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.
Commun Biol ; 6(1): 145, 2023 02 03.
Article in English | MEDLINE | ID: mdl-36737664

ABSTRACT

Cancer cells experience confinement as they navigate the tumour microenvironment during metastasis. Recent studies have revealed that the nucleus can function as a 'ruler' for measuring physical confinement via membrane tension, allowing for compression-sensitive changes in migration. Cell nuclei contain many nuclear bodies that form when their components phase separate and condense within permissive local regions within the nucleus. However, how sub-nuclear organisation and phase separation changes with cell confinement and compression is largely unknown. Here we focus on paraspeckles, stress-responsive subnuclear bodies that form by phase separation around the long non-coding RNA NEAT1. As cells entered moderate confinement, a significant increase in paraspeckle number and size was observed compared to unconfined cells. Paraspeckle polarization bias towards the leading edge was also observed in confinement, correlating with regions of euchromatin. Increasing paraspeckle abundance resulted in increases in confined migration likelihood, speed, and directionality, as well as an enhancement of paraspeckle polarization towards the leading edge. This polarization of paraspeckle condensates may play a key role in regulating confined migration and invasion in cancer cells, and illustrates the utility of microchannel-based assays for identifying phenomena not observed on 2D or 3D bulk substrates.


Subject(s)
Paraspeckles , RNA, Long Noncoding , Cell Nucleus/genetics , RNA, Long Noncoding/genetics
2.
Oncotarget ; 7(46): 74602-74611, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27806341

ABSTRACT

Major histocompatibility complex (MHC) class I molecules present antigenic peptides to cytotoxic T cells. During an adaptive immune response, MHC molecules are regulated by several mechanisms including lipopolysaccharide (LPS) and interferon gamma (IFN-g). However, it is unclear whether the serine protease cathepsin G (CatG), which is generally secreted by neutrophils at the site of inflammation, might regulate MHC I molecules. We identified CatG, and to a higher extend CatG and lactoferrin (LF), as an exogenous regulator of cell surface MHC I expression of immune cells and glioblastoma stem cells. In addition, levels of MHC I molecules are reduced on dendritic cells from CatG deficient mice compared to their wild type counterparts. Furthermore, cell surface CatG on immune cells, including T cells, B cells, and NK cells triggers MHC I on THP-1 monocytes suggesting a novel mechanism for CatG to facilitate intercellular communication between infiltrating cells and the respective target cell. Subsequently, our findings highlight the pivotal role of CatG as a checkpoint protease which might force target cells to display their intracellular MHC I:antigen repertoire.


Subject(s)
Cathepsin G/pharmacology , Glioblastoma/genetics , Glioblastoma/metabolism , Histocompatibility Antigens Class I/metabolism , Immune System/cytology , Immune System/metabolism , Animals , Cathepsin G/genetics , Cathepsin G/metabolism , Cell Line , Cell Line, Tumor , Cell Membrane/metabolism , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/immunology , Histocompatibility Antigens Class I/genetics , Humans , Immune System/immunology , Lactoferrin/metabolism , Male , Mice , Mice, Knockout , Monocytes/drug effects , Monocytes/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Proteolysis
3.
Cancer Immunol Immunother ; 65(3): 283-91, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26837514

ABSTRACT

To mount an adaptive immune response, MHC I molecules present antigenic peptides to CTLs. Transcriptional reduction of MHC I molecules is a strategy of immune evasion, which impairs the detection of infected or tumorous cells by CTLs. Natural killer (NK) cells, on the other hand, eliminate target cells specifically in the absence of MHC I. Consequently, infected or tumorous cells partly retain their MHC I at the cell surface to avoid NK recognition. However, it remains unclear which protease degrades MHC I molecules and how these cells maintain a limited set of MHC I at the cell surface. Here, we demonstrate that cathepsin G (CatG), a serine protease, found in the endocytic compartment of APCs and, to a lesser extent, CatD and CatS proteolytically degrade MHC I molecules. Inhibition of CatG boosted MHC I expression at the cell surface of primary human immune cells. In contrast, human glioblastoma cells do not harbor active CatG and might have lost the ability to proteolytically degrade MHC I during tumorigenesis to avoid NK-mediated killing. Overexpression of CatG in glioblastoma cells resulted in a rapid and efficient MHC I degradation. In conclusion, CatG is an essential protease for regulating MHC I molecules and thus modulation of CatG activity might present a new avenue for therapeutic intervention.


Subject(s)
Brain Neoplasms/immunology , Cathepsin G/physiology , Glioblastoma/immunology , Histocompatibility Antigens Class I/metabolism , Cell Line, Tumor , Histocompatibility Antigens Class I/analysis , Humans , Proteolysis
SELECTION OF CITATIONS
SEARCH DETAIL
...