Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Signal ; 10(482)2017 Jun 06.
Article in English | MEDLINE | ID: mdl-28588081

ABSTRACT

Increased protein translation in cells and various factors in the tumor microenvironment can induce endoplasmic reticulum (ER) stress, which initiates the unfolded protein response (UPR). We have previously reported that factors released from cancer cells mounting a UPR induce a de novo UPR in bone marrow-derived myeloid cells, macrophages, and dendritic cells that facilitates protumorigenic characteristics in culture and tumor growth in vivo. We investigated whether this intercellular signaling, which we have termed transmissible ER stress (TERS), also operates between cancer cells and what its functional consequences were within the tumor. We found that TERS signaling induced a UPR in recipient human prostate cancer cells that included the cell surface expression of the chaperone GRP78. TERS also activated Wnt signaling in recipient cancer cells and enhanced resistance to nutrient starvation and common chemotherapies such as the proteasome inhibitor bortezomib and the microtubule inhibitor paclitaxel. TERS-induced activation of Wnt signaling required the UPR kinase and endonuclease IRE1. However, TERS-induced enhancement of cell survival was predominantly mediated by the UPR kinase PERK and a reduction in the abundance of the transcription factor ATF4, which prevented the activation of the transcription factor CHOP and, consequently, the induction of apoptosis. When implanted in mice, TERS-primed cancer cells gave rise to faster growing tumors than did vehicle-primed cancer cells. Collectively, our data demonstrate that TERS is a mechanism of intercellular communication through which tumor cells can adapt to stressful environments.


Subject(s)
Bortezomib/pharmacology , Cell Survival/drug effects , Drug Resistance , Endoplasmic Reticulum Stress/drug effects , Paclitaxel/pharmacology , Prostatic Neoplasms/pathology , Unfolded Protein Response/drug effects , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Animals , Antineoplastic Agents/pharmacology , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Tubulin Modulators/pharmacology , Tumor Cells, Cultured , Wnt Proteins/genetics , Wnt Proteins/metabolism , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
2.
Proc Natl Acad Sci U S A ; 110(50): 20182-7, 2013 Dec 10.
Article in English | MEDLINE | ID: mdl-24277816

ABSTRACT

Evolutionarily conserved short (20-30 nucleotides) noncoding RNAs (microRNAs) are powerful regulators of gene expression in a variety of physiological and pathological processes. As such, means to efficiently modulate microRNA function constitute an important therapeutic opportunity. Here we demonstrate that primary B lymphocytes can be genetically programmed with nonviral plasmid DNA for the biogenesis and delivery of antisense sequences (anti-microRNA) against microRNA-150 (miR-150). Within 18 h of transfection with an anti-miR-150 construct, primary B lymphocytes secrete ∼3,000 copies of anti-miR-150 molecules per cell. Anti-miR-150 molecules released by B lymphocytes were internalized by CD8 T lymphocytes during cross-priming in vitro and in vivo, resulting in marked down-regulation of endogenous miR-150. However, such internalization was not observed in the absence of cross-priming. These results suggest that shuttling anti-miR-150 molecules from B lymphocytes to T cells requires the activation of receiver T cells via the antigen receptor. Finally, anti-miR-150 synthesized in B cells were secreted both as free and extracellular vesicle-associated fractions, but only extracellular vesicle-associated anti-miR-150 were apparently taken up by CD8 T cells. Collectively, these data indicate that primary B lymphocytes represent an efficient platform for the synthesis and delivery of short, noncoding RNA, paving the way for an approach to immunogenomic therapies.


Subject(s)
B-Lymphocytes/metabolism , Gene Expression Regulation/genetics , Gene Targeting/methods , Immunotherapy/methods , MicroRNAs/metabolism , RNA, Small Untranslated/biosynthesis , RNA, Small Untranslated/metabolism , Animals , Antibodies/immunology , Cross-Priming , Flow Cytometry , Humans , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Microscopy, Fluorescence , Oligonucleotides/genetics , Plasmids/genetics , Real-Time Polymerase Chain Reaction , Transfection
3.
PLoS One ; 7(12): e51845, 2012.
Article in English | MEDLINE | ID: mdl-23272178

ABSTRACT

Tumor-infiltrating myeloid cells, such as dendritic cells (BMDC), are key regulators of tumor growth. However, the tumor-derived signals polarizing BMDC to a phenotype that subverts cell-mediated anti-tumor immunity have yet to be fully elucidated. Addressing this unresolved problem we show that the tumor unfolded protein response (UPR) can function in a cell-extrinsic manner via the transmission of ER stress (TERS) to BMDC. TERS-imprinted BMDC upregulate the production of pro-inflammatory, tumorigenic cytokines but also the immunosuppressive enzyme arginase. Importantly, they downregulate cross-presentation of high-affinity antigen and fail to effectively cross-prime CD8(+) T cells, causing T cell activation without proliferation and similarly dominantly suppress cross-priming by bystander BMDC. Lastly, TERS-imprinted BMDC facilitate tumor growth in vivo with fewer tumor-infiltrating CD8(+) T cells. In sum, we demonstrate that tumor-borne ER stress imprints ab initio BMDC to a phenotype that recapitulates several of the inflammatory/suppressive characteristics ascribed to tumor-infiltrating myeloid cells, highlighting the tumor UPR as a critical controller of anti-tumor immunity and a new target for immune modulation in cancer.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Endoplasmic Reticulum Stress , Neoplasms/immunology , Neoplasms/metabolism , Animals , Cell Line, Tumor , Cross-Priming/immunology , Humans , Lymphocyte Activation/immunology , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Tumor Burden
SELECTION OF CITATIONS
SEARCH DETAIL
...