Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters

Database
Language
Affiliation country
Publication year range
1.
Immunity ; 56(8): 1825-1843.e6, 2023 08 08.
Article in English | MEDLINE | ID: mdl-37451265

ABSTRACT

Glioblastoma (GBM), a highly lethal brain cancer, is notorious for immunosuppression, but the mechanisms remain unclear. Here, we documented a temporospatial patterning of tumor-associated myeloid cells (TAMs) corresponding to vascular changes during GBM progression. As tumor vessels transitioned from the initial dense regular network to later scant and engorged vasculature, TAMs shifted away from perivascular regions and trafficked to vascular-poor areas. This process was heavily influenced by the immunocompetence state of the host. Utilizing a sensitive fluorescent UnaG reporter to track tumor hypoxia, coupled with single-cell transcriptomics, we revealed that hypoxic niches attracted and sequestered TAMs and cytotoxic T lymphocytes (CTLs), where they were reprogrammed toward an immunosuppressive state. Mechanistically, we identified chemokine CCL8 and cytokine IL-1ß as two hypoxic-niche factors critical for TAM trafficking and co-evolution of hypoxic zones into pseudopalisading patterns. Therefore, perturbation of TAM patterning in hypoxic zones may improve tumor control.


Subject(s)
Glioblastoma , T-Lymphocytes, Cytotoxic , Humans , Tumor-Associated Macrophages , Macrophages , Immunosuppression Therapy , Glioblastoma/pathology , Tumor Microenvironment
2.
Commun Biol ; 4(1): 145, 2021 01 29.
Article in English | MEDLINE | ID: mdl-33514835

ABSTRACT

Infiltrative growth is a major cause of high lethality of malignant brain tumors such as glioblastoma (GBM). We show here that GBM cells upregulate guidance receptor Plexin-B2 to gain invasiveness. Deletion of Plexin-B2 in GBM stem cells limited tumor spread and shifted invasion paths from axon fiber tracts to perivascular routes. On a cellular level, Plexin-B2 adjusts cell adhesiveness, migratory responses to different matrix stiffness, and actomyosin dynamics, thus empowering GBM cells to leave stiff tumor bulk and infiltrate softer brain parenchyma. Correspondingly, gene signatures affected by Plexin-B2 were associated with locomotor regulation, matrix interactions, and cellular biomechanics. On a molecular level, the intracellular Ras-GAP domain contributed to Plexin-B2 function, while the signaling relationship with downstream effectors Rap1/2 appeared variable between GBM stem cell lines, reflecting intertumoral heterogeneity. Our studies establish Plexin-B2 as a modulator of cell biomechanics that is usurped by GBM cells to gain invasiveness.


Subject(s)
Brain Neoplasms/metabolism , Cell Movement , Glioblastoma/metabolism , Nerve Tissue Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Biomechanical Phenomena , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell-Matrix Junctions/genetics , Cell-Matrix Junctions/metabolism , Cell-Matrix Junctions/pathology , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Male , Mice, Inbred ICR , Mice, SCID , Neoplasm Invasiveness , Nerve Tissue Proteins/genetics , Semaphorins/genetics , Semaphorins/metabolism , Shelterin Complex , Signal Transduction , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome , YAP-Signaling Proteins , rap GTP-Binding Proteins/genetics , rap GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL