Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 1496, 2024 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-38383468

RESUMO

Pancreatic ductal adenocarcinoma (PDAC), a lethal disease, requires a grasp of its biology for effective therapies. Exosomes, implicated in cancer, are poorly understood in living systems. Here we use the genetically engineered mouse model (ExoBow) to map the spatiotemporal distribution of exosomes from healthy and PDAC pancreas in vivo to determine their biological significance. We show that, within the PDAC microenvironment, cancer cells establish preferential communication routes through exosomes with cancer associated fibroblasts and endothelial cells. The latter being a conserved event in the healthy pancreas. Inhibiting exosomes secretion in both scenarios enhances angiogenesis, underscoring their contribution to vascularization and to cancer. Inter-organ communication is significantly increased in PDAC with specific organs as most frequent targets of exosomes communication occurring in health with the thymus, bone-marrow, brain, and intestines, and in PDAC with the kidneys, lungs and thymus. In sum, we find that exosomes mediate an organized intra- and inter- pancreas communication network with modulatory effects in vivo.


Assuntos
Carcinoma Ductal Pancreático , Exossomos , Neoplasias Pancreáticas , Camundongos , Animais , Exossomos/patologia , Células Endoteliais/patologia , Linhagem Celular Tumoral , Movimento Celular , Neoplasias Pancreáticas/patologia , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Pâncreas/patologia , Microambiente Tumoral
2.
Nat Commun ; 13(1): 405, 2022 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-35058437

RESUMO

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused the pandemic of the coronavirus induced disease 2019 (COVID-19) with evolving variants of concern. It remains urgent to identify novel approaches against broad strains of SARS-CoV-2, which infect host cells via the entry receptor angiotensin-converting enzyme 2 (ACE2). Herein, we report an increase in circulating extracellular vesicles (EVs) that express ACE2 (evACE2) in plasma of COVID-19 patients, which levels are associated with severe pathogenesis. Importantly, evACE2 isolated from human plasma or cells neutralizes SARS-CoV-2 infection by competing with cellular ACE2. Compared to vesicle-free recombinant human ACE2 (rhACE2), evACE2 shows a 135-fold higher potency in blocking the binding of the viral spike protein RBD, and a 60- to 80-fold higher efficacy in preventing infections by both pseudotyped and authentic SARS-CoV-2. Consistently, evACE2 protects the hACE2 transgenic mice from SARS-CoV-2-induced lung injury and mortality. Furthermore, evACE2 inhibits the infection of SARS-CoV-2 variants (α, ß, and δ) with equal or higher potency than for the wildtype strain, supporting a broad-spectrum antiviral mechanism of evACE2 for therapeutic development to block the infection of existing and future coronaviruses that use the ACE2 receptor.


Assuntos
Enzima de Conversão de Angiotensina 2/imunologia , COVID-19/imunologia , Vesículas Extracelulares/imunologia , SARS-CoV-2/imunologia , Células A549 , Enzima de Conversão de Angiotensina 2/genética , Enzima de Conversão de Angiotensina 2/metabolismo , Animais , COVID-19/sangue , COVID-19/epidemiologia , Chlorocebus aethiops , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Células HEK293 , Células HeLa , Humanos , Camundongos Transgênicos , Testes de Neutralização/métodos , Pandemias/prevenção & controle , Ligação Proteica , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/imunologia , Glicoproteína da Espícula de Coronavírus/metabolismo , Análise de Sobrevida , Células Vero
3.
Gut ; 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-35012996

RESUMO

OBJECTIVE: Intratumor heterogeneity drives cancer progression and therapy resistance. However, it has yet to be determined whether and how subpopulations of cancer cells interact and how this interaction affects the tumour. DESIGN: We have studied the spontaneous flow of extracellular vesicles (EVs) between subpopulations of cancer cells: cancer stem cells (CSC) and non-stem cancer cells (NSCC). To determine the biological significance of the most frequent communication route, we used pancreatic ductal adenocarcinoma (PDAC) orthotopic models, patient-derived xenografts (PDXs) and genetically engineered mouse models (GEMMs). RESULTS: We demonstrate that PDAC tumours establish an organised communication network between subpopulations of cancer cells using EVs called the EVNet). The EVNet is plastic and reshapes in response to its environment. Communication within the EVNet occurs preferentially from CSC to NSCC. Inhibition of this communication route by impairing Rab27a function in orthotopic xenographs, GEMMs and PDXs is sufficient to hamper tumour growth and phenocopies the inhibition of communication in the whole tumour. Mechanistically, we provide evidence that CSC EVs use agrin protein to promote Yes1 associated transcriptional regulator (YAP) activation via LDL receptor related protein 4 (LRP-4). Ex vivo treatment of PDXs with antiagrin significantly impairs proliferation and decreases the levels of activated YAP.Patients with high levels of agrin and low inactive YAP show worse disease-free survival. In addition, patients with a higher number of circulating agrin+ EVs show a significant increased risk of disease progression. CONCLUSION: PDAC tumours establish a cooperation network mediated by EVs that is led by CSC and agrin, which allows tumours to adapt and thrive. Targeting agrin could make targeted therapy possible for patients with PDAC and has a significant impact on CSC that feeds the tumour and is at the centre of therapy resistance.

4.
Biosensors (Basel) ; 9(2)2019 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-31137820

RESUMO

Rapid multiplex cell surface marker analysis can expedite investigations in which large number of antigens need to be analyzed. Simultaneous analysis of multiple surface antigens at the same level of sensitivity is however limited in the current golden standard analysis method, flow cytometry. In this paper we introduce a surface plasmon resonance imaging (SPRi)-based technique for 44-plex parameter analysis using a single sample, in less than 20 min. We analyzed the expression on cells from five different cancer cell lines by SPRi on a 44-plex antibody array including 4 negative controls and compared the output with flow cytometry. The combined correlation of the markers that showed expression by flow cytometry was 0.76. The results demonstrate as a proof of principle that SPRi can be applied for rapid semi-quantitative multiplex cell surface marker analysis.


Assuntos
Antígenos de Neoplasias/análise , Citometria de Fluxo/métodos , Ressonância de Plasmônio de Superfície/métodos , Humanos , Células MCF-7
5.
Semin Cell Dev Biol ; 78: 13-21, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-28803894

RESUMO

Exosomes are small extracellular vesicles with a significant role in most processes associated with cancer. On one hand, exosomes role in the different hallmarks of cancer has been widely described, highlighting the urge to understand the potential to target communication mediated by exosomes as a novel therapeutic approach in cancer. On the other hand, exosomes stability in circulation and tumor-targeting capacity shows their applicability in the delivery of anti-cancer molecules. This review will discuss the dual applicability of exosomes in cancer focusing on their usage for therapy improvement, or their targeting to block their supportive role in tumor progression and response to therapy. We highlight the current developments and the strategies used to enhance the potential of exosomes to become clinical partners in the treatment of cancer.


Assuntos
Comunicação Celular/fisiologia , Exossomos/patologia , Neoplasias/patologia , Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/sangue , Comunicação Celular/efeitos dos fármacos , Exossomos/efeitos dos fármacos , Humanos , Neoplasias/tratamento farmacológico
6.
Cancer Res ; 77(23): 6480-6488, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29162616

RESUMO

Exosomes are a subclass of extracellular vesicles involved in intercellular communication that are released by all cell types, including cancer cells. Cancer exosomes carry malignant information in the form of proteins, lipids, and nucleic acids that can reprogram recipient cells. Exosomes have emerged as putative biological mediators in cancer contributing to major steps of disease progression. A leading role exists for cancer exosomes in specific aspects of tumor progression: modulation of immune response, tumor microenvironment reprogramming, and metastasis. This review will address the functions attributed to cancer exosomes in these three aspects of cancer biology, highlighting recent advances and potential limitations. Finally, we explore alternative strategies to develop better models to study cancer exosomes biology. Cancer Res; 77(23); 6480-8. ©2017 AACR.


Assuntos
Exossomos/imunologia , Exossomos/patologia , Metástase Neoplásica/patologia , Neoplasias/imunologia , Neoplasias/patologia , Biomarcadores Tumorais , Linfócitos T CD4-Positivos/imunologia , Comunicação Celular , Células Dendríticas/imunologia , Progressão da Doença , Humanos , Metástase Neoplásica/imunologia , Microambiente Tumoral/imunologia , Microambiente Tumoral/fisiologia
7.
Nature ; 546(7659): 498-503, 2017 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-28607485

RESUMO

The mutant form of the GTPase KRAS is a key driver of pancreatic cancer but remains a challenging therapeutic target. Exosomes are extracellular vesicles generated by all cells, and are naturally present in the blood. Here we show that enhanced retention of exosomes, compared to liposomes, in the circulation of mice is likely due to CD47-mediated protection of exosomes from phagocytosis by monocytes and macrophages. Exosomes derived from normal fibroblast-like mesenchymal cells were engineered to carry short interfering RNA or short hairpin RNA specific to oncogenic KrasG12D, a common mutation in pancreatic cancer. Compared to liposomes, the engineered exosomes (known as iExosomes) target oncogenic KRAS with an enhanced efficacy that is dependent on CD47, and is facilitated by macropinocytosis. Treatment with iExosomes suppressed cancer in multiple mouse models of pancreatic cancer and significantly increased overall survival. Our results demonstrate an approach for direct and specific targeting of oncogenic KRAS in tumours using iExosomes.


Assuntos
Exossomos/metabolismo , Inativação Gênica , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/terapia , Proteínas Proto-Oncogênicas p21(ras)/genética , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Animais , Antígeno CD47/metabolismo , Modelos Animais de Doenças , Exossomos/imunologia , Feminino , Terapia Genética , Lipossomos/imunologia , Camundongos , Monócitos/citologia , Monócitos/imunologia , Metástase Neoplásica/prevenção & controle , Neoplasias Pancreáticas/sangue , Neoplasias Pancreáticas/patologia , Pinocitose , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Taxa de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...