Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Elife ; 92020 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-32909947

RESUMO

Programmed cell death protein-1 (PD-1) checkpoint immunotherapy efficacy remains unpredictable in glioblastoma (GBM) patients due to the genetic heterogeneity and immunosuppressive tumor microenvironments. Here, we report a microfluidics-based, patient-specific 'GBM-on-a-Chip' microphysiological system to dissect the heterogeneity of immunosuppressive tumor microenvironments and optimize anti-PD-1 immunotherapy for different GBM subtypes. Our clinical and experimental analyses demonstrated that molecularly distinct GBM subtypes have distinct epigenetic and immune signatures that may lead to different immunosuppressive mechanisms. The real-time analysis in GBM-on-a-Chip showed that mesenchymal GBM niche attracted low number of allogeneic CD154+CD8+ T-cells but abundant CD163+ tumor-associated macrophages (TAMs), and expressed elevated PD-1/PD-L1 immune checkpoints and TGF-ß1, IL-10, and CSF-1 cytokines compared to proneural GBM. To enhance PD-1 inhibitor nivolumab efficacy, we co-administered a CSF-1R inhibitor BLZ945 to ablate CD163+ M2-TAMs and strengthened CD154+CD8+ T-cell functionality and GBM apoptosis on-chip. Our ex vivo patient-specific GBM-on-a-Chip provides an avenue for a personalized screening of immunotherapies for GBM patients.


Assuntos
Glioblastoma/terapia , Imunoterapia/instrumentação , Dispositivos Lab-On-A-Chip , Receptor de Morte Celular Programada 1/metabolismo , Microambiente Tumoral/imunologia
2.
Adv Healthc Mater ; 8(4): e1801234, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30537061

RESUMO

Macrophages are a predominant immune cell population that drive inflammatory responses and exhibit transitions in phenotype and function during tissue remodeling in disease and repair. Thus, engineering an immunomodulatory biomaterial has significant implications for resolving inflammation. Here, a biomimetic and photoresponsive hyaluronan (HA) hydrogel nanocomposite with tunable 3D extracellular matrix (ECM) adhesion sites for dynamic macrophage immunomodulation is engineered. Photodegradative alkoxylphenacyl-based polycarbonate (APP) nanocomposites are exploited to permit user-controlled Arg-Gly-Asp (RGD) adhesive peptide release and conjugation to a HA-based ECM for real-time integrin activation of macrophages encapsulated in 3D HA-APP nanocomposite hydrogels. It is demonstrated that photocontrolled 3D ECM-RGD peptide conjugation can activate αvß3 integrin of macrophages, and periodic αvß3 integrin activation can enhance anti-inflammatory M2 macrophage polarization. Altogether, an emerging use of biomimetic, photoresponsive, and bioactive HA-APP nanocomposite hydrogel is highlighted to command 3D cell-ECM interactions for modulating macrophage polarization, which may shed light on cell-ECM interactions in innate immunity and inspire new biomaterial-based immunomodulatory therapies.


Assuntos
Ácido Hialurônico , Hidrogéis , Imunomodulação , Macrófagos/imunologia , Nanocompostos/química , Raios Ultravioleta , Animais , Ácido Hialurônico/química , Ácido Hialurônico/farmacologia , Hidrogéis/química , Hidrogéis/farmacologia , Imunomodulação/efeitos dos fármacos , Imunomodulação/efeitos da radiação , Integrina alfaVbeta3/imunologia , Camundongos , Oligopeptídeos/química , Oligopeptídeos/farmacologia , Células RAW 264.7
3.
Biomaterials ; 161: 164-178, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29421553

RESUMO

Glioblastoma (GBM) is the most lethal primary adult brain tumor and its pathology is hallmarked by distorted neovascularization, diffuse tumor-associated macrophage infiltration, and potent immunosuppression. Reconstituting organotypic tumor angiogenesis models with biomimetic cell heterogeneity and interactions, pro-/anti-inflammatory milieu and extracellular matrix (ECM) mechanics is critical for preclinical anti-angiogenic therapeutic screening. However, current in vitro systems do not accurately mirror in vivo human brain tumor microenvironment. Here, we engineered a three-dimensional (3D), microfluidic angiogenesis model with controllable and biomimetic immunosuppressive conditions, immune-vascular and cell-matrix interactions. We demonstrate in vitro, GL261 and CT-2A GBM-like tumors steer macrophage polarization towards a M2-like phenotype for fostering an immunosuppressive and proangiogenic niche, which is consistent with human brain tumors. We distinguished that GBM and M2-like immunosuppressive macrophages promote angiogenesis, while M1-like pro-inflammatory macrophages suppress angiogenesis, which we coin "inflammation-driven angiogenesis." We observed soluble immunosuppressive cytokines, predominantly TGF-ß1, and surface integrin (αvß3) endothelial-macrophage interactions are required in inflammation-driven angiogenesis. We demonstrated tuning cell-adhesion receptors using an integrin (αvß3)-specific collagen hydrogel regulated inflammation-driven angiogenesis through Src-PI3K-YAP signaling, highlighting the importance of altered cell-ECM interactions in inflammation. To validate the preclinical applications of our 3D organoid model and mechanistic findings of inflammation-driven angiogenesis, we screened a novel dual integrin (αvß3) and cytokine receptor (TGFß-R1) blockade that suppresses GBM tumor neovascularization by simultaneously targeting macrophage-associated immunosuppression, endothelial-macrophage interactions, and altered ECM. Hence, we provide an interactive and controllable GBM tumor microenvironment and highlight the importance of macrophage-associated immunosuppression in GBM angiogenesis, paving a new direction of screening novel anti-angiogenic therapies.


Assuntos
Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/metabolismo , Glioblastoma/imunologia , Glioblastoma/metabolismo , Macrófagos/metabolismo , Animais , Linhagem Celular Tumoral , Matriz Extracelular/metabolismo , Macrófagos/imunologia , Camundongos , Microscopia Confocal , Neovascularização Patológica/imunologia , Neovascularização Patológica/metabolismo , Células RAW 264.7 , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA