Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 2 de 2
1.
Brief Bioinform ; 23(1)2022 01 17.
Article En | MEDLINE | ID: mdl-34891154

Long non-coding ribonucleic acids (RNAs) (lncRNAs) are key players in tumorigenesis and immune responses. The nature of their cell type-specific gene expression and other functional evidence support the idea that lncRNAs have distinct cellular functions in the tumor immune microenvironment (TIME). To date, the majority of lncRNA studies have heavily relied on bulk RNA-sequencing data in which various cell types contribute to an averaged signal, limiting the discovery of cell type-specific lncRNA functions. Single-cell RNA-sequencing (scRNA-seq) is a potential solution for tackling this limitation despite the lack of annotations for low abundance yet cell type-specific lncRNAs. Hence, updated annotations and further understanding of the cellular expression of lncRNAs will be necessary for characterizing cell type-specific functions of lncRNA genes in the TIME. In this review, we discuss lncRNAs that are specifically expressed in tumor and immune cells, summarize the regulatory functions of the lncRNAs at the cell type level and highlight how a scRNA-seq approach can help to study the cell type-specific functions of TIME lncRNAs.


Immunity , Neoplasms , RNA, Long Noncoding , Tumor Microenvironment , Base Sequence , Humans , Immunity/genetics , Neoplasms/genetics , Neoplasms/immunology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Sequence Analysis, RNA , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
2.
Front Immunol ; 12: 631472, 2021.
Article En | MEDLINE | ID: mdl-33643317

Splenic long-lived plasma cells are abnormally numerous and deleterious in systemic autoimmune diseases, yet how they accumulate remains poorly understood. We demonstrate here that a pathological role of spleen-derived CD11b+Gr-1+ myeloid cells (SDMCs) underpins the accumulation of splenic long-lived plasma cells in a lupus-prone model named sanroque. We found that SDMCs were progressively accumulated in sanroque mice from the early clinical phase. Transcriptome profiles revealed that SDMCs have a predominant shift toward an inflammatory phenotype relative to the bone marrow-derived counterparts and are distinct from neutrophils and monocytes. SDMCs were expanded in situ via splenic extramedullary myelopoiesis under the proinflammatory cytokine milieu during lupus progression. SDMCs promoted the development of IFN-γ-secreting Th1 and follicular helper T cells, thereby licensing CD4+ T cells to be pathologic activators of SDMCs and plasma cells. SDMCs also directly promoted the survival of plasma cells by providing B-cell activating factor of the TNF family. The frequency of SDMCs correlated with that of splenic long-lived plasma cells. Selective depletion of CD11b+Gr-1+ cells reduced autoantibody production in sanroque mice. Thus, our findings suggest that SDMCs expanded in situ establish a positive feedback loop with CD4+ T cells, leading to accumulation of long-lived plasma cells which exacerbates lupus autoimmunity.


Autoimmunity/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Survival/immunology , Inflammation/immunology , Lupus Erythematosus, Systemic/immunology , Myeloid Cells/immunology , Plasma Cells/physiology , Spleen/cytology , Animals , CD4-Positive T-Lymphocytes/metabolism , Female , Lupus Erythematosus, Systemic/physiopathology , Mice , Mice, Inbred C57BL , Myeloid Cells/metabolism , Myeloid Cells/pathology , Plasma Cells/immunology
...