/thy), EfnB2(thy/thy), EfnB1(thy/thy)EfnB2(thy/thy) mice) or TECs (EfnB1(tec/tec), EfnB2(tec/tec), EfnB1(tec/tec)EfnB2(tec/tec) mice) and have analyzed their role on the medullary compartment. In all the studied mutants, medullary areas are smaller and more compact than in the wt thymuses. In most of them, we observe abundant big cysts and a higher proportion of UEA(hi)MTS10(-) cells than in wt mice, which are often forming small cysts. On EfnB1(tec/tec)EfnB2(tec/tec), changes affecting organ size and medullary compartment start at perinatal stage. Our data shed some light on knowledge about wt medulla histological structure and cysts meaning and formation process and on the role played by ephrinB in them.


Assuntos
Efrina-B1/deficiência , Efrina-B2/deficiência , Células Epiteliais/metabolismo , Deleção de Genes , Cisto Mediastínico/metabolismo , Timócitos/metabolismo , Timo/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Efrina-B1/genética , Efrina-B2/genética , Células Epiteliais/patologia , Genótipo , Cisto Mediastínico/genética , Cisto Mediastínico/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tamanho do Órgão , Organogênese , Fenótipo , Nicho de Células-Tronco , Timócitos/patologia , Timo/patologia
11.
Histol Histopathol ; 30(5): 589-99, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25491481

RESUMO

The thymus represents an epithelial microenvironment specialized in the generation of T-cells. The mechanisms or signals that determine the initial differentiation of the two well distinguished histological compartments of the thymus, cortex and medulla, remain unknown. Here, we report a three-dimensional analysis of the distribution of some established thymic epithelial markers in relation to thymic anatomical development during the first steps of thymus organogenesis. In the thymic primordium, initial lumen is lined by claudin (Cld)3/4+K5+ cells, after thymus growth and lobulation they form a continuous branched structure that increases its length and branching degree. Within it, the presence of luminal structures can be distinguished, even at E13.5. The medullary marker mouse thymic stroma 10 (MTS10) is upregulated in these Cld3/4+ lumen forming cells in a proximal-distal sequence. This structural organisation is histologically similar to that described in other epithelial organs undergoing a branching morphogenesis process. These results indicate that the thymic medulla can be evidenced as a continuous branched structure from early stages and suggest a thymic developmental program based on or containing elements of a branching morphogenesis program modified by the presence of lymphoid cells, in which medullary epithelial cell commitment is initially determined by lumen formation.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Imageamento Tridimensional , Microscopia de Fluorescência , Timo/embriologia , Animais , Claudina-3/metabolismo , Claudina-4/metabolismo , Células Epiteliais/citologia , Perfilação da Expressão Gênica , Camundongos , Morfogênese , Linfócitos T/metabolismo , Timócitos/citologia
12.
J Immunol ; 190(6): 2670-81, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23408838

RESUMO

Previous analysis on the thymus of erythropoietin-producing hepatocyte kinases (Eph) B knockout mice and chimeras revealed that Eph-Eph receptor-interacting proteins (ephrins) are expressed both on T cells and thymic epithelial cells (TECs) and play a role in defining the thymus microenvironments. In the current study, we have used the Cre-LoxP system to selectively delete ephrin-B1 and/or ephrin-B2 in either thymocytes (EfnB1(thy/thy), EfnB2(thy/thy), and EfnB1(thy/thy)EfnB2(thy/thy) mice) or TECs (EfnB1(tec/tec), EfnB2(tec/tec), and EfnB1(tec/tec)EfnB2(tec/tec) mice) and determine the relevance of these Eph ligands in T cell differentiation and thymus histology. Our results indicate that ephrin-B1 and ephrin-B2 expressed on thymocytes play an autonomous role in T cell development and, expressed on TECs, their nonautonomous roles are partially overlapping. The effects of the lack of ephrin-B1 and/or ephrin-B2 on either thymocytes or TECs are more severe and specific on thymic epithelium, contribute to the cell intermingling necessary for thymus organization, and affect cortical TEC subpopulation phenotype and location. Moreover, ephrin-B1 and ephrin-B2 seem to be involved in the temporal appearance of distinct cortical TECs subsets defined by different Ly51 levels of expression on the ontogeny.


Assuntos
Comunicação Celular/imunologia , Diferenciação Celular/imunologia , Efrina-B1/fisiologia , Efrina-B2/fisiologia , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Timo/citologia , Timo/imunologia , Animais , Efrina-B1/biossíntese , Efrina-B1/deficiência , Efrina-B2/biossíntese , Efrina-B2/deficiência , Células Epiteliais/citologia , Regulação da Expressão Gênica/imunologia , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Camundongos Transgênicos , Receptor Cross-Talk/imunologia , Timo/anatomia & histologia
13.
Neuroimmunomodulation ; 18(5): 271-80, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21952679

RESUMO

In the present study, we review available information on the relevance of Eph and ephrins in numerous processes occurring in the thymus that regulate not only T cell differentiation but also thymic epithelial cell (TEC) development and organization. Eph/ephrins are a large family of receptors and ligands involved in organogenesis and homeostasis of adult tissues. They are extensively expressed in the thymus and seem to be involved in the colonization of lymphoid progenitor cells and their migration throughout the thymic parenchyma necessary to provide an adequate topological location of developing thymocytes in the epithelial network that ensures their correct differentiation. In addition, EphB2 and EphB3 play a cell-autonomous role in regulating the transitions of double-negative to double-positive cells and of double-positive to single-positive thymocytes and the lack of these molecules or their ligands ephrin B1 and ephrin B2 induces profound alterations of the TEC maturation and in the arrangement of epithelial network. We emphasize that these results are largely reflecting the role played by this family of molecules in controlling thymocyte-TEC interactions within the thymus.


Assuntos
Efrinas/metabolismo , Células Epiteliais/fisiologia , Transdução de Sinais/fisiologia , Timócitos/fisiologia , Timo/metabolismo , Animais , Comunicação Celular , Diferenciação Celular , Movimento Celular , Humanos , Timo/citologia
14.
Immunol Cell Biol ; 89(8): 844-52, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21243004

RESUMO

In order to carry out an in-depth study of the roles of EphB receptors in T-cell development and to determine the specific relevance of forward and reverse signals in the process, we established severe combined immunodeficient (SCID) mice chimeras with wild-type (WT) or EphB-deficient bone marrow cells. The obtained results demonstrate that EphB2 contributes more significantly than EphB3 in the control of CD4(-)CD8(-) (DN)-CD4(+)CD8(+) (DP) progression, and that reverse signals generated in SCID mice receiving EphB2LacZ precursors, which express the EphB2 extracellular domain, partially rescue the blockade of DN cell maturation observed in EphB2-null chimeras. In addition, increased apoptotic DP thymocytes occurring in EphB2 and/or EphB3 SCID chimeras also contribute to the reduced proportions of DP cells. However, EphB2LacZ chimeras do not show any changes in the proportions of apoptotic DP cells, thus suggesting that there is a role for ephrinB reverse signaling in thymocyte survival. The maturation of DP to CD4(+)CD8(-) or CD4(-)CD8(+) seems to need EphB2 forward signaling and EphB3; a fact that was confirmed in reaggregates formed with either EphB2- or EphB3-deficient DP thymocytes and WT thymic epithelial cells (TECs). The DP thymocyte-TEC conjugate formation was also affected by the absence of EphB receptors. Finally, EphB-deficient SCID chimeras show profoundly altered thymic epithelial organization that confirms a significant role for EphB2 and EphB3 receptors in the thymocyte-TEC crosstalk.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Receptores da Família Eph/metabolismo , Transdução de Sinais , Timócitos/fisiologia , Timo/imunologia , Animais , Apoptose , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Efrinas/metabolismo , Linfopoese , Camundongos , Camundongos Knockout , Camundongos SCID , Receptores da Família Eph/deficiência , Receptores da Família Eph/genética , Timócitos/imunologia , Timo/citologia , Quimeras de Transplante
15.
J Leukoc Biol ; 88(3): 483-94, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20504947

RESUMO

The ephrin-Eph ligand receptor pair is known to control the repulsion/adhesion process in different tissues, including the immune system. Herein, we evaluated the role of EphB2 receptors in T cell progenitor migration during in vitro thymus colonization and to ECM or chemokine stimuli. EphB2 and their ligands, ephrin-B1 and ephrin-B2, are expressed in BM-derived progenitors, and EphB2(-/-) cells had diminished thymus colonization capacity. Conversely, EphB2(LacZ) cells, which maintain a preserved ephrin-binding domain, were capable of colonizing WT thymuses similarly to WT progenitors, highlighting the importance of reverse signals transmitted to normal fetal thymus. However, the EphB2 receptor expressed by microenvironmental cells also drives progenitor immigration, as recolonization of EphB2-deficient fetal thymuses was compromised profoundly. Additionally, we observed lower depositions of ECM and chemokines on EphB2-deficient thymuses but no changes in their receptor expression on BM-derived progenitors and developing thymocytes. Migration of EphB2-deficient progenitors and thymocytes was also reduced through ECM or chemokine stimuli. Furthermore, ephrin-B1 costimulation also inhibited haptotaxis and chemotaxis of WT but not EphB2(LacZ) cells, demonstrating the specific involvement of EphB2 signaling on T cell progenitor migration. Our data suggest the relevance of a nonactivated EphB2 for regulating T cell progenitor migration and its modulation upon ephrin-B engagement.


Assuntos
Comunicação Celular/imunologia , Movimento Celular/imunologia , Efrina-B2/metabolismo , Feto/citologia , Células-Tronco/citologia , Linfócitos T/citologia , Timo/embriologia , Animais , Células da Medula Óssea/citologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Membrana Celular/metabolismo , Quimiocinas/metabolismo , Ensaio de Unidades Formadoras de Colônias , Efrina-B1/metabolismo , Efrina-B2/deficiência , Proteínas da Matriz Extracelular/metabolismo , Ligantes , Camundongos , Receptor EphB2/metabolismo , Receptores Fc/metabolismo , Células-Tronco/imunologia , Linfócitos T/imunologia , Timo/citologia , Timo/imunologia
16.
Cell Cycle ; 8(24): 4119-26, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19923894

RESUMO

Bone morphogenetic proteins (BMPs) play a pivotal role during vertebrate embryogenesis and organogenesis, and have also been described to function in regulating cell fate and determination in self-renewing tissues in adults. Recent results have demonstrated that the different components of the BMP2/4 signaling pathway are expressed in the human thymus. In this study, we provide evidence that BMP4 and IL-7 interplay is important in the maintenance of the human thymic progenitor population. Intrathymic CD34(+) cells express BMP receptors (BMPRIA, BMPRIB, ActRIA, BMPRII), signal transduction molecules (Smad1, 5, 8 and 4), and produce BMP4. Neutralization of endogenous BMP4 by treatment with the antagonist Noggin reduces thymic precursor cell survival, and the addition of exogenous BMP4 decreases their proliferation. The treatment of chimeric human-mouse fetal thymus organ cultures with BMP4 inhibits cell expansion, arrests thymocyte differentiation, and leads to the accumulation of human CD34(+) precursor cells. This effect is mainly attributed to the ability of BMP4 to counteract the IL-7-induced proliferation and differentiation of CD34(+) cells. BMP4 downregulates in the precursor cell population the expression of CD127 and inhibits the IL-7-dependent STAT5 phosphorylation. In addition, BMP signaling is promoted by IL-7. Our results also demonstrate that in thymic progenitors BMPs act downstream of Sonic Hedgehog, previously described to function as a maintenance factor for human intrathymic CD34(+) precursor cells.


Assuntos
Proteína Morfogenética Óssea 4/metabolismo , Interleucina-7/metabolismo , Células-Tronco/metabolismo , Timo/metabolismo , Antígenos CD34/metabolismo , Proteína Morfogenética Óssea 4/genética , Receptores de Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Transporte/farmacologia , Diferenciação Celular/fisiologia , Proliferação de Células , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Pré-Escolar , Quimera , Proteínas Hedgehog/metabolismo , Humanos , Lactente , Recém-Nascido , Interleucina-7/genética , Subunidade alfa de Receptor de Interleucina-7/metabolismo , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais/fisiologia , Proteínas Smad/genética , Proteínas Smad/metabolismo , Células-Tronco/citologia , Linfócitos T/citologia , Linfócitos T/metabolismo , Timo/citologia
17.
Eur J Immunol ; 39(10): 2916-24, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19731361

RESUMO

The role of EphB2 and EphB3 in the organization of thymic epithelial cells has been studied in EphB-deficient fetal thymus lobes grafted under the kidney capsule of WT mice. The deficient lobes, as compared with WT ones, showed altered distribution of medullary areas, shortening of medullary epithelial cell processes and presence of K5(-)K8(-) areas. EphB2 and EphB3 expressed on thymic epithelial cells play an autonomous role in their organization. The relevance of Eph/ephrinB forward and reverse signals for this process was evaluated in grafted fetal thymus lobes from mice expressing a truncated EphB2 receptor capable of activating reverse, but not forward, signaling. These deficient lobes showed important alterations of the thymic epithelial organization as compared with the grafted WT lobes, but a less severe phenotype than the grafted EphB2-deficient thymus lobes, which confirms the relevance of EphB2 forward signal for the thymic epithelial organization but, also, a role of the reverse signaling in determining the final epithelial phenotype.


Assuntos
Células Epiteliais/patologia , Epitélio/crescimento & desenvolvimento , Receptor EphB2/fisiologia , Receptor EphB3/fisiologia , Timo/crescimento & desenvolvimento , Animais , Comunicação Celular/genética , Diferenciação Celular/genética , Quimera/genética , Células Epiteliais/metabolismo , Epitélio/patologia , Feminino , Transplante de Tecido Fetal , Queratina-15 , Queratina-5/metabolismo , Queratina-8/metabolismo , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Camundongos SCID , Modelos Biológicos , Deleção de Sequência/genética , Linfócitos T/patologia , Timo/patologia , Timo/transplante
18.
Int J Dev Biol ; 53(7): 971-82, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19598115

RESUMO

In the current study, we extend our own previous results on the thymocyte phenotype of EphB2 and/or EphB3 deficient mice by analyzing the phenotype and the histological organization of their thymic epithelial stroma. All studied adult EphB-deficient thymi showed profound alterations with respect to the wild-type (WT) ones. Each mutant exhibited a specific phenotype, but also showed common features including occurrence of K5+K8+MTS10+ immature medullary epithelial cells, numerous K5-K8-MTS20+ cells and K5+K8+ cells in the thymic cortex and cortical and medullary K5-K8- areas devoid of epithelial cell markers. In addition, comparative analysis of WT and EphB-deficient embryonic and newborn thymi demonstrated that the observed adult phenotype was a consequence of the gradual accumulation of early phenotypic and morphological defects, becoming more severe at the end of embryonic life and in newborn animals. Together, these results confirm a role for EphB2 and EphB3 in thymus morphogenesis. The obtained data are discussed from the point of view of the recognized role played by these two Ephs in the homeostasis of other epithelia and their possible relationships with molecules known to be involved in thymic epithelial cell development.


Assuntos
Receptor EphB2/fisiologia , Receptor EphB3/fisiologia , Timo/embriologia , Timo/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Epitélio/anormalidades , Epitélio/embriologia , Epitélio/crescimento & desenvolvimento , Epitélio/fisiologia , Feminino , Antígenos de Histocompatibilidade Classe II/metabolismo , Queratina-15 , Queratina-5/metabolismo , Queratina-8/metabolismo , Laminina/metabolismo , Camundongos , Camundongos Knockout , Morfogênese/genética , Morfogênese/fisiologia , Fenótipo , Gravidez , Receptor EphB2/deficiência , Receptor EphB2/genética , Receptor EphB3/deficiência , Receptor EphB3/genética , Transdução de Sinais , Timo/anormalidades , Timo/fisiologia
19.
Ann N Y Acad Sci ; 1153: 14-9, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19236323

RESUMO

Eph receptors and their ligands, ephrins, are molecules involved in the morphogenesis of numerous tissues, including the central nervous system in which they play a key role in determining cell positioning and tissue domains containing or excluding nerve fibers. Because common features have been suggested to occur in the microenvironmental organization of brain and thymus, a highly compartmentalized organ central for T cell differentiation, we examined the expression and possible role of Eph/ephrins in the biology of the thymus gland. We reviewed numerous in vivo and in vitro results that confirm a role for Eph and ephrins in the maturation of the thymic epithelial cell (TEC) network and T cell differentiation. Their possible involvement in different steps of early thymus organogenesis, including thymus primordium branching, lymphoid colonization, and thymocyte-TEC interactions, that determine the organization of a mature three-dimensional thymic epithelial network is also analyzed.


Assuntos
Timo/embriologia , Timo/metabolismo , Animais , Diferenciação Celular , Efrinas/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Humanos , Receptores da Família Eph/metabolismo , Transdução de Sinais , Timo/citologia
20.
Immunology ; 125(1): 131-43, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18397270

RESUMO

In the present study, we have analysed the phenotype of EphB2 and/or EphB3 deficient thymocytes confirming and extending previous studies on the role of this family of molecules in T-cell differentiation. In all mutant thymuses statistically significant reduced cell contents were observed. This reduction of thymic cellularity correlated with increased proportions of apoptotic cells, largely both double negative (DN; CD4- CD8-) and double positive (CD4+ CD8+) cells, and decreased proportions of DN cycling cells. Adult deficient thymuses also showed increased proportions of DN cells but not significant variations in the percentages of other thymocyte subsets. In absolute terms, the thymocyte number decreased significantly in all thymocyte compartments from the DN3 (CD44- CD25+) cell stage onward, without variations in the numbers of both DN1 (CD44+ CD25-) and DN2 (CD44+ CD25+) cells. Remarkably, all these changes also occurred from the 15-day fetal EphB2 and/or EphB3 deficient mice, suggesting that adult phenotype results from the gradual accumulations of defects appearing early in the thymus ontogeny. As a reflection of thymus condition, a reduction in the number of T lymphocytes occurred in the peripheral blood and mesenteric lymph nodes, but not in spleen, maintaining the proportions of T-cell subsets defined by CD4/CD8 marker expression, in all cases.


Assuntos
Receptores da Família Eph/deficiência , Subpopulações de Linfócitos T/imunologia , Timo/imunologia , Animais , Apoptose/imunologia , Ciclo Celular/imunologia , Diferenciação Celular/imunologia , Efrinas/metabolismo , Imunofenotipagem , Contagem de Linfócitos , Camundongos , Camundongos Knockout , Receptor EphB2/metabolismo , Receptor EphB3/metabolismo , Receptores da Família Eph/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Timo/crescimento & desenvolvimento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA