scholarly journals RORα Regulates Cholesterol Metabolism of CD8+ T Cells for Anticancer Immunity

Cancers ◽  
2020 ◽  
Vol 12 (7) ◽  
pp. 1733
Author(s):  
In Kyu Lee ◽  
Hyerin Song ◽  
Hyerim Kim ◽  
Ik Soo Kim ◽  
Na Ly Tran ◽  
...  

Retinoic acid-related orphan receptor α (RORα) functions as a transcription factor for various biological processes, including circadian rhythm, inflammation, cancer, and lipid metabolism. Here, we demonstrate that RORα is crucial for maintaining cholesterol homeostasis in CD8+ T cells by attenuating NF-κB transcriptional activity. Cholesterol sulfate, the established natural agonist of RORα, exhibits cellular cytotoxicity on, and increased effector responses in, CD8+ T cells. Transcript analysis reveals that the suppression of RORα leads to the upregulation of NF-κB target genes in T cells. Chromatin immunoprecipitation analysis was used to determine the corecruitment of RORα and histone deacetylase (HDAC) on NF-κB target promoters and the subsequent dismissal of coactivators for transcriptional repression. We demonstrate that RORα/HDAC-mediated attenuation of NF-κB signaling controls the balance of cholesterol metabolism in CD8+ T cells, and that therapeutic strategies targeting this epigenetic regulation could be beneficial to the treatment of solid tumors including colon cancers.

2021 ◽  
Vol 2021 ◽  
pp. 1-11
Author(s):  
Liang Dong ◽  
Xi Yang ◽  
Yangyanqiu Wang ◽  
Yin Jin ◽  
Qing Zhou ◽  
...  

Background. T cell-mediated antitumor immune response is the basis of colorectal cancer (CRC) immunotherapy. Cholesterol plays an important role in T cell signal transduction and function. Apolipoprotein E (APOE) plays a major role in cholesterol metabolism. Objective. To screen and analyze key markers involved in the anticolon cancer response of CD8+ T cells through the regulation of cholesterol metabolism. Methods. Based on the median cutoff of the expression value of APOE according to the data downloaded from The Cancer Genome Atlas and Gene Expression Omnibus database, patients were grouped into low and high expression groups. Differences in clinical factors were assessed, and survival analysis was performed. Differentially expressed genes (DEGs) in the high and low expression groups were screened, followed by the analysis of differences in tumor-infiltrating immune cells and weighted gene coexpression network analysis results. The closely related genes to APOE were identified, followed by enrichment analysis, protein–protein interaction (PPI) network analysis, and differential expression analysis. Immunohistochemical staining (IHC) was used to detect the expression of CD8 in CRC tissues. Results. There were significant differences in prognosis and pathologic_N between the APOE low and high expression groups. A total of 2,349 DEGs between the high and low expression groups were selected. A total of 967 genes were obtained from the blue and brown modules. The probability of distribution of CD8+ T cells differed significantly between the two groups, and 320 closely related DEGs of APOE were screened. Genes including the HLA gene family, B2M, IRF4, and STAT5A had a higher degree in the PPI network. GEO datasets verified the prognosis and the related DEGs of APOE. IHC staining verified the relationship between the distribution of CD8+ T cells and APOE expression. Conclusion. Genes including the HLA gene family, B2M, IRF4, and STAT5A might be the key genes involved in the anticolon cancer response of CD8+ T cells through the regulation of cholesterol metabolism.


2019 ◽  
Vol 150 (3) ◽  
pp. 464-474 ◽  
Author(s):  
Guanghui Chen ◽  
Kun Wu ◽  
Tao Zhao ◽  
Shicheng Ling ◽  
Wei Liu ◽  
...  

ABSTRACT Background microRNAs (miRNAs) post-transcriptionally regulate gene expression and act as important modulators of cholesterol homeostasis. Objective The study explores the mechanism by which miRNAs mediate high fat–induced changes of cholesterol metabolism in yellow catfish. Methods Yellow catfish (weight: 3.79 ± 0.16 g, 3 mo old, mixed sex) were fed 2 diets containing lipids at 11.3% [control (CON)] or 15.4% [high-fat diet (HFD)] (by weight) for 8 wk. Cholesterol content was measured; hematoxylin-eosin (H&E) staining, qPCR assays, and small RNA sequencing were conducted in the liver. Hepatocytes were isolated from separate, untreated fish and incubated for 24 h in control solution or palmitic acid (PA; 0.25 mM)/oleic acid (OA; 0.5 mM) after 4 h pretreatment with or without miR-144 inhibitor/mimic (40 nM). Cholesterol content was measured; qPCR assays and Western blotting were conducted in the hepatocytes. HEK293T cells were co-transfected with plasmids to validate miR-144 target genes. The promoter activities of miR-144 were analyzed in HEK293T cells with PA (0.25 mM) or OA (0.25 or 0.5 mM) treatment for 24 h. Luciferase activity assays, electrophoretic mobility shift assay, and Western blotting were conducted in HEK293T cells. Results Compared with CON, HFD induced hepatic cholesterol accumulation (31.5%), and upregulated miR-144 expression (8.40-fold, P < 0.05). miR-144 directly targeted hydroxymethylglutaryl-CoA reductase (hmgcr), cholesterol 7α-monooxygenase A1 (cyp7a1), and adenosine triphosphate binding cassette transporter A1 (abca1) in HEK293T cells. In the hepatocytes of yellow catfish, miR-144 inversely regulated the expression of hmgcr, cyp7a1, and abca1 (30.3–78.5%, P < 0.05); loss of miR-144 function alleviated PA- or OA-induced cholesterol accumulation (19.5–61.1%, P < 0.05). We also characterized the C/EBPα binding site in the miR-144 promoter, and found that C/EBPα positively regulated miR-144 expression through binding to the miR-144 promoter. Conclusions miR-144 mediated HFD-induced changes in the liver and hepatocytes of yellow catfish, suggesting a possible mechanism for HFD-induced dysfunction in cholesterol metabolism.


Nature ◽  
2016 ◽  
Vol 531 (7596) ◽  
pp. 651-655 ◽  
Author(s):  
Wei Yang ◽  
Yibing Bai ◽  
Ying Xiong ◽  
Jin Zhang ◽  
Shuokai Chen ◽  
...  

2020 ◽  
Vol 10 (1) ◽  
Author(s):  
Haiyan Yu ◽  
Hongwei Wu ◽  
Fengping Zheng ◽  
Chengxin Zhu ◽  
Lianghong Yin ◽  
...  

Abstract A detailed understanding of the gene-regulatory network in ankylosing spondylitis (AS) is vital for elucidating the mechanisms of AS pathogenesis. Assaying transposase-accessible chromatin in single cell sequencing (scATAC-seq) is a suitable method for revealing such networks. Thus, scATAC-seq was applied to define the landscape of active regulatory DNA in AS. As a result, there was a significant change in the percent of CD8+ T cells in PBMCs, and 37 differentially accessible transcription factor (TF) motifs were identified. T cells, monocytes-1 and dendritic cells were found to be crucial for the IL-17 signaling pathway and TNF signaling pathway, since they had 73 potential target genes regulated by 8 TF motifs with decreased accessibility in AS. Moreover, natural killer cells were involved in AS by increasing the accessibility to TF motifs TEAD1 and JUN to induce cytokine-cytokine receptor interactions. In addition, CD4+ T cells and CD8+ T cells may be vital for altering host immune functions through increasing the accessibility of TF motifs NR1H4 and OLIG (OLIGI and OLIG2), respectively. These results explain clear gene regulatory variation in PBMCs from AS patients, providing a foundational framework for the study of personal regulomes and delivering insights into epigenetic therapy.


2021 ◽  
Author(s):  
Ramdane Igalouzene ◽  
Hector Hernandez-Vargas ◽  
Nicolas Benech ◽  
David Bauché ◽  
Célia Barrachina ◽  
...  

AbstractSMAD4, a key mediator of TGF-β signaling, plays a crucial role in T cells to prevent chronic gut inflammation. However, the molecular mechanisms underlying this control remain elusive. Using different genetic and epigenetic approaches, we unexpectedly reveal that SMAD4 in CD8 T cells prevents chronic intestinal inflammation by a feedforward mechanism that is TGF-β-independent. Prior to any TGF-β-receptor engagement, SMAD4 acts as an active and basal repressor of epigenetic, transcriptional and functional TGF-β imprinting in CD8 T cells. Thus, in sharp opposition to total TGF-β signaling deletion, SMAD4 deletion impairs naïve CD8 T cell effector predisposition but promotes CD8 T cell accumulation and epithelial retention by promoting their response to IL-7 and their expression of integrins such as Itgae. Besides, SMAD4 deletion unleashes the induction of a wide range of TGF-β-signaling-repressors such as Smad7, Ski, Skil, and Smurf2 and hampers TGF-β-mediated CD8 T cell immunosuppression. Mechanistically, prior to any TGF-β signal, SMAD4 binds to the loci of several TGF-β-target genes, and by regulating histone acetylation, represses their expression. The massive gut epithelial colonization, associated with their escape from the immunoregulatory TGF-β effects overtakes their poor effector preconditioning and elicits microbiota-driven chronic epithelial CD8 T cell activation. Hence, in an anticipatory manner, independently of TGF-β, SMAD4 governs a feedforward regulation of TGF-β effects in CD8 T cells, preventing chronic intestinal inflammation.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2940-2940
Author(s):  
Kohei Hosokawa ◽  
Pawel Muranski ◽  
Xingmin Feng ◽  
Keyvan Keyvanfar ◽  
Danielle M. Townsley ◽  
...  

Abstract Background. Emerging evidence indicates that microRNAs (miRNAs) may regulate immunity. Their role in acquired aplastic anemia (AA), a T cell-mediated autoimmune disease, has not been investigated. We characterized miRNA signatures in patient T cells to identify novel genes involved in the regulation of immune responses of CD4+ and CD8+ T cells in AA. Cells from age-matched healthy donors and patients with myelodysplastic syndrome (MDS) and transfusion-dependent sickle cell disease (SCD) were used for comparison. Method. MiRNAs and mRNAs from flow cytometrically sorted T cells from patients and controls were extracted and subjected to profiling using miRNA PCR array platform (miScript miRNA PCR array, MIHS-111Z, Qiagen Inc). Real-time PCR was employed for validation of specific miRNA expression changes. To define identified miRNA functions, their target genes were predicted using bioinformatic software (TargetScan, miRBase, DIANA-miRPath). MiRNA target gene profilings were performed using custom PCR array platform (Custom PCR array, Qiagen Inc) in CD4+ and CD8+ T cells in AA. MiRNA knockdown (miR-126-3p and/or miR-145-5p) on healthy CD4+ and CD8+ T cells was performed using the Human T Cell Nucleofector kit (Lonza AG). Cell proliferation was estimated by CFSE assays. Effects of miRNA knockdown on the target genes were evaluated by qPCR and immunoblot. MiRNA expression profilings were examined in CD4+ and CD8+ T cells in a mouse model of lymph node cell infusion-induced bone marrow failure (BMF). Results and Discussion. As compared to healthy controls, miRNA expression profiling revealed significant down-regulation of miRNAs in a disease-specific pattern. MiR-199a-5p expression was reduced in CD4+ and CD8+ T cells from patients with all diseases that were tested (AA, MDS, SCD). Reduced expression of miR-223-3p (more than 3 fold change (FC), P <0.05) in T cells was seen in low-risk MDS. Concurrent downregulation of miR-126-3p, miR-145-5p, miR-223-3p and miR-199a-5p (more than 3 FC, P <0.05) was uniquely observed only in CD4+ and CD8+ T cells from AA patients (Figure 1A). Analysis of these four miRNAs in T cells from AA patients after successful immunosuppressive therapy (IST) at 3 or 6 months (N=6) revealed restoration of the expression levels of miR-126-3p and miR-145-5p (more than 2 FC, P <0.05) as compared to those of patients before IST. Bioinformatic analysis predicted MYC, PIK3R2, ETS1, STAT1, CD28, and HIF1A as potential target transcriptomes for these four miRNAs. Gene expression profiling revealed significant up-regulation of MYC in CD4+ T cells (more than 1.5 FC, P <0.05) and PIK3R2 in CD8+ T cells (more than 1.5 FC, P <0.05) in AA patients compared to healthy controls (Figure 1B). qPCR and immunoblot confirmed up-regulation of MYC (~2.0 FC) in CD4+ T cells and PIK3R2 (~2.0 FC) in CD8+ T cells after transfection with anti-miR-145-5p and anti-miR-126-3p, respectively. Knock-down of the miR-126-3p and miR-145-5p promoted proliferation of both CD4+ and CD8+ T cells and increased IFN-γ production in CD8+ T cells, suggesting enhanced effector differentiation and functionality of this subset as a result of reduced activity of both miRNAs. Expression levels of miR-126-3p and miR-145-5p were not decreased in CD4+ and CD8+ T cells in a BMF mouse model. However, expression levels of miR-142-5p and miR-29a-3p, which have been reported as miRNAs associated with allo-reactivity, were decreased (more than 2 FC, P <0.05) in this model, concordant with the mouse models basis in H2 mismatched reactivity. Conclusion. This work describes previously unknown potentially regulatory role of miRNAs 145-5p and 126-3p in T cell activation in AA. Further investigation is warranted to determine the role of these miRNAs in the more general pathogenesis of autoimmune diseases. Understanding these novel pathways might offer new therapeutic strategies and a novel mechanistic insight into the epigenetic control of human T cell effector responses. Figure 1 Figure 1. Disclosures Townsley: GSK: Research Funding.


Cancers ◽  
2021 ◽  
Vol 13 (20) ◽  
pp. 5249
Author(s):  
Jason Roszik ◽  
Lisa Maria Mustachio ◽  
John A. Livingston ◽  
Roman Groisberg ◽  
Roberto Carmagnani Pestana ◽  
...  

Soft tissue sarcomas, depending on the subtype and grade, frequently recur and become metastatic after localized treatment. There is now great interest in applying immunotherapy to sarcomas to immuno-profile the different subtypes and immune monitor for prognosis. Our group previously showed that key immunotherapy target genes are present in sarcomas. Here, we extend our findings by demonstrating that sarcomas with a relatively high mutational load are likely to be more sensitive to immunotherapy compared to sarcomas with a lower mutation load. We also show that sarcomas with a higher mutation load are associated with the expression of key immune-related genes. We found that CD8+ T cells are present in sarcoma subtypes and that PD-L2 is highly expressed. These findings further define potential mechanisms behind the immunotherapy response of specific sarcoma subtypes and can be used to develop more optimal treatments in the future.


2015 ◽  
Vol 29 (9) ◽  
pp. 1269-1285 ◽  
Author(s):  
Paul Childress ◽  
Keith R. Stayrook ◽  
Marta B. Alvarez ◽  
Zhiping Wang ◽  
Yu Shao ◽  
...  

Abstract PTH is an osteoanabolic for treating osteoporosis but its potency wanes. Disabling the transcription factor nuclear matrix protein 4 (Nmp4) in healthy, ovary-intact mice enhances bone response to PTH and bone morphogenetic protein 2 and protects from unloading-induced osteopenia. These Nmp4−/− mice exhibit expanded bone marrow populations of osteoprogenitors and supporting CD8+ T cells. To determine whether the Nmp4−/− phenotype persists in an osteoporosis model we compared PTH response in ovariectomized (ovx) wild-type (WT) and Nmp4−/− mice. To identify potential Nmp4 target genes, we performed bioinformatic/pathway profiling on Nmp4 chromatin immunoprecipitation sequencing (ChIP-seq) data. Mice (12 w) were ovx or sham operated 4 weeks before the initiation of PTH therapy. Skeletal phenotype analysis included microcomputed tomography, histomorphometry, serum profiles, fluorescence-activated cell sorting and the growth/mineralization of cultured WT and Nmp4−/− bone marrow mesenchymal stem progenitor cells (MSPCs). ChIP-seq data were derived using MC3T3-E1 preosteoblasts, murine embryonic stem cells, and 2 blood cell lines. Ovx Nmp4−/− mice exhibited an improved response to PTH coupled with elevated numbers of osteoprogenitors and CD8+ T cells, but were not protected from ovx-induced bone loss. Cultured Nmp4−/− MSPCs displayed enhanced proliferation and accelerated mineralization. ChIP-seq/gene ontology analyses identified target genes likely under Nmp4 control as enriched for negative regulators of biosynthetic processes. Interrogation of mRNA transcripts in nondifferentiating and osteogenic differentiating WT and Nmp4−/− MSPCs was performed on 90 Nmp4 target genes and differentiation markers. These data suggest that Nmp4 suppresses bone anabolism, in part, by regulating IGF-binding protein expression. Changes in Nmp4 status may lead to improvements in osteoprogenitor response to therapeutic cues.


Sign in / Sign up

Export Citation Format

Share Document