Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 19(1): e1010753, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36689549

RESUMO

Kaposi's sarcoma herpesvirus (KSHV) is the etiologic agent of Kaposi's sarcoma (KS), a hyperplasia consisting of enlarged malformed vasculature and spindle-shaped cells, the main proliferative component of KS. While spindle cells express markers of lymphatic and blood endothelium, the origin of spindle cells is unknown. Endothelial precursor cells have been proposed as the source of spindle cells. We previously identified two types of circulating endothelial colony forming cells (ECFCs), ones that expressed markers of blood endothelium and ones that expressed markers of lymphatic endothelium. Here we examined both blood and lymphatic ECFCs infected with KSHV. Lymphatic ECFCs are significantly more susceptible to KSHV infection than the blood ECFCs and maintain the viral episomes during passage in culture while the blood ECFCs lose the viral episome. Only the KSHV-infected lymphatic ECFCs (K-ECFCLY) grew to small multicellular colonies in soft agar whereas the infected blood ECFCs and all uninfected ECFCs failed to proliferate. The K-ECFCLYs express high levels of SOX18, which supported the maintenance of high copy number of KSHV genomes. When implanted subcutaneously into NSG mice, the K-ECFCLYs persisted in vivo and recapitulated the phenotype of KS tumor cells with high number of viral genome copies and spindling morphology. These spindle cell hallmarks were significantly reduced when mice were treated with SOX18 inhibitor, SM4. These data suggest that KSHV-infected lymphatic ECFCs can be utilized as a KSHV infection model for in vivo translational studies to test novel inhibitors representing potential treatment modalities for KS.


Assuntos
Herpesvirus Humano 8 , Sarcoma de Kaposi , Animais , Camundongos , Herpesvirus Humano 8/genética , Células Endoteliais , Endotélio Vascular/patologia
2.
Cancer Res ; 80(15): 3116-3129, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32518203

RESUMO

Kaposi sarcoma is a tumor caused by Kaposi sarcoma herpesvirus (KSHV) infection and is thought to originate from lymphatic endothelial cells (LEC). While KSHV establishes latency in virtually all susceptible cell types, LECs support spontaneous expression of oncogenic lytic genes, high viral genome copies, and release of infectious virus. It remains unknown the contribution of spontaneous virus production to the expansion of KSHV-infected tumor cells and the cellular factors that render the lymphatic environment unique to KSHV life cycle. We show here that expansion of the infected cell population, observed in LECs, but not in blood endothelial cells, is dependent on the spontaneous virus production from infected LECs. The drivers of lymphatic endothelium development, SOX18 and PROX1, regulated different steps of the KSHV life cycle. SOX18 enhanced the number of intracellular viral genome copies and bound to the viral origins of replication. Genetic depletion or chemical inhibition of SOX18 caused a decrease of KSHV genome copy numbers. PROX1 interacted with ORF50, the viral initiator of lytic replication, and bound to the KSHV genome in the promoter region of ORF50, increasing its transactivation activity and KSHV spontaneous lytic gene expression and infectious virus release. In Kaposi sarcoma tumors, SOX18 and PROX1 expression correlated with latent and lytic KSHV protein expression. These results demonstrate the importance of two key transcriptional drivers of LEC fate in the regulation of the tumorigenic KSHV life cycle. Moreover, they introduce molecular targeting of SOX18 as a potential novel therapeutic avenue in Kaposi sarcoma. SIGNIFICANCE: SOX18 and PROX1, central regulators of lymphatic development, are key factors for KSHV genome maintenance and lytic cycle in lymphatic endothelial cells, supporting Kaposi sarcoma tumorigenesis and representing attractive therapeutic targets.


Assuntos
Transformação Celular Viral/genética , Herpesvirus Humano 8/fisiologia , Proteínas de Homeodomínio/fisiologia , Fatores de Transcrição SOXF/fisiologia , Sarcoma de Kaposi/genética , Proteínas Supressoras de Tumor/fisiologia , Replicação Viral/genética , Carcinogênese/genética , Células Cultivadas , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Células Endoteliais/virologia , Regulação Viral da Expressão Gênica/genética , Genoma Viral/genética , Células HEK293 , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/patogenicidade , Proteínas de Homeodomínio/genética , Humanos , Sistema Linfático/metabolismo , Sistema Linfático/patologia , Sistema Linfático/virologia , Fatores de Transcrição SOXF/genética , Sarcoma de Kaposi/patologia , Sarcoma de Kaposi/virologia , Proteínas Supressoras de Tumor/genética
3.
Sci Rep ; 8(1): 9531, 2018 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-29934628

RESUMO

The transcription factor PROX1 is essential for development and cell fate specification. Its function in cancer is context-dependent since PROX1 has been shown to play both oncogenic and tumour suppressive roles. Here, we show that PROX1 suppresses the transcription of MMP14, a metalloprotease involved in angiogenesis and cancer invasion, by binding and suppressing the activity of MMP14 promoter. Prox1 deletion in murine dermal lymphatic vessels in vivo and in human LECs increased MMP14 expression. In a hepatocellular carcinoma cell line expressing high endogenous levels of PROX1, its silencing increased both MMP14 expression and MMP14-dependent invasion in 3D. Moreover, PROX1 ectopic expression reduced the MMP14-dependent 3D invasiveness of breast cancer cells and angiogenic sprouting of blood endothelial cells in conjunction with MMP14 suppression. Our study uncovers a new transcriptional regulatory mechanism of cancer cell invasion and endothelial cell specification.


Assuntos
Proteínas de Homeodomínio/metabolismo , Metaloproteinase 14 da Matriz/genética , Transcrição Gênica , Proteínas Supressoras de Tumor/metabolismo , Animais , Linhagem Celular , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Técnicas de Inativação de Genes , Proteínas de Homeodomínio/genética , Humanos , Vasos Linfáticos/metabolismo , Camundongos , Regiões Promotoras Genéticas/genética , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética
4.
Elife ; 72018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29712618

RESUMO

Lymphatic invasion and lymph node metastasis correlate with poor clinical outcome in melanoma. However, the mechanisms of lymphatic dissemination in distant metastasis remain incompletely understood. We show here that exposure of expansively growing human WM852 melanoma cells, but not singly invasive Bowes cells, to lymphatic endothelial cells (LEC) in 3D co-culture facilitates melanoma distant organ metastasis in mice. To dissect the underlying molecular mechanisms, we established LEC co-cultures with different melanoma cells originating from primary tumors or metastases. Notably, the expansively growing metastatic melanoma cells adopted an invasively sprouting phenotype in 3D matrix that was dependent on MMP14, Notch3 and ß1-integrin. Unexpectedly, MMP14 was necessary for LEC-induced Notch3 induction and coincident ß1-integrin activation. Moreover, MMP14 and Notch3 were required for LEC-mediated metastasis of zebrafish xenografts. This study uncovers a unique mechanism whereby LEC contact promotes melanoma metastasis by inducing a reversible switch from 3D growth to invasively sprouting cell phenotype.


Assuntos
Neoplasias da Mama/patologia , Endotélio Linfático/patologia , Integrina beta1/metabolismo , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/secundário , Metaloproteinase 14 da Matriz/metabolismo , Receptor Notch3/metabolismo , Animais , Apoptose , Neoplasias da Mama/metabolismo , Movimento Celular , Proliferação de Células , Células Cultivadas , Endotélio Linfático/metabolismo , Feminino , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Pulmonares/metabolismo , Metástase Linfática , Camundongos , Camundongos SCID , Invasividade Neoplásica , Ensaios Antitumorais Modelo de Xenoenxerto , Peixe-Zebra
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...