Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 135
Filter
1.
Database (Oxford) ; 20242024 Jul 06.
Article in English | MEDLINE | ID: mdl-38970503

ABSTRACT

The relationship between different ribonucleic acids (RNAs) and tumor immunity has been widely investigated. However, a systematic description of tumor immune-related RNAs in different tumors is still lacking. We collected the relationship of tumor immune-related RNAs from the published literature and presented them in a user-friendly interface, "ImmRNA" (http://www.immrna.cn/), to provide a resource to study immune-RNA-cancer regulatory relations. The ImmRNA contains 49 996 curated entries. Each entry includes gene symbols, gene types, target genes, downstream effects, functions, immune cells, and other information. By rearranging and reanalyzing the data, our dataset contains the following key points: (i) providing the links between RNAs and the immune in cancers, (ii) displaying the downstream effects and functions of RNAs, (iii) listing immune cells and immune pathways related to RNA function, (iv) showing the relationship between RNAs and prognostic outcomes, and (v) exhibiting the experimental methods described in the article. ImmRNA provides a valuable resource for understanding the functions of tumor immune-related RNAs. Database URL:  http://www.immrna.cn/.


Subject(s)
Neoplasms , Humans , Neoplasms/genetics , Neoplasms/immunology , Databases, Genetic , Databases, Nucleic Acid , RNA, Neoplasm/genetics , RNA, Neoplasm/immunology , RNA/genetics , RNA/immunology
2.
Mol Cell Biochem ; 477(4): 1281-1293, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35129780

ABSTRACT

Triple-Negative Breast Cancer (TNBC) is one of the most aggressive and hot BC subtypes. Our research group has recently shed the light on the utility of natural compounds as effective immunotherapeutic agents. The aim of this study is to investigate the role of a methoxylated quercetin glycoside (MQG) isolated from Cleome droserifolia in harnessing TNBC progression and tuning the tumor microenvironment and natural killer cells cytotoxicity. Results showed that MQG showed the highest potency (IC50 = 12 µM) in repressing cellular proliferation, colony-forming ability, migration, and invasion capacities. Mechanistically, MQG was found to modulate a circuit of competing endogenous RNAs where it was found to reduce the oncogenic MALAT-1 lncRNA and induce TP53 and its downstream miRNAs; miR-155 and miR-146a. Accordingly, this leads to alteration in several downstream signaling pathways such as nitric oxide synthesizing machinery, natural killer cells' cytotoxicity through inducing the expression of its activating ligands such as MICA/B, ULBP2, CD155, and ICAM-1 and trimming of the immune-suppressive cytokines such as TNF-α and IL-10. In conclusion, this study shows that MQG act as a compelling anti-cancer agent repressing TNBC hallmarks, activating immune cell recognition, and alleviating the immune-suppressive tumor microenvironment experienced by TNBC patients.


Subject(s)
Glycosides/pharmacology , MicroRNAs/immunology , RNA, Long Noncoding/immunology , RNA, Neoplasm/immunology , Triple Negative Breast Neoplasms/immunology , Tumor Suppressor Protein p53/immunology , Female , Humans , MCF-7 Cells , MicroRNAs/genetics , RNA, Long Noncoding/genetics , RNA, Neoplasm/genetics , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Tumor Suppressor Protein p53/genetics
3.
Int J Mol Sci ; 22(18)2021 Sep 11.
Article in English | MEDLINE | ID: mdl-34576006

ABSTRACT

Tumor immune escape is a common process in the tumorigenesis of non-small cell lung cancer (NSCLC) cells where programmed death ligand-1 (PD-L1) expression, playing a vital role in immunosuppression activity. Additionally, epidermal growth factor receptor (EGFR) phosphorylation activates Janus kinase-2 (JAK2) and signal transduction, thus activating transcription 3 (STAT3) to results in the regulation of PD-L1 expression. Chemotherapy with commercially available drugs against NSCLC has struggled in the prospect of adverse effects. Nobiletin is a natural flavonoid isolated from the citrus peel that exhibits anti-cancer activity. Here, we demonstrated the role of nobiletin in evasion of immunosuppression in NSCLC cells by Western blotting and real-time polymerase chain reaction methods for molecular signaling analysis supported by gene silencing and specific inhibitors. From the results, we found that nobiletin inhibited PD-L1 expression through EGFR/JAK2/STAT3 signaling. We also demonstrated that nobiletin exhibited p53-independent PD-L1 suppression, and that miR-197 regulates the expression of STAT3 and PD-L1, thereby enhancing anti-tumor immunity. Further, we evaluated the combination ability of nobiletin with an anti-PD-1 monoclonal antibody in NSCLC co-culture with peripheral blood mononuclear cells. Similarly, we found that nobiletin assisted the induction of PD-1/PD-L1 blockade, which is a key factor for the immune escape mechanism. Altogether, we propose nobiletin as a modulator of tumor microenvironment for cancer immunotherapy.


Subject(s)
B7-H1 Antigen/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Flavones/pharmacology , Lung Neoplasms/immunology , MicroRNAs/immunology , Neoplasm Proteins/immunology , RNA, Neoplasm/immunology , STAT3 Transcription Factor/immunology , Signal Transduction/drug effects , Tumor Escape/drug effects , A549 Cells , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Signal Transduction/immunology
4.
Immunol Res ; 69(6): 558-567, 2021 12.
Article in English | MEDLINE | ID: mdl-34559381

ABSTRACT

Approximately 25% of breast cancer (BC) patients are HER2-positive. Trastuzumab is used as a targeted therapy drug to treat HER2-positive BC patients; however, the drug resistance remains a big challenge. Circular RNAs (circRNAs) are reported to be involved in drug resistance, but the role of circ_0001598 has never been studied in BC. First, we identified the expression of circ_0001598 by RT-qPCR in BC. The gain-of-function and loss-of-function studies were applied to study the functional roles of circ_0001598 and its target gene. We observed upregulation of circ_0001598 in BC tissues, especially in trastuzumab-resistant BC samples. We further identified that miR-1184 is a functional target of circ_0001598. Moreover, it was found that programmed death-ligand 1 (PD-L1) was a direct target of miR-1184. The oncogenic effects of circ_0001598 in promoting BC cell growth, trastuzumab-resistance, PD-L1 expression, and escaping of CD8 T cell killing were abolished after the restoration of miR-1184. In conclusion, we demonstrate that circ_0001598/miR-1184/PD-L1 signaling plays a crucial role in the regulation of BC progression and trastuzumab-resistance phonotypes, which suggests that circ_0001598 may be a molecular target to treat HER2-positive BC patients.


Subject(s)
B7-H1 Antigen/immunology , Breast Neoplasms/immunology , Drug Resistance, Neoplasm/immunology , MicroRNAs/immunology , Neoplasm Proteins/immunology , RNA, Circular/immunology , RNA, Neoplasm/immunology , Trastuzumab/pharmacology , Tumor Escape , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Female , Humans
5.
Front Immunol ; 12: 697026, 2021.
Article in English | MEDLINE | ID: mdl-34526985

ABSTRACT

N6-methylation of adenosine (m6A), a post-transcriptional regulatory mechanism, is the most abundant nucleotide modification in almost all types of RNAs. The biological function of m6A in regulating the expression of oncogenes or tumor suppressor genes has been widely investigated in various cancers. However, recent studies have addressed a new role of m6A modification in the anti-tumor immune response. By modulating the fate of targeted RNA, m6A affects tumor-associated immune cell activation and infiltration in the tumor microenvironment (TME). In addition, m6A-targeting is found to affect the efficacy of classical immunotherapy, which makes m6A a potential target for immunotherapy. Although m6A modification together with its regulators may play the exact opposite role in different tumor types, targeting m6A regulators has been shown to have wide implications in several cancers. In this review, we discussed the link between m6A modification and tumor with an emphasis on the importance of m6A in anti-tumor immune response and immunotherapy.


Subject(s)
Adenosine/analogs & derivatives , Antineoplastic Agents/therapeutic use , Immunotherapy , Neoplasms/drug therapy , RNA, Neoplasm/metabolism , Tumor Microenvironment , Adenosine/genetics , Adenosine/immunology , Adenosine/metabolism , Animals , Gene Expression Regulation, Neoplastic , Humans , Molecular Targeted Therapy , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/immunology , Tumor Microenvironment/immunology
6.
Clin Exp Immunol ; 206(1): 36-46, 2021 10.
Article in English | MEDLINE | ID: mdl-34195995

ABSTRACT

The recurrence of patients with epithelial ovarian cancer (EOC) is largely attributed to tumour cells escaping from the surveillance of immune cells. However, to date there is a lack of studies that have systematically evaluated the associations between the infiltration fraction of immune cells and the recurrence risk of EOC. Based on the micro-ribonucleic acid (microRNA) expression profiles of 441 EOC patients, we constructed a microRNA-based panel with recurrence prediction potential using non-negative matrix factorization consensus clustering. Then, we evaluated the association between recurrence risk and infiltration proportions among 10 immune cell types by CIBERSORT and a multivariable Cox regression model. As a result, we identified a 72-microRNA-based panel that could stratify patients into high and low risk of recurrence. The infiltration of plasma cells and M1 macrophages was consistently significantly associated with the risk of recurrence in patients with EOC. Plasma cells were significantly associated with a decreased risk of relapse [hazard ratio (HR) = 0.58, p = 0.006), while M1 macrophages were associated with an increased risk of relapse (HR = 1.59, p = 0.003). Therefore, the 72-microRNA-based panel, M1 macrophages and plasma cells may hold potential to serve as recurrence predictors of EOC patients in clinical practice.


Subject(s)
Carcinoma, Ovarian Epithelial , Lymphocytes, Tumor-Infiltrating/immunology , Models, Immunological , Neoplasm Recurrence, Local , Ovarian Neoplasms , Plasma Cells/immunology , Carcinoma, Ovarian Epithelial/diagnosis , Carcinoma, Ovarian Epithelial/immunology , Female , Gene Expression Regulation, Neoplastic/immunology , Humans , MicroRNAs/immunology , Middle Aged , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/immunology , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/immunology , Prognosis , RNA, Neoplasm/immunology
7.
Front Immunol ; 12: 641507, 2021.
Article in English | MEDLINE | ID: mdl-33777035

ABSTRACT

RNA methylation is a kind of RNA modification that exists widely in eukaryotes and prokaryotes. RNA methylation occurs not only in mRNA but also in ncRNA. According to the different sites of methylation, RNA methylation includes m6A, m5C, m7G, and 2-O-methylation modifications. Modifications affect the splicing, nucleation, stability and immunogenicity of RNA. RNA methylation is involved in many physiological and pathological processes. In the immune system, especially for tumor immunity, RNA methylation affects the maturation and response function of immune cells. Through the influence of RNA immunogenicity and innate immune components, modifications regulate the innate immunity of the body. Some recent studies verified that RNA methylation can regulate tumor immunity, which also provides a new idea for the future of treating immunological diseases and tumor immunotherapy.


Subject(s)
Immunity, Innate , Neoplasms/immunology , RNA Splicing/immunology , RNA, Messenger/immunology , RNA, Neoplasm/immunology , Animals , Humans , Methylation , Neoplasms/pathology
8.
Comput Math Methods Med ; 2021: 6676692, 2021.
Article in English | MEDLINE | ID: mdl-33727952

ABSTRACT

A common cancer in females, breast cancer (BRCA) mortality has been recently reduced; however, the prognosis of BRCA patients remains poor. This study attempted to develop prognostic immune-related long noncoding RNAs (lncRNAs) for BRCA and identify the effects of these lncRNAs on the tumor microenvironment (TME). Gene expression data from The Cancer Genome Atlas (TCGA) database were collected in order to select differentially expressed lncRNAs. Immune-related lncRNAs were downloaded from the ImmLnc database, where 316 immune-related lncRNAs were identified, 12 of which were found to be significantly related to the prognosis of BRCA patients. Multivariate cox regression analysis was then applied to construct prognostic immune-related lncRNAs as the risk model, including C6orf99, LINC00987, SIAH2-AS1, LINC01010, and ELOVL2-AS1. High-risk and low-risk groups were distinguished according to the median of immune-related risk scores. Accordingly, the overall survival (OS) in the high-risk group was observed to be shorter than that in the low-risk group. qRT-PCR analysis demonstrated that lncRNA expression levels in BRCA cell lines were in basic agreement with predictions except for LINC00987. By validating numerous clinical samples, lncRNA C6orf99 was shown to be highly expressed in the advanced stage, while LINC01010 and SIAH2-AS1 decreased in the advanced T-stage and M-stage. Moreover, the expression of LINC0098 was found to be significantly decreased among the groups (>50 years old). Gene set enrichment analysis (GSEA) was applied to analyze the cancer hallmarks and immunological characteristics of the high-risk and low-risk groups. Importantly, the TIMER database demonstrated that this immune-related lncRNA risk model for breast cancer is related to the infiltration of immune cells. In conclusion, the results indicated that five immune-related lncRNAs could be used as a prognostic model and may even accelerate immunotherapy for BRCA patients.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/immunology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/immunology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Cell Line, Tumor , Computational Biology , Databases, Genetic , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Genes, myc , Glycolysis/genetics , Glycolysis/immunology , Humans , Kaplan-Meier Estimate , Lymphocytes, Tumor-Infiltrating/immunology , Oxidative Phosphorylation , Prognosis , Proportional Hazards Models , RNA, Neoplasm/genetics , RNA, Neoplasm/immunology , Risk Factors , T-Lymphocyte Subsets/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
9.
Front Immunol ; 12: 762243, 2021.
Article in English | MEDLINE | ID: mdl-35069534

ABSTRACT

Background: The role of RNA N6-methyladenosine (m6A) modification in tumor progression and metastasis has been demonstrated. Nonetheless, potential biological function of m6A modification patterns in nasopharyngeal carcinoma (NPC) remains unknown. Methods: The m6A modification patterns were comprehensively evaluated based on 26 m6A regulators in NPC, and m6A subtype and also m6A score were identified and systematically correlated with representative tumor characteristics. Results: Two distinct m6A subtypes were determined and were highly consistent with immune activated and immune suppressed phenotypes, respectively. More representative m6A scores of individual tumors could predict tumor microenvironment (TME) infiltration, mRNA based stemness index (mRNAsi), EBV gene expression, genetic variation, and prognosis of NPC patients. Low m6A score, characterized by activation of immunity and suppression of mRNAsi and EBV gene, indicated an activated TME phenotype and better PFS and also lower risk of recurrence and metastasis. High m6A score, characterized by activation of Wnt and NF-κB signaling pathway and lack of effective immune infiltration, indicated an immune suppressed TME phenotype and poorer survival. Low m6A score was also correlated with increased tumor mutation burden (TMB) and better response to immunotherapy, and vice versa. A significant therapeutic advantage in patients with low m6A score was confirmed with an anti-PDL1 immunotherapy cohort. Conclusions: m6A patterns played an important role in the diversity and complexity of TME. m6A score could be used to evaluate the m6A pattern of individual tumor to enhance our understanding of TME infiltration and guide more effective immunotherapy strategies.


Subject(s)
Adenosine/analogs & derivatives , Nasopharyngeal Carcinoma/immunology , Nasopharyngeal Neoplasms/immunology , RNA, Neoplasm/immunology , Tumor Microenvironment/immunology , Wnt Signaling Pathway/immunology , Adenosine/immunology , Humans , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Carcinoma/therapy , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/therapy , Neoplasm Metastasis
10.
Int J Mol Sci ; 21(22)2020 Nov 19.
Article in English | MEDLINE | ID: mdl-33228245

ABSTRACT

Extracellular vesicles (EVs) are a heterogeneous group of cell-derived submicron vesicles released under physiological or pathological conditions. EVs mediate the cellular crosstalk, thus contributing to defining the tumor microenvironment, including in epithelial ovarian cancer (EOC). The available literature investigating the role of EVs in EOC has been reviewed following PRISMA guidelines, focusing on the role of EVs in early disease diagnosis, metastatic spread, and the development of chemoresistance in EOC. Data were identified from searches of Medline, Current Contents, PubMed, and from references in relevant articles from 2010 to 1 April 2020. The research yielded 194 results. Of these, a total of 36 papers, 9 reviews, and 27 original types of research were retained and analyzed. The literature findings demonstrate that a panel of EV-derived circulating miRNAs may be useful for early diagnosis of EOC. Furthermore, it appears clear that EVs are involved in mediating two crucial processes for metastatic and chemoresistance development: the epithelial-mesenchymal transition, and tumor escape from the immune system response. Further studies, more focused on in vivo evidence, are urgently needed to clarify the role of EV assessment in the clinical management of EOC patients.


Subject(s)
Carcinoma, Ovarian Epithelial/genetics , Epithelial-Mesenchymal Transition/genetics , Extracellular Vesicles/genetics , MicroRNAs/genetics , Ovarian Neoplasms/genetics , RNA, Neoplasm/genetics , Antineoplastic Agents/therapeutic use , Carcinoma, Ovarian Epithelial/diagnosis , Carcinoma, Ovarian Epithelial/drug therapy , Carcinoma, Ovarian Epithelial/immunology , Cell Communication/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Early Detection of Cancer , Epithelial-Mesenchymal Transition/immunology , Extracellular Vesicles/immunology , Extracellular Vesicles/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Lymphatic Metastasis , MicroRNAs/immunology , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/immunology , RNA, Neoplasm/immunology , Signal Transduction , Tumor Escape , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
11.
Arch Biochem Biophys ; 695: 108583, 2020 11 30.
Article in English | MEDLINE | ID: mdl-32956633

ABSTRACT

miRNAs are short non-coding RNA molecules that regulate the expression of mRNA post-transcriptionally. MiRNAs that are secreted into the circulation, also termed circulating miRNAs, have been studied extensively for their roles in diagnosis, treatment and prognosis of human breast cancer. Breast cancer is the most prevalent female cancer and is associated with key cancer hallmarks including sustained proliferation, evasion of apoptosis, increased invasion, enhanced metastases, initation of inflammation, induction of angiogenesis, metabolic derangement and immune dysregulation. This review aimed to explore the relationships between circulating miRNAs and different breast cancer hallmarks. Besides, the advantages, challenges and clinical application of using circulating miRNAs in human breast cancer management were also discussed.


Subject(s)
Apoptosis , Breast Neoplasms/blood , Cell Proliferation , Circulating MicroRNA/blood , Neovascularization, Pathologic/blood , RNA, Neoplasm/blood , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Circulating MicroRNA/immunology , Female , Humans , Inflammation/blood , Inflammation/immunology , Inflammation/pathology , Inflammation/therapy , Neoplasm Invasiveness , Neoplasm Metastasis , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/therapy , RNA, Neoplasm/immunology
12.
Exp Hematol ; 90: 30-38, 2020 10.
Article in English | MEDLINE | ID: mdl-32827560

ABSTRACT

Germinal center-associated nuclear protein (GANP) is a unique and multifunctional protein that plays a critical role in cell biology, neurodegenerative disorders, immunohematology, and oncogenesis. GANP is an orthologue of Saccharomyces Sac3, one of the components of the transcription export 2 (TREX-2) complex and a messenger RNA (mRNA) nuclear export factor. GANP is widely conserved in all mammals, including humans. Although GANP was originally discovered as a molecule upregulated in the germinal centers of secondary lymphoid follicles in peripheral lymphoid organs, it is expressed ubiquitously in many tissues. It serves numerous functions, including making up part of the mammalian TREX-2 complex; mRNA nuclear export via nuclear pores; prevention of R-loop formation, genomic instability, and hyper-recombination; and B-cell affinity maturation. In this review, we first overview the extensive analyses that have revealed the basic functions of GANP and its ancestor molecule Sac3, including mRNA nuclear export and regulation of R-loop formation. We then describe how aberrant expression of GANP is significantly associated with cancer development. Moreover, we discuss a crucial role for GANP in B-cell development, especially affinity maturation in germinal centers. Finally, we illustrate that overexpression of GANP in B cells leads to lymphomagenesis resembling Hodgkin lymphoma derived from germinal center B cells, and that GANP may be involved in transdifferentiation of B cells to macrophages, which strongly affects Hodgkin lymphomagenesis.


Subject(s)
Acetyltransferases/immunology , Carcinogenesis/immunology , Hematologic Neoplasms/immunology , Hodgkin Disease/immunology , Intracellular Signaling Peptides and Proteins/immunology , Neoplasm Proteins/immunology , Active Transport, Cell Nucleus/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Carcinogenesis/pathology , Cell Transdifferentiation/immunology , Germinal Center/immunology , Germinal Center/pathology , Hematologic Neoplasms/pathology , Hodgkin Disease/pathology , Humans , Macrophages/immunology , Macrophages/pathology , RNA, Messenger/immunology , RNA, Neoplasm/immunology
13.
Med Sci Monit ; 26: e924604, 2020 Aug 02.
Article in English | MEDLINE | ID: mdl-32740646

ABSTRACT

BACKGROUND Increasing studies have shown the important clinical role of immune and stromal cells in gastric cancer microenvironment. Based on information of immune and stromal cells in The Cancer Genome Atlas, this study aimed to construct a prognostic risk assessment model for gastric cancer. MATERIAL AND METHODS Based on the immune/structural scores, differentially expressed genes (DEGs) were filtered and analyzed. Afterwards, DEGs associated with prognosis were screened and the risk assessment model was constructed in the training set. Moreover, the validity of the model was verified both in the testing set and the overall sample. RESULTS In this study, patients were divided into high-score and low-score groups based on immune/stromal score, and 919 DEGs were identified. By applying least absolute shrinkage and selection operator (LASSO) and Cox analysis, 10 mRNAs were selected to form a prognostic risk assessment model, risk score=(0.294*SLC17A9) + (-0.477*FERMT3) + (0.866*NRP1) + (0.350*MMRN1) + (0.381*RNASE1) + (0.189*TRIB3) + (0.230*PGAP3) + (0.087*MAGEA3) + (0.182*TACR2) + (0.368*CYP51A1). In the training set, the low-risk group divided by the model was found to have better overall survival, and the prediction efficiency of the model was demonstrated to be good. Multivariate Cox analysis indicated that the model could work as a prognostic factor independently. Similar results were shown in the testing group and overall patients cohort group. Finally, the risk assessment model and other clinical variables were integrated to construct a nomogram. CONCLUSIONS In general, this study constructs a prognostic risk assessment model for gastric cancer, which could improve the prognosis stratification of patients combined with other clinical indicators.


Subject(s)
Adenocarcinoma/genetics , Biomarkers, Tumor/genetics , Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , RNA, Neoplasm/genetics , Stomach Neoplasms/genetics , Adenocarcinoma/diagnosis , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Aged , Area Under Curve , Biomarkers, Tumor/immunology , Carcinogenesis/immunology , Carcinogenesis/pathology , Databases, Genetic , Female , Gene Expression Profiling , Gene Regulatory Networks , Humans , Immunity, Innate , Male , Middle Aged , Models, Genetic , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasm Staging , Prognosis , RNA, Messenger/genetics , RNA, Messenger/immunology , RNA, Neoplasm/immunology , ROC Curve , Stomach Neoplasms/diagnosis , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Stromal Cells/immunology , Stromal Cells/pathology , Survival Analysis , Transcriptome , Tumor Microenvironment/genetics
14.
Blood ; 136(24): 2812-2823, 2020 12 10.
Article in English | MEDLINE | ID: mdl-32730593

ABSTRACT

Somatic gene mutations are key determinants of outcome in patients with myelodysplastic syndromes (MDS) and secondary AML (sAML). In particular, patients with TP53 mutations represent a distinct molecular cohort with uniformly poor prognosis. The precise pathogenetic mechanisms underlying these inferior outcomes have not been delineated. In this study, we characterized the immunological features of the malignant clone and alterations in the immune microenvironment in patients with TP53-mutant and wild-type MDS or sAML. Notably, PDL1 expression is significantly increased in hematopoietic stem cells of patients with TP53 mutations, which is associated with MYC upregulation and marked downregulation of MYC's negative regulator miR-34a, a p53 transcription target. Notably, patients with TP53 mutations display significantly reduced numbers of bone marrow-infiltrating OX40+ cytotoxic T cells and helper T cells, as well as decreased ICOS+ and 4-1BB+ natural killer cells. Further, highly immunosuppressive regulatory T cells (Tregs) (ie, ICOShigh/PD-1-) and myeloid-derived suppressor cells (PD-1low) are expanded in cases with TP53 mutations. Finally, a higher proportion of bone marrow-infiltrating ICOShigh/PD-1- Treg cells is a highly significant independent predictor of overall survival. We conclude that the microenvironment of TP53 mutant MDS and sAML has an immune-privileged, evasive phenotype that may be a primary driver of poor outcomes and submit that immunomodulatory therapeutic strategies may offer a benefit for this molecularly defined subpopulation.


Subject(s)
Leukemia, Myeloid, Acute , Mutation , Myelodysplastic Syndromes , Myeloid-Derived Suppressor Cells/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Suppressor Protein p53 , Adult , Aged , Aged, 80 and over , Female , Humans , Immunosuppression Therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/pathology , Male , MicroRNAs/genetics , MicroRNAs/immunology , Middle Aged , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/immunology , Myelodysplastic Syndromes/pathology , Myeloid-Derived Suppressor Cells/pathology , RNA, Neoplasm/genetics , RNA, Neoplasm/immunology , T-Lymphocytes, Regulatory/pathology , Tumor Suppressor Protein p53/immunology
15.
J Leukoc Biol ; 107(6): 917-932, 2020 06.
Article in English | MEDLINE | ID: mdl-32272497

ABSTRACT

T cells are crucial for the success of immune-based cancer therapy. Reinvigorating antitumor T cell activity by blocking checkpoint inhibitory receptors has provided clinical benefits for many cancer patients. However, the efficacy of these treatments varies in cancer patients and the mechanisms underlying these diverse responses remain elusive. The density and status of tumor-infiltrating T cells have been shown to positively correlate with patient response to checkpoint blockades. Therefore, further understanding of the heterogeneity, clonal expansion, migration, and effector functions of tumor-infiltrating T cells will provide fundamental insights into antitumor immune responses. To this end, recent advances in single-cell RNA sequencing technology have enabled profound and extensive characterization of intratumoral immune cells and have improved our understanding of their dynamic relationships. Here, we summarize recent progress in single-cell RNA sequencing technology and current strategies to uncover heterogeneous tumor-infiltrating T cell subsets. In particular, we discuss how the coupling of deep transcriptome information with T cell receptor (TCR)-based lineage tracing has furthered our understanding of intratumoral T cell populations. We also discuss the functional implications of various T cell subsets in tumors and highlight the identification of novel T cell markers with therapeutic or prognostic potential.


Subject(s)
Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms/immunology , RNA, Neoplasm/genetics , T-Lymphocyte Subsets/immunology , Transcriptome , Cell Communication/genetics , Cell Communication/immunology , Gene Expression , Genetic Heterogeneity , Humans , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/pathology , Neoplasms/genetics , Neoplasms/pathology , RNA, Neoplasm/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Sequence Analysis, RNA , Single-Cell Analysis , T-Lymphocyte Subsets/classification , T-Lymphocyte Subsets/pathology , Tumor Microenvironment/immunology
16.
Sci Immunol ; 5(44)2020 02 21.
Article in English | MEDLINE | ID: mdl-32086381

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are innate immune cells that acquire the capacity to suppress adaptive immune responses during cancer. It remains elusive how MDSCs differ from their normal myeloid counterparts, which limits our ability to specifically detect and therapeutically target MDSCs during cancer. Here, we sought to determine the molecular features of breast cancer-associated MDSCs using the widely studied mouse model based on the mouse mammary tumor virus (MMTV) promoter-driven expression of the polyomavirus middle T oncoprotein (MMTV-PyMT). To identify MDSCs in an unbiased manner, we used single-cell RNA sequencing to compare MDSC-containing splenic myeloid cells from breast tumor-bearing mice with wild-type controls. Our computational analysis of 14,646 single-cell transcriptomes revealed that MDSCs emerge through an aberrant neutrophil maturation trajectory in the spleen that confers them an immunosuppressive cell state. We establish the MDSC-specific gene signature and identify CD84 as a surface marker for improved detection and enrichment of MDSCs in breast cancers.


Subject(s)
Breast Neoplasms/pathology , Myeloid-Derived Suppressor Cells/pathology , Single-Cell Analysis , Transcriptome , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Breast Neoplasms/immunology , Cell Differentiation/genetics , Female , Humans , Mice , Mice, Inbred Strains , Mice, Transgenic , Myeloid-Derived Suppressor Cells/immunology , RNA, Neoplasm/genetics , RNA, Neoplasm/immunology , Signaling Lymphocytic Activation Molecule Family/genetics , Signaling Lymphocytic Activation Molecule Family/immunology
17.
Cancer Biomark ; 27(2): 265-275, 2020.
Article in English | MEDLINE | ID: mdl-31929144

ABSTRACT

BACKGROUND: Tumor-infiltrating immune cells are indispensable to the progression and prognosis of clear cell renal cell carcinoma (ccRCC). OBJECTIVE: The aim of this study was to explore the clinical implications of immune cell infiltrates in ccRCC. METHODS: The Cancer Genome Atlas (TCGA) database (N= 515) and E-MTAB-1980 cohort of patients (N= 101) were adopted to estimate the prognostic value of immune cell infiltration. Twenty-four types of immune cells were evaluated using single-sample gene set enrichment analysis. Cox regression analyses were conducted to develop an immune risk score. RESULTS: Survival analyses revealed that 13 genes significantly associated with the overall survival (OS). Furthermore, multivariate Cox analysis identified an immune risk score on the basis of mast cells, natural killer CD56bright cells, T helper 17 (Th17) cells, and Th2 cells. The immune risk score was associated with OS, with hazard ratios of 2.72 (95% CI 2.17-3.40) and 3.24 (95% CI 1.64-6.44) in TCGA and E-MTAB-1980 datasets, respectively. This immune risk score was significantly correlated with some immunotherapy-related biomarkers. CONCLUSIONS: We profiled a prognostic signature and established an immune risk score model for ccRCC, which could provide novel predictive markers for patients with ccRCC and an indicator for immunotherapy response measurement.


Subject(s)
Carcinoma, Renal Cell/immunology , Kidney Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Carcinoma, Renal Cell/classification , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cohort Studies , Female , Gene Expression Profiling , Humans , Kidney Neoplasms/classification , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Male , Middle Aged , Prognosis , RNA, Neoplasm/genetics , RNA, Neoplasm/immunology , Survival Rate
18.
J Immunol ; 203(7): 1897-1908, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31484735

ABSTRACT

Sepsis is characterized as life-threatening organ dysfunction caused by a dysregulated host immune response to infection. The purpose of this investigation was to determine the differential effect of sepsis on innate versus adaptive immunity, in humans, by examining RNA expression in specific immune cell subsets, including monocytes/macrophages and CD4 and CD8 T cells. A second aim was to determine immunosuppressive mechanisms operative in sepsis that might be amenable to immunotherapy. Finally, we examined RNA expression in peripheral cells from critically ill nonseptic patients and from cancer patients to compare the unique immune response in these disorders with that occurring in sepsis. Monocytes, CD4 T cells, and CD8 T cells from septic patients, critically ill nonseptic patients, patients with metastatic colon cancer, and healthy controls were analyzed by RNA sequencing. Sepsis induced a marked phenotypic shift toward downregulation of multiple immune response pathways in monocytes suggesting that impaired innate immunity may be fundamental to the immunosuppression that characterizes the disorder. In the sepsis cohort, there was a much more pronounced effect on gene transcription in CD4 T cells than in CD8 T cells. Potential mediators of sepsis-induced immunosuppression included Arg-1, SOCS-1, and SOCS-3, which were highly upregulated in multiple cell types. Multiple negative costimulatory molecules, including TIGIT, Lag-3, PD-1, and CTLA-4, were also highly upregulated in sepsis. Although cancer had much more profound effects on gene transcription in CD8 T cells, common immunosuppressive mechanisms were present in all disorders, suggesting that immunoadjuvant therapies that are effective in one disease may also be efficacious in the others.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Gene Expression Regulation, Neoplastic/immunology , Monocytes/immunology , Neoplasms/immunology , RNA, Neoplasm/immunology , Sepsis/immunology , Sequence Analysis, RNA , Adult , Aged , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Critical Illness , Female , Humans , Immune Tolerance , Male , Middle Aged , Monocytes/pathology , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasms/genetics , Neoplasms/pathology , Prospective Studies , RNA, Neoplasm/genetics , Sepsis/genetics , Sepsis/pathology
19.
Front Immunol ; 10: 1734, 2019.
Article in English | MEDLINE | ID: mdl-31404149

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are a heterogeneous cell population with potent immunosuppressive functions. They play major roles in cancer and many of the pathologic conditions associated with inflammation. Long non-coding RNAs (lncRNAs) are untranslated functional RNA molecules. The lncRNAs are involved in the control of a wide variety of cellular processes and are dysregulated in different diseases. They can participate in the modulation of immune function and activity of inflammatory cells, including MDSCs. This mini review focuses on the emerging role of lncRNAs in MDSC activity. We summarize how lncRNAs modulate the generation, recruitment, and immunosuppressive functions of MDSCs and the underlying mechanisms.


Subject(s)
Inflammation/immunology , Myeloid-Derived Suppressor Cells/immunology , Neoplasms/immunology , RNA, Long Noncoding/physiology , Animals , CCAAT-Enhancer-Binding Protein-beta/physiology , Cell Lineage , Epigenesis, Genetic , Forecasting , Gene Expression Regulation, Neoplastic/immunology , Humans , Immunotherapy , Mice , Myeloid-Derived Suppressor Cells/classification , Neoplasms/genetics , Neoplasms/therapy , Nitric Oxide/metabolism , Pseudogenes , RNA, Long Noncoding/genetics , RNA, Long Noncoding/immunology , RNA, Neoplasm/immunology , RNA, Neoplasm/physiology , Reactive Oxygen Species/metabolism , Tumor Escape , Tumor Microenvironment
20.
Sci Rep ; 9(1): 2855, 2019 02 27.
Article in English | MEDLINE | ID: mdl-30814560

ABSTRACT

Tumor cells tend to behave differently in response to immune selective conditions. Contrary to those in therapeutic antitumor conditions, tumor cells in prophylactic antitumor conditions lose antigen expression for antitumor immune escape. Here, using a CT26/HER2 tumor model, we investigate the underlying mechanism(s). We selected tumor cell variants (CT26/HER2-A1 and -A2) displaying resistance to antitumor protective immunity and loss of HER2 antigen expression. These immune-resistant cells failed to induce Ag-specific IgG and IFN-γ responses while forming tumors at the same rate as CT26/HER2 cells. RT-PCR, qRT-PCR, PCR, Western blot and DNA sequencing analyses demonstrated that HER2 expression was inhibited at the post-transcriptional level in these immune-resistant cells, suggesting that tumor cells may escape antitumor immunity through the post-transcriptional regulation of antigen gene expression. The proteasome and lysosomal protein degradation pathways were not responsible for antigen loss, as determined by an inhibitor assay. Finally, HER2 mRNA was found to be not present in the monosomes and polysomes of CT26/HER2-A2 cells, as opposed to CT26/HER2 cells, suggesting that the translation activity of HER2 mRNAs may be suppressed in these immune-resistant cells. Taken together, our results report a new mechanism by which tumor cells respond to antitumor protective immunity for antitumor immune evasion.


Subject(s)
Antigens, Neoplasm/immunology , Neoplasms, Experimental/immunology , RNA, Messenger/immunology , RNA, Neoplasm/immunology , Receptor, ErbB-2/immunology , Tumor Escape , Animals , Antigens, Neoplasm/genetics , Cell Line, Tumor , Female , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Receptor, ErbB-2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL