Comparison of NK cells and ILC1 in lean versus obese mouse livers by RNASeq
Ontology highlight
ABSTRACT: Mice were weaned onto standard RM1 diet or onto a highly palatable obesogenic diet (824018 – ‘45% AFE fat) supplemented with condensed milk. After 12 weeks, mice were killed, and liver NK cells (Lin- NK1.1+ CD49a- CD49b+) or ILC1 (Lin- NK1.1+ CD49a+ CD49b-) were sorted. Total RNA was extracted from sorted cells, cDNA generated and RNASeq performed.
Project description:Bone marrow cells were isolated from wild type and Tbx21 knockout mice. iNK (Lin- NK1.1+ CD11b- CD27+) and mNK1 (Lin- NK1.1+ CD11b+ CD27+) were sorted from WT and Tbx21 KO mice. mNK2 (Lin- NK1.1+ CD11b+ CD27-) were sorted from WT mice only, since these cells are not present in Tbx21 KO mice.
Project description:Purpose: To understanding the effects of RORa deficiency on ILC1 cells, we conducted bulk mRNA-seqencing Method: Firstly, we purified liver ILC1 cells (CD45.2+CD3-CD19-NKp46+NK1.1+CD49a+CD49b-) via Fluorescence Activated Cell Sorting, then frozen in -80 °C ultra-low temperature refrigerator, followed by High-throughput sequencing, in three replicates for WT (Rorafl/fl) mice and one replicate for cKO (Ncr1Cre-Rorafl/fl) , using Illumina Hiseq 1500 platform.
Project description:We report the identification of immature thymic CD4(-),CD8(-) double-negative (DN)1e cells with the NKT cell lineage potential. We also analyzed the gene expression profiles of DN1e thymocytes compared with those of mature thymic NKT cell developmental stages termed NKT stage-1, 2, and -3, which are characterized by differential expression levels of NK1.1 and CD44 antigens in C57BL/6 mouse strain. Next generation sequencing of total transcriptomes using total RNA isolated from FACS sorted ex vivo thymic DN1eP (Lin-/CD44+/CD25-/CD24low/CD5+/CD27+/Ly108-/CXCR3+) fraction, and mature thymic alphaGalCer-loaded CD1d dimer+TCRbeta+ NKT cell developmental stage-1 (CD44-/NK1.1-), stage-2 (CD44+/NK1.1-), and stage-3 (CD44+/NK1.1+) cells.
Project description:The indicated thymic progenitor population was sorted via FACS and then loaded into a Fluidigm C1 small cell capture chip for single-cell capture, lysis, reverse transcription, and preamplification. Preamplified products were analyzed on a BioMark HD with EvaGreen chemistry. The genes analyzed were selected based on bulk RNA sequencing data and/or prior publications, including genes relevant for gamma/delta T cells and T cell progenitors. B6=C57BL/6J, Rag=B6.Rag1-/-, Tcrd=B6.Tcrd-/-, DN1d=Live/TCRd-/Lin-/CD44+/CD25-/CD24+/cKit-, DN2=Live/TCRd-/Lin-/CD44+/CD25+/cKit+, cKit-=Live/TCRd-/Lin-/CD44+/CD25-/cKit-. Lin=CD3/CD8/CD11b/CD11c/CD19/Gr-1/NK1.1/TCRb/Ter-119 Sample naming nomenclature is as follows: MouseLine_CellPopoulation_DateCaptured_CaptureChamber_CellNumberInChamber
Project description:Natural killer (NK) cells are present in large populations at the maternal-fetal interface during early pregnancy, but their roles in fetal growth are unclear. Here, we identify a CD49a+Eomes+ subset of NK cells that have the capability to secrete growth-promoting-factors (GPF), including pleiotrophin, osteoglycin and osteopontin in both humans and mice. Decreases in this GPF-secreting NK subset impair fetal development, and leads to fetal growth restriction. The transcriptional factor Nfil3 but not T-bet affects the function and the number of this decidual NK subset. The crosstalk of the HLA-G-ILT2-KIR2DL4 axis promotes the GPF-secreting function of this NK subset. Adoptive transfer of these induced CD49a+Eomes+NK cells can reverse the impaired fetal growth and rebuild an appropriate local microenvironment. These findings reveal new properties of NK cells in promoting fetal growth as well as novel approaches for therapeutically administering NK cells to reverse restricted nourishments within the microenvironment during early pregnancy. To investigate novel molecular signatures of human decidual NK cells, we performed microarray analysis on dNK cells (CD3−CD56+CD49a+CD49b−) and pNK cells (CD3−CD56+CD49a−CD49b+). dNK cells were purified from first-trimester deciduas. pNK cells were purified from adult peripheral blood mononuclear cells. Samples were collected from healthy adult donors after obtaining informed consent according to the Ethics Committee of the University of Science & Technology of China.
Project description:This study was undertaken to assess heterogeneity within neuroblastoma cell populations, and to assess whether the integrin CD49b can distinguish different neuroblastoma phenotypes. Methods: We performed bulk RNA sequencing on Neuro-2a and SH-SY5Y neuroblastoma cells sorted based on CD49b expression, and CUT&RUN for H3K4me1 and H3K27ac on Neuro-2a cells sorted based on CD49b expression. Results: We find that CD49b expression identifies transcriptionally distinct neuroblastoma cell populations. High CD49b expression marks cells expressing mesenchymal genes, while lack of CD49 denotes cells expression neuronal genes. CUT&RUN analysis shows that CD49b distinguishes cells with distinct enhancer and super enhancer profiles. Conclusion: RNA sequencing and CUT&RUN demonstrate that neuroblastoma cell lines include heterogeneous populations.
Project description:Co-culture of human CD4+ T cells with allogeneeic tolerogenic dendritic cells (DC-10) results in a cell product, called T-allo10, enriched with alloantigen-specific type 1 regulatory T cells (Tr1 cells). CD49b+LAG3+ Tr1 cells and CD49b-LAG3- non-Tr1 cells (DN) were sorted from total T-allo10 cells, and their transcriptomes compared to parental CD4+ T cells, as well as to and control CD49b-LAG3- effector T cells (Teff) sorted from T-allo cells, which were made equivalently to T-allo10 except with using mature dendirtic cells instead of DC-10.
Project description:This goal of this study was to identify genes that are deregulated in the absence of EZH2 in early lymphocyte progenitors. We examined gene expression by RNA-sequencing in sorted CLPs (Lin-CD117int CD127+ CD135+), pro-B cells (B220+CD19+CD43+), DN3 cells (Lin- CD25+ CD117-), splenic NK cells (Lin-NK1.1+DX5+) and bone marrow ILC2 cells (Lin- Sca1+CD127+) from Ezh2fl/fl Il7racre/+ and Il7racre/+ control mice. Reads were aligned to the mm10 reference genome by Tophat2.1.0. Reads were assigned to genes using the htseq-count tool from HTSeq v 0.6.1 and gene annotations from Ensembl release 78. Differential expression was calculated across 2-3 independent replicates by EdgeR. We found that CLPs, ILC2s, and splenic NK cells maintained their normal transcriptional programs despite loss of EZH2. In contrast, loss of EZH2 caused over 1000 genes to be deregulated in pro-B and DN3 cells indicating that EZH2 is required for transcriptomic stability in adaptive, but not innate lymphocyte progenitors.
Project description:To investigate whether liver-resident ILC1s could develop from local hematopoietic progenitors, we analyzed the phenotypic properties of liver CD45+Lin- progenitors. We found that the adult mouse liver contained Lin-Sca-1+Mac-1+ (LSM) hematopoietic progenitors derived from the fetal liver. This population included Lin-CD122+CD49a+ progenitors that could generate liver ILC1s but not conventional NK (cNK) cells. By performing single-cell RNA seq, we show the heterogeneous composition of these hematopoietic progenitors.
Project description:Innate lymphoid cells (ILCs) comprise several subsets that were originally classified based on their cytokine production profiles. Natural killer (NK) cells and type 1 ILCs (ILC1s) were initially classified together, but recent data supported their separation into different lineages. Here we describe how infection with the parasite Toxoplasma gondii induces changes to NK1.1+ NKp46+ cells that persist independent of ongoing infection. Notably, there is an expansion of Eomes– CD49a+ cells that superficially resemble ILC1s, but express unique genes, circulate throughout the vasculature, and possess distinct epigenetic marks. Single-cell RNA sequencing confirms T. gondii-induced Eomes– CD49a+ cells are distinct from both conventional NK cells and ILC1s. Furthermore, there is heterogeneity within this population, as both conventional NK cells and ILC1s contribute to their formation. Indeed, downregulation of Eomes within conventional NK cells accounts for most T. gondii-induced Eomes– CD49a+ cells, indicating that NK cells can give rise to cells resembling ILC1s during infection.