Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Circulation ; 135(8): 786-805, 2017 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-27899395

RESUMO

BACKGROUND: During adult life, blood vessel formation is thought to occur via angiogenic processes involving branching from existing vessels. An alternate proposal suggests that neovessels form from endothelial progenitors able to assemble the intimal layers. We here aimed to define vessel-resident endothelial progenitors in vivo in a variety of tissues in physiological and pathological situations such as normal aorta, lungs, and wound healing, tumors, and placenta, as well. METHODS: Based on protein expression levels of common endothelial markers using flow cytometry, 3 subpopulations of endothelial cells could be identified among VE-Cadherin+ and CD45- cells. RESULTS: Lineage tracing by using Cdh5creERt2/Rosa-YFP reporter strategy demonstrated that the CD31-/loVEGFR2lo/intracellular endothelial population was indeed an endovascular progenitor (EVP) of an intermediate CD31intVEGFR2lo/intracellular transit amplifying (TA) and a definitive differentiated (D) CD31hiVEGFR2hi/extracellular population. EVP cells arose from vascular-resident beds that could not be transferred by bone marrow transplantation. Furthermore, EVP displayed progenitor-like status with a high proportion of cells in a quiescent cell cycle phase as assessed in wounds, tumors, and aorta. Only EVP cells and not TA and D cells had self-renewal capacity as demonstrated by colony-forming capacity in limiting dilution and by transplantation in Matrigel plugs in recipient mice. RNA sequencing revealed prominent gene expression differences between EVP and D cells. In particular, EVP cells highly expressed genes related to progenitor function including Sox9, Il33, Egfr, and Pdfgrα. Conversely, D cells highly expressed genes related to differentiated endothelium including Ets1&2, Gata2, Cd31, Vwf, and Notch. The RNA sequencing also pointed to an essential role of the Sox18 transcription factor. The role of SOX18 in the differentiation process was validated by using lineage-tracing experiments based on Sox18CreERt2/Rosa-YFP mice. Besides, in the absence of functional SOX18/SOXF, EVP progenitors were still present, but TA and D populations were significantly reduced. CONCLUSIONS: Our findings support an entirely novel endothelial hierarchy, from EVP to TA to D, as defined by self-renewal, differentiation, and molecular profiling of an endothelial progenitor. This paradigm shift in our understanding of vascular-resident endothelial progenitors in tissue regeneration opens new avenues for better understanding of cardiovascular biology.


Assuntos
Células Endoteliais/metabolismo , Células-Tronco/metabolismo , Animais , Antígenos CD/metabolismo , Aorta/metabolismo , Aorta/patologia , Transplante de Medula Óssea , Caderinas/metabolismo , Diferenciação Celular , Células Endoteliais/citologia , Endotélio Vascular/citologia , Feminino , Antígenos Comuns de Leucócito/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica , Placenta/metabolismo , Placenta/patologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Gravidez , Fatores de Transcrição SOXF/metabolismo , Células-Tronco/citologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Ferimentos e Lesões/patologia , Ferimentos e Lesões/terapia
2.
FASEB J ; 31(2): 610-624, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28045376

RESUMO

The prospect of using endothelial progenitors is currently hampered by their low engraftment upon transplantation. We report that mesenchymal stem/stromal cells (MSCs), independent of source and age, improve the engraftment of endothelial colony forming cells (ECFCs). MSC coculture altered ECFC appearance to an elongated mesenchymal morphology with reduced proliferation. ECFC primed via MSC contact had reduced self-renewal potential, but improved capacity to form tube structures in vitro and engraftment in vivo Primed ECFCs displayed major differences in transcriptome compared to ECFCs never exposed to MSCs, affecting genes involved in the cell cycle, up-regulating of genes influencing mesenchymal transition, adhesion, extracellular matrix. Inhibition of NOTCH signaling, a potential upstream regulator of mesenchymal transition, in large part modulated this gene expression pattern and functionally reversed the mesenchymal morphology of ECFCs. The collective results showed that primed ECFCs survive better and undergo a mesenchymal transition that is dependent on NOTCH signaling, resulting in significantly increased vasculogenic potential.-Shafiee, A., Patel, J., Wong, H. Y., Donovan, P., Hutmacher, D. W., Fisk, N. M., Khosrotehrani, K. Priming of endothelial colony-forming cells in a mesenchymal niche improves engraftment and vasculogenic potential by initiating mesenchymal transition orchestrated by NOTCH signaling.


Assuntos
Células Endoteliais/metabolismo , Células-Tronco Mesenquimais/fisiologia , Receptores Notch/metabolismo , Transdução de Sinais/fisiologia , Animais , Diferenciação Celular , Proliferação de Células , Técnicas de Cocultura , Feminino , Regulação da Expressão Gênica/fisiologia , Humanos , Camundongos , Placenta , Gravidez , Receptores Notch/genética
3.
Nat Commun ; 10(1): 18, 2019 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-30604758

RESUMO

Tumor vascularization is a hallmark of cancer central to disease progression and metastasis. Current anti-angiogenic therapies have limited success prompting the need to better understand the cellular origin of tumor vessels. Using fate-mapping analysis of endothelial cell populations in melanoma, we report the very early infiltration of endovascular progenitors (EVP) in growing tumors. These cells harbored self-renewal and reactivated the expression of SOX18 transcription factor, initiating a vasculogenic process as single cells, progressing towards a transit amplifying stage and ultimately differentiating into more mature endothelial phenotypes that comprised arterial, venous and lymphatic subtypes within the core of the tumor. Molecular profiling by RNA sequencing of purified endothelial fractions characterized EVPs as quiescent progenitors remodeling the extracellular matrix with significant paracrine activity promoting growth. Functionally, EVPs did not rely on VEGF-A signaling whereas endothelial-specific loss of Rbpj depleted the population and strongly inhibited metastasis. The understanding of endothelial heterogeneity opens new avenues for more effective anti-vascular therapies in cancer.


Assuntos
Transformação Celular Neoplásica/patologia , Células Progenitoras Endoteliais/patologia , Melanoma Experimental/patologia , Neovascularização Patológica/patologia , Fatores de Transcrição SOXF/metabolismo , Animais , Diferenciação Celular , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Endotélio Vascular/citologia , Endotélio Vascular/patologia , Matriz Extracelular/patologia , Feminino , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/genética , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Vasos Linfáticos/citologia , Vasos Linfáticos/patologia , Masculino , Melanoma Experimental/irrigação sanguínea , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Invasividade Neoplásica/patologia , Fatores de Transcrição SOXF/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
4.
Cell Rep ; 22(8): 2176-2189, 2018 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-29466742

RESUMO

Furin trafficking, and that of related proprotein convertases (PCs), may regulate which substrates are accessible for endoproteolysis, but tools to directly test this hypothesis have been lacking. Here, we develop targeted biosensors that indicate Furin activity in endosomes is 10-fold less inhibited by decanoyl-RVKR-chloromethylketone and enriched >3-fold in endosomes compared to the trans-Golgi network (TGN). Endogenous PC7, which resists this inhibitor, was active in distinct vesicles. Only overexpressed PC7 activity reached the cell surface, endosomes, and the TGN. A PLC motif in the cytosolic tail of PC7 was dispensable for endosomal activity, but it was specifically required for TGN recycling and to rescue proActivin-A cleavage in Furin-depleted B16F1 melanoma cells. In sharp contrast, PC7 complemented Furin in cleaving Notch1 independently of PLC-mediated TGN access. Our study provides a proof in principle that compartment-specific biosensors can be used to gain insight into the regulation of PC trafficking and to map the tropism of PC-specific inhibitors.


Assuntos
Técnicas Biossensoriais , Compartimento Celular , Furina/metabolismo , Subtilisinas/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Proteína 9 Associada à CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Endocitose , Endossomos/metabolismo , Exocitose , Transferência Ressonante de Energia de Fluorescência , Edição de Genes , Células HEK293 , Células HeLa , Humanos , Melanoma/genética , Melanoma/patologia , Melanoma Experimental/patologia , Proteínas Mutantes/metabolismo , Proteólise , Frações Subcelulares/metabolismo , Especificidade por Substrato , Rede trans-Golgi/metabolismo
5.
J Invest Dermatol ; 137(12): 2578-2587, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28844941

RESUMO

The secreted growth factor Activin-A of the transforming growth factor ß family and its receptors can promote or inhibit several cancer hallmarks including tumor cell proliferation and differentiation, vascularization, lymphangiogenesis and inflammation. However, a role in immune evasion and its relationship with tumor-induced muscle wasting and tumor vascularization, and the relative contributions of autocrine versus paracrine Activin signaling remain to be evaluated. To address this, we compared the effects of truncated soluble Activin receptor IIB as a ligand trap, or constitutively active mutant type IB receptor versus secreted Activin-A or the related ligand Nodal in mouse and human melanoma cell lines and tumor grafts. We found that although cell-autonomous receptor activation arrested tumor cell proliferation, Activin-A secretion stimulated melanoma cell dedifferentiation and tumor vascularization by functional blood vessels, and it increased primary and metastatic tumor burden and muscle wasting. Importantly, in mice with impaired adaptive immunity, the tumor-promoting effect of Activin-A was lost despite sustained vascularization and cachexia, suggesting that Activin-A promotes melanoma progression by inhibiting antitumor immunity. Paracrine Activin-A signaling emerges as a potential target for personalized therapies, both to reduce cachexia and to enhance the efficacy of immunotherapies.


Assuntos
Ativinas/metabolismo , Evasão da Resposta Imune , Melanoma/metabolismo , Neoplasias Cutâneas/metabolismo , Animais , Caquexia , Ciclo Celular , Progressão da Doença , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Sistema Imunitário , Antígeno Ki-67/metabolismo , Melanoma/patologia , Melanoma Experimental , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Neovascularização Patológica , Fenótipo , Transdução de Sinais , Neoplasias Cutâneas/patologia , Microambiente Tumoral
6.
Stem Cells Transl Med ; 5(10): 1302-1306, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27381992

RESUMO

: Since the discovery of endothelial progenitor cells (EPCs) almost 2 decades ago, there has been great hope in their use in treating chronic ischemic disease. Unfortunately, to date, many of the clinical trials using EPCs have been hampered by the lack of clear definition of this cell population. Attributes of a progenitor population are self-renewal and multipotentiality. Major progress has been achieved moving from a definition of EPCs based on a candidate cell surface molecule to a functional definition based essentially on self-renewal hierarchy of endothelial colony-forming cells (ECFCs). More recent work has seized on this functional characterization to associate gene expression signatures with the self-renewal capacity of ECFCs. In particular, Notch signaling driving the quiescence of progenitors has been shown to be central to progenitor self-renewal. This new molecular definition has tremendous translational consequences, because progenitors have been shown to display greater vasculogenic potential. Also, this molecular definition of EPC self-renewal allows assessment of the quality of presumed EPC preparations. This promises to be the initial stage in progressing EPCs further into mainstream clinical use. SIGNIFICANCE: The development of a therapy using endothelial progenitor cells provides great hope for patients in treating cardiovascular diseases going forward. For continual development of this therapy toward the clinical, further understanding of the fundamental biology of these cells is required. This will enable a greater understanding of their stemness capacity and provide insight into their ability to differentiate and drive tissue regeneration when injected into a host.


Assuntos
Células Progenitoras Endoteliais/citologia , Animais , Células Progenitoras Endoteliais/classificação , Células Progenitoras Endoteliais/metabolismo , Humanos
7.
Mol Biosyst ; 10(4): 741-58, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24448662

RESUMO

Here we report the identification of a proliferative, viable, and hyperdiploid tumor cell subpopulation present within Glioblastoma (GB) patient tumors. Using xenograft tumor models, we demonstrate that hyperdiploid cell populations are maintained in xenograft tumors and that clonally expanded hyperdiploid cells support tumor formation and progression in vivo. In some patient tumorsphere lines, hyperdiploidy is maintained during long-term culture and in vivo within xenograft tumor models, suggesting that hyperdiploidy can be a stable cell state. In other patient lines hyperdiploid cells display genetic drift in vitro and in vivo, suggesting that in these patients hyperdiploidy is a transient cell state that generates novel phenotypes, potentially facilitating rapid tumor evolution. We show that the hyperdiploid cells are resistant to conventional therapy, in part due to infrequent cell division due to a delay in the G0/G1 phase of the cell cycle. Hyperdiploid tumor cells are significantly larger and more metabolically active than euploid cancer cells, and this correlates to an increased sensitivity to the effects of glycolysis inhibition. Together these data identify GB hyperdiploid tumor cells as a potentially important subpopulation of cells that are well positioned to contribute to tumor evolution and disease recurrence in adult brain cancer patients, and suggest tumor metabolism as a promising point of therapeutic intervention against this subpopulation.


Assuntos
Neoplasias Encefálicas/genética , Divisão Celular/genética , Diploide , Pontos de Checagem da Fase G1 do Ciclo Celular/genética , Glioblastoma/genética , Animais , Antineoplásicos Alquilantes/farmacologia , Proliferação de Células , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Feminino , Glicólise , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Recidiva Local de Neoplasia/genética , Transplante de Neoplasias , Esferoides Celulares , Temozolomida , Transplante Heterólogo , Células Tumorais Cultivadas
8.
Int J Biochem Cell Biol ; 45(3): 706-10, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23262292

RESUMO

Ubiquitination of proteins by the Nedd4 family of ubiquitin ligases is a significant mechanism in protein trafficking and degradation and provides for tight spatiotemporal regulation. Ubiquitination is gaining increasing recognition as a central mechanism underpinning the regulation of neuronal development and homeostasis in the brain. This review will focus on the Nedd4 and Nedd4-2 E3 ubiquitin ligases that are implicated in an increasing number of neuronal protein-protein interactions. Understanding of the contribution of Nedd4 and Nedd4-2 in regulating key functions in the brain is shedding new light on the ubiquitination signal not only in orchestrating degradation events but also in protein trafficking. Furthermore, the description of several novel Nedd4/4-2 targets in neurons is changing the way we conceptualize how neurons maintain normal function and how this is altered in disease.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte , Neurônios/metabolismo , Ubiquitina-Proteína Ligases , Ubiquitinação , Comunicação Celular , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Humanos , Ubiquitina-Proteína Ligases Nedd4 , Neuritos/metabolismo , Neurônios/citologia , Transporte Proteico , Proteólise , Ubiquitina/genética , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
9.
Oncotarget ; 8(49): 84618-84619, 2017 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-29156660
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa