Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Biol Rep ; 51(1): 467, 2024 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-38551765

RESUMO

BACKGROUND: Osteosarcoma (OS) stands out as the most common bone tumor, with approximately 20% of the patients receiving a diagnosis of metastatic OS at their initial assessment. A significant challenge lies in the frequent existence of undetected metastases during the initial diagnosis. Mesenchymal stem cells (MSCs) possess unique abilities that facilitate tumor growth, and their interaction with OS cells is crucial for metastatic spread. METHODS AND RESULTS: We demonstrated that, in vitro, MSCs exhibited a heightened migration response toward the secretome of non-metastatic OS cells. When challenged to a secretome derived from lungs preloaded with OS cells, MSCs exhibited greater migration toward lungs colonized with metastatic OS cells. Moreover, in vivo, MSCs displayed preferential migratory and homing behavior toward lungs colonized by metastatic OS cells. Metastatic OS cells, in turn, demonstrated an increased migratory response to the MSCs' secretome. This behavior was associated with heightened cathepsin D (CTSD) expression and the release of active metalloproteinase 2 (MMP2) by metastatic OS cells. CONCLUSIONS: Our assessment focused on two complementary tumor capabilities crucial to metastatic spread, emphasizing the significance of inherent cell features. The findings underscore the pivotal role of signaling integration within the niche, with a complex interplay of migratory responses among established OS cells in the lungs, prometastatic OS cells in the primary tumor, and circulating MSCs. Pulmonary metastases continue to be a significant factor contributing to OS mortality. Understanding these mechanisms and identifying differentially expressed genes is essential for pinpointing markers and targets to manage metastatic spread and improve outcomes for patients with OS.


Assuntos
Neoplasias Ósseas , Osteossarcoma , Animais , Humanos , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Proliferação de Células/genética , Pulmão/metabolismo , Osteossarcoma/genética , Osteossarcoma/patologia , Células Estromais/patologia , Neoplasias Ósseas/metabolismo , Linhagem Celular Tumoral , Microambiente Tumoral
2.
Apoptosis ; 26(7-8): 447-459, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34024019

RESUMO

Osteosarcoma (OS) is the most frequent malignant bone tumor, affecting predominantly children. Metastases represent a major clinical challenge and an estimated 80% would present undetectable micrometastases at diagnosis. The identification of metastatic traits and molecules would impact in micrometastasis management. We demonstrated that OS LM7 metastatic cells secretome was able to induce microvascular endothelium cell rearrangements, an angiogenic-related trait. A proteomic analysis indicated a gain in angiogenic-related pathways in these cells, as compared to their parental-non-metastatic OS SAOS2 cells counterpart. Further, factors with proangiogenic functions like VEGF and PDGF were upregulated in LM7 cells. However, no differential angiogenic response was induced by LM7 cells in vivo. Regulation of the Fas-FasL axis is key for OS cells to colonize the lungs in this model. Analysis of the proteomic data with emphasis in apoptosis pathways and related processes revealed that the percentage of genes associated with those, presented similar levels in SAOS2 and LM7 cells. Further, the balance of expression levels of proteins with pro- and antiapoptotic functions in both cell types was subtle. Interestingly and of relevance to the model, Fas associated Factor 1 (FAF1), which participates in Fas signaling, was present in LM7 cells and was not detected in SAOS2 cells. The subtle differences in apoptosis-related events and molecules, together with the reported cell-survival functions of the identified angiogenic factors and the increased survival features that we observed in LM7 cells, suggest that the gain in angiogenesis-related pathways in metastatic OS cells would relate to a prosurvival switch rather to an angiogenic switch as an advantage feature to colonize the lungs. OS metastatic cells also displayed higher adhesion towards microvascular endothelium cells suggesting an advantage for tissue colonization. A gain in angiogenesis pathways and molecules does not result in major angiogenic potential. Together, our results suggest that metastatic OS cells would elicit signaling associated to a prosurvival phenotype, allowing homing into the hostile site for metastasis. During the gain of metastatic traits process, cell populations displaying higher adhesive ability to microvascular endothelium, negative regulation of the Fas-FasL axis in the lung parenchyma and a prosurvival switch, would be selected. This opens a new scenario where antiangiogenic treatments would affect cell survival rather than angiogenesis, and provides a molecular panel of expression that may help in distinguishing OS cells with different metastatic potential.


Assuntos
Neoplasias Ósseas , Neoplasias Pulmonares , Osteossarcoma , Proteínas Adaptadoras de Transdução de Sinal , Apoptose , Proteínas Reguladoras de Apoptose , Neoplasias Ósseas/genética , Linhagem Celular Tumoral , Sobrevivência Celular , Humanos , Neoplasias Pulmonares/genética , Osteossarcoma/genética , Proteômica , Secretoma , Regulação para Cima
3.
Biochim Biophys Acta Gen Subj ; 1864(4): 129522, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31945406

RESUMO

BACKGROUND: Osteosarcoma (OS) is the most frequent malignant bone tumor, affecting predominantly children and young adults. Metastases are a major clinical challenge in OS. In this context, 20% of OS patients are diagnosed with metastatic OS, but near 80% of all OS patients could present non-detectable micrometastases at the moment of diagnosis. METHODS: Osteogenic differentiation; doxorubicin exclusion assay; fluorescence microscopy; RT-qPCR; proteomic analysis. RESULTS: Our results suggest that metastatic OS cells possess a diminished osteoblastic differentiation potential with a gain of metastatic traits like the capacity to modify intracellular localization of chemodrugs and higher levels of expression of stemness-related genes. On the opposite hand, non-metastatic OS cells possess bone-associated traits like higher osteoblastic differentiation and also an osteoblastic-inducer secretome. OS cells also differ in the nature of their interaction with mesenchymal stem cells (MSCs), with opposites impacts on MSCs phenotype and behavior. CONCLUSIONS: All this suggests that a major trait acquired by metastatic cells is a switch into a stem-like state that could favor its survival in the pulmonary niche, opening new possibilities for personalized chemotherapeutic schemes. GENERAL SIGNIFICANCE: Our work provides new insights regarding differences among metastatic and non-metastatic OS cells, with particular emphasis on differentiation potential, multidrug resistance and interaction with MSCs.


Assuntos
Neoplasias Ósseas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Osteossarcoma/metabolismo , Antibióticos Antineoplásicos/farmacologia , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/secundário , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Doxorrubicina/farmacologia , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/patologia , Osteossarcoma/tratamento farmacológico , Osteossarcoma/secundário , Fenótipo , Relação Estrutura-Atividade , Células Tumorais Cultivadas
4.
Int J Mol Sci ; 20(8)2019 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-31010037

RESUMO

Mesenchymal stem cells (MSCs) represent an interesting population due to their capacity to release a variety of cytokines, chemokines, and growth factors, and due to their motile nature and homing ability. MSCs can be isolated from different sources, like adipose tissue or bone marrow, and have the capacity to differentiate, both in vivo and in vitro, into adipocytes, chondrocytes, and osteoblasts, making them even more interesting in the regenerative medicine field. Tumor associated stroma has been recognized as a key element in tumor progression, necessary for the biological success of the tumor, and MSCs represent a functionally fundamental part of this associated stroma. Exosomes represent one of the dominant signaling pathways within the tumor microenvironment. Their biology raises high interest, with implications in different biological processes involved in cancer progression, such as the formation of the pre-metastatic niche. This is critical during the metastatic cascade, given that it is the formation of a permissive context that would allow metastatic tumor cells survival within the new environment. In this context, we explored the role of exosomes, particularly MSCs-derived exosomes as direct or indirect modulators. All this points out a possible new tool useful for designing better treatment and detection strategies for metastatic progression, including the management of chemoresistance.


Assuntos
Exossomos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Metástase Neoplásica/patologia , Animais , Humanos , Tropismo , Microambiente Tumoral
5.
Oncotarget ; 9(93): 36585-36602, 2018 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-30564299

RESUMO

Hyaluronan, the main glycosaminoglycan of extracellular matrices, is concentrated in tissues with high cell proliferation and migration rates. In cancer, hyaluronan expression is altered and it becomes fragmented into low-molecular-weight forms, affecting mechanisms associated with cell proliferation, invasion, angiogenesis and multidrug resistance. Here, we analyzed the effect of low-molecular-weight hyaluronan on the response of T lymphoma, osteosarcoma, and mammary adenocarcinoma cell lines to the antineoplastic drug doxorubicin, and whether co-treatment with hyaluronan and doxorubicin modified the behavior of endothelial cells. Our aim was to associate the hyaluronan-doxorubicin response with angiogenic alterations in these tumors. After hyaluronan and doxorubicin co-treatment, hyaluronan altered drug accumulation and modulated the expression of ATP-binding cassette transporters in T-cell lymphoma cells. In contrast, no changes in drug accumulation were observed in cells from solid tumors, indicating that hyaluronan might not affect drug efflux. However, when we evaluated the effect on angiogenic mechanisms, the supernatant from tumor cells treated with doxorubicin exhibited a pro-angiogenic effect on endothelial cells. Hyaluronan-doxorubicin co-treatment increased migration and vessel formation in endothelial cells. This effect was independent of vascular endothelial growth factor but related to fibroblast growth factor-2 expression. Besides, we observed a pro-angiogenic effect on endothelial cells during hyaluronan and doxorubicin co-treatment in the in vivo murine model of T-cell lymphoma. Our results demonstrate for the first time that hyaluronan is a potential modulator of doxorubicin response by mechanisms that involve not only drug efflux but also angiogenic processes, providing an adverse tumor stroma during chemotherapy.

6.
Stem Cells Dev ; 21(14): 2689-702, 2012 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-22462538

RESUMO

The homing properties of mesenchymal stromal cells (MSCs) toward tumors turn them into attractive tools for combining cell and gene therapy. The aim of this study was to select in a feasible way a human bone marrow-derived MSC subpopulation that might exhibit a selective ability to target the tumor mass. Using differential in vitro adhesive capacities during cells isolation, we selected a specific MSC subpopulation (termed MO-MSCs) that exhibited enhanced multipotent capacity and increased cell surface expression of specific integrins (integrins α2, α3, and α5), which correlated with an enhanced MO-MSCs adhesiveness toward their specific ligands. Moreover, MO-MSCs exhibited a higher migration toward conditioned media from different cancer cell lines and fresh human breast cancer samples in the presence or not of a human microendothelium monolayer. Further in vivo studies demonstrated increased tumor homing of MO-MSCs toward established 578T and MD-MBA-231 breast cancer and A375N melanoma tumor xenografts. Tumor penetration by MO-MSCs was highly dependent on metallopeptidases production as it was inhibited by the specific inhibitor 1,10 phenantroline. Finally, systemically administered MO-MSCs preloaded with an oncolytic adenovirus significantly inhibited tumor growth in mice harboring established A375N melanomas, overcoming the natural resistance of the tumor to in situ administration of the oncolytic adenovirus. In summary, this work characterizes a novel MSC subpopulation with increased tumor homing capacity that can be used to transport therapeutic compounds.


Assuntos
Adenoviridae/metabolismo , Melanoma/terapia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Terapia Viral Oncolítica/métodos , Adenoviridae/genética , Animais , Antineoplásicos/uso terapêutico , Adesão Celular , Técnicas de Cultura de Células , Diferenciação Celular , Linhagem Celular Tumoral , Movimento Celular , Condrogênese , Meios de Cultivo Condicionados , Humanos , Cadeias alfa de Integrinas/metabolismo , Melanoma/patologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Nus , Vírus Oncolíticos/genética , Vírus Oncolíticos/metabolismo , Fenantrolinas/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
7.
Mol Pharm ; 8(5): 1538-48, 2011 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-21770423

RESUMO

Hepatocellular carcinoma (HCC) is the fifth most common cancer and the third cause of cancer-related death. Fibrogenesis is an active process characterized by the production of several proinflammatory cytokines, chemokines and growth factors. It involves the activation of hepatic stellate cells (HSCs) which accumulate at the site of injury and are the main source of the extracellular matrix deposits. There are no curative treatments for advanced HCC, thus, new therapies are urgently needed. Mesenchymal stromal cells (MSCs) have the ability to migrate to sites of injury or to remodeling tissues after in vivo administration; however, in several cancer models they demonstrated limited efficacy to eradicate experimental tumors partially due to poor engraftment. Thus, the aim of this work was to analyze the capacity of human MSCs (hMSCs) to migrate and anchor to HCC tumors. We observed that HCC and HSCs, but not nontumoral stroma, produce factors that induce hMSC migration in vitro. Conditioned media (CM) generated from established HCC cell lines were found to induce higher levels of hMSC migration than CM derived from fresh patient tumor samples. In addition, after exposure to CM from HCC cells or HSCs, hMSCs demonstrated adhesion and invasion capability to endothelial cells, type IV collagen and fibrinogen. Consistently, these cells were found to increase metalloproteinase-2 activity. In vivo studies with subcutaneous and orthotopic HCC models indicated that intravenously infused hMSCs migrated to lungs, spleen and liver. Seven days post-hMSC infusion cells were located also in the tumor in both models, but the signal intensity was significantly higher in orthotopic than in subcutaneous model. Interestingly, when orthotopic HCC tumors where established in noncirrhotic or cirrhotic livers, the amount of hMSCs localized in the liver was higher in comparison with healthy animals. A very low signal was found in lungs and spleens, indicating that liver tumors are able to recruit them at high efficiency. Taken together our results indicate that HCC and HSC cells produce factors that efficiently induce hMSC migration toward tumor microenvironment in vitro and in vivo and make MSCs candidates for cell-based therapeutic strategies to hepatocellular carcinoma associated with fibrosis.


Assuntos
Células da Medula Óssea/metabolismo , Carcinoma Hepatocelular/patologia , Movimento Celular , Cirrose Hepática/metabolismo , Neoplasias Hepáticas/patologia , Células-Tronco Mesenquimais/patologia , Microambiente Tumoral , Animais , Células da Medula Óssea/patologia , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/fisiopatologia , Carcinoma Hepatocelular/terapia , Adesão Celular , Linhagem Celular , Linhagem Celular Tumoral , Meios de Cultivo Condicionados , Endotélio Vascular/metabolismo , Células Estreladas do Fígado/metabolismo , Células Estreladas do Fígado/patologia , Humanos , Cirrose Hepática/etiologia , Cirrose Hepática/patologia , Cirrose Hepática/terapia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/fisiopatologia , Neoplasias Hepáticas/terapia , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Nus , Proteínas de Neoplasias/metabolismo , Esferoides Celulares/metabolismo , Esferoides Celulares/patologia , Células Tumorais Cultivadas , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Cancer ; 110(7): 1568-77, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17694551

RESUMO

BACKGROUND: Ewing sarcoma (ES) is a highly vascular malignancy. It has been demonstrated that both angiogenesis and vasculogenesis contribute to the growth of ES tumors. Granulocyte-colony-stimulating factor (G-CSF), a cytokine known to stimulate bone marrow (BM) stem cell production and angiogenesis, is routinely administered to ES patients after chemotherapy. Whether ES cells and patient tumor samples express G-CSF and its receptor (G-CSFR) and whether treatment with this factor enhances tumor growth was examined. METHODS: Human ES cell lines were analyzed for expression of G-CSF and G-CSFR in vitro and in vivo. Sixty-eight paraffin-embedded and 15 frozen tumor specimens from patients with ES were also evaluated for the presence of G-CSF and G-CSFR. The in vivo effect of G-CSF on angiogenesis and BM cell migration was determined. Using a TC/7-1 human ES mouse model, the effect of G-CSF administration on ES tumors was investigated. RESULTS: G-CSF and G-CSFR protein and RNA expression was identified in all ES cell lines and patient samples analyzed. In addition, G-CSF was found to stimulate angiogenesis and BM cell migration in vivo. Tumor growth was found to be significantly increased in mice treated with G-CSF. The average tumor volume for the group treated with G-CSF was 1218 mm(3) compared with 577 mm(3) for the control group (P = .006). CONCLUSIONS: The findings that ES cells and patient tumors expressed both G-CSF and its receptor in vitro and in vivo and that the administration of G-CSF promoted tumor growth in vivo suggest that the potential consequences of G-CSF administration should be investigated further.


Assuntos
Neoplasias Ósseas/química , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Fator Estimulador de Colônias de Granulócitos/análise , Receptores de Fator Estimulador de Colônias de Granulócitos/análise , Sarcoma de Ewing/química , Neoplasias Ósseas/irrigação sanguínea , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Quimiotaxia , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Neovascularização Patológica , RNA/análise , Receptores de Fator Estimulador de Colônias de Granulócitos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sarcoma de Ewing/irrigação sanguínea , Sarcoma de Ewing/patologia
9.
Clin Cancer Res ; 13(16): 4867-73, 2007 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-17699866

RESUMO

PURPOSE: We previously showed that bone marrow cells participate in new tumor vessel formation in Ewing's sarcoma, and that vascular endothelial growth factor 165 (VEGF(165)) is critical to this process. The purpose of this study was to determine whether blocking VEGF receptor 2 (VEGFR-2) with DC101 antibody suppresses tumor growth, reduces tumor vessel formation, and inhibits the migration of bone marrow cells into the tumor. EXPERIMENTAL DESIGN: An H-2 MHC-mismatched bone marrow transplant Ewing's sarcoma mouse model was used. Bone marrow cells from CB6F1 (MHC H-2(b/d)) mice were injected into irradiated BALB/cAnN mice (MHC H-2(d)). TC71 Ewing's sarcoma cells were s.c. injected 4 weeks after the bone marrow transplantation. Mice were then treated i.p. with DC101 antibody or immunoglobulin G (control) twice a week for 3 weeks starting 3 days after tumor cell injection. RESULTS: DC101 antibody therapy significantly reduced tumor growth and tumor mean vessel density (P < 0.05) and increased tumor cell apoptosis. Decreased bone marrow cell migration into the tumor was also shown after DC101 therapy as assessed by the colocalization of H-2K(b) and CD31 using immunohistochemistry. DC101 inhibited the migration of both human and mouse vessel endothelial cells in vitro. CONCLUSION: These results indicated that blocking VEGFR-2 with DC101 antibodies may be a useful therapeutic approach for treating patients with Ewing's sarcoma.


Assuntos
Anticorpos/uso terapêutico , Neovascularização Patológica/prevenção & controle , Sarcoma de Ewing/irrigação sanguínea , Sarcoma de Ewing/terapia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Células da Medula Óssea/fisiologia , Linhagem Celular Tumoral , Movimento Celular , Células Endoteliais/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Pericitos/fisiologia , Ratos , Sarcoma de Ewing/patologia
10.
Int J Cancer ; 119(4): 839-46, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16557578

RESUMO

The Ewing's sarcoma cell line TC71 overexpresses vascular endothelial growth factor isoform 165 (VEGF165), a potent proangiogenic molecule that induces endothelial cell proliferation, migration, and chemotaxis. CD34+ bone marrow stem cells can differentiate into endothelial and hematopoietic cells. We used a transplant model to determine whether CD34+ cells migrate from the bone marrow to Ewing's sarcoma tumors and participate in the neovascularization process that supports tumor growth. We also examined the role of VEGF165 in CD34+ cell migration. Human umbilical cord CD34+ cells were transplanted into sublethally irradiated severe combined immunodeficient mice. Seven days later, the mice were injected subcutaneously with TC71 tumor cells. Tumors were excised 2 weeks later and analyzed by immunohistochemistry. The tumor sections expressed both human VE-cadherin and mouse CD31, indicating involvement of donor-derived human cells in the tumor vessels. To determine the role of VEGF165 in the chemoattraction of CD34+ cells, we generated two VEGF165-deficient TC71 clones, a stable anti-sense VEGF165 cell line (Clone 17) and a VEGF165 siRNA-inhibited clone (TC/siVEGF(7-1)). The resulting VEGF165-deficient tumor cells had normal growth rates in vitro, but had delayed growth when implanted into mice. Immunohistochemical analysis revealed decreased infiltration of CD34+ cells into both VEGF165-deficient tumors. These data show that bone marrow stem cells contribute to the growing tumor vasculature in Ewing's sarcoma and that VEGF165 is critical for the migration of CD34+ cells from the bone marrow into the tumor.


Assuntos
Antígenos CD34/metabolismo , Sarcoma de Ewing/irrigação sanguínea , Sarcoma de Ewing/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Fator A de Crescimento do Endotélio Vascular/biossíntese , Animais , Movimento Celular , Colágeno , DNA Antissenso/genética , Combinação de Medicamentos , Regulação Neoplásica da Expressão Gênica , Humanos , Laminina , Camundongos , Transplante de Neoplasias , Proteoglicanas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sarcoma de Ewing/genética , Sarcoma de Ewing/patologia , Fator A de Crescimento do Endotélio Vascular/deficiência , Fator A de Crescimento do Endotélio Vascular/genética
11.
Clin Exp Metastasis ; 21(8): 747-53, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-16035619

RESUMO

INTRODUCTION: Expression of adhesion molecules such as alphavbeta3 integrin has been associated with the metastatic potential of tumor cells. The purpose of this study was to determine whether alphavbeta3 expression correlated with the metastatic potential of human osteosarcoma cells. MATERIALS AND METHODS: We developed a series of sublines (LM2-LM7) from human osteosarcoma SAOS parental cells, with progressively increasing potential to form lung metastases in nude mice after intravenous injection. SAOS parental and LM2 cells were poorly metastatic, but LM7 cells resulted in visible metastatic lung nodules by 6-8 weeks. We quantified alphavbeta3 integrin expression using flow cytometry. RESULTS: alphavbeta3 expression correlated with the metastatic potential of the cells, with LM7 cells showing the highest expression. LM7 cell adhesion to vitronectin decreased after treatment with echistatin, a RGD-containing peptide antagonist of alphavbeta3. LM7 cells demonstrated higher chemotactic activity than SAOS cells to a homogenate made from lung tissue. This chemotactic activity was also inhibited by echistatin. These data indicated that alphavbeta3 was critical for the migration of LM7 cells to the lung homogenate. Chemotaxis to a liver homogenate was the same for LM7 and SAOS cells. Migration of LM7 cells through lung endothelial cells was higher than that through liver endothelial cells, and echistatin again inhibited this migration. CONCLUSIONS: alphavbeta3 integrin expression may play a role in the metastatic potential of osteosarcoma cells by enhancing the ability of the cells to migrate specifically to the lung. Alphavbeta3 integrin may therefore be a potential new target for osteosarcoma.


Assuntos
Neoplasias Ósseas/metabolismo , Movimento Celular , Integrina alfaVbeta3/metabolismo , Osteossarcoma/metabolismo , Animais , Antineoplásicos/farmacologia , Neoplasias Ósseas/patologia , Adesão Celular , Quimiotaxia , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Integrina alfaVbeta3/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intercelular , Fígado/citologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Pulmão/citologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Masculino , Camundongos , Camundongos Nus , Oligopeptídeos/farmacologia , Osteossarcoma/secundário , Peptídeos/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Receptores de Vitronectina/antagonistas & inibidores , Células Tumorais Cultivadas , Vitronectina/metabolismo
12.
Clin Cancer Res ; 8(11): 3622-7, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12429654

RESUMO

Vasculogenesis, the process by which endothelial cell precursors are recruited and organized to form a vasculature, has traditionally been thought to play a role only in embryonic development. However, several studies have now been published suggesting that vasculogenesis may have a role in the formation of new vascular networks during postnatal life. Recent studies suggest the existence of circulating endothelial precursor cells that arise from outside the place of vascularization. Using a mouse bone marrow (BM) transplantation model that takes advantage of MHC haplotype differences between donor and recipient mice, we examined the contribution of donor BM-derived cells to neovascularization in recipient nude mice with developing Ewing's sarcoma tumors. We found that the donor BM cells gave rise to endothelial cells in vitro and colocalized with neovessels in Ewing's sarcomas in vivo. We also found that donor BM-derived cells were involved in the formation of the tumor vasculature. Our findings indicate that not only angiogenesis but also vasculogenesis was involved in the development of Ewing's sarcoma in our mouse model.


Assuntos
Neovascularização Patológica , Sarcoma de Ewing/irrigação sanguínea , Animais , Antígenos CD , Transplante de Medula Óssea , Células Cultivadas , Endoglina , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Feminino , Haplótipos , Humanos , Imuno-Histoquímica , Interleucina-1/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Modelos Biológicos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/biossíntese , Reação em Cadeia da Polimerase , Polimorfismo Genético , Receptores de Superfície Celular , Células Tumorais Cultivadas , Molécula 1 de Adesão de Célula Vascular/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA