Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
Add more filters

Publication year range
1.
Immunity ; 52(5): 872-884.e5, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32433950

ABSTRACT

Some endocrine organs are frequent targets of autoimmune attack. Here, we addressed the origin of autoimmune disease from the viewpoint of feedback control. Endocrine tissues maintain mass through feedback loops that balance cell proliferation and removal according to hormone-driven regulatory signals. We hypothesized the existence of a dedicated mechanism that detects and removes mutant cells that missense the signal and therefore hyperproliferate and hypersecrete with potential to disrupt organismal homeostasis. In this mechanism, hypersecreting cells are preferentially eliminated by autoreactive T cells at the cost of a fragility to autoimmune disease. The "autoimmune surveillance of hypersecreting mutants" (ASHM) hypothesis predicts the presence of autoreactive T cells in healthy individuals and the nature of self-antigens as peptides from hormone secretion pathway. It explains why some tissues get prevalent autoimmune disease, whereas others do not and instead show prevalent mutant-expansion disease (e.g., hyperparathyroidism). The ASHM hypothesis is testable, and we discuss experimental follow-up.


Subject(s)
Autoimmune Diseases/immunology , Diabetes Mellitus, Type 1/immunology , Endocrine Glands/immunology , Endocrine System/immunology , Immunologic Surveillance/immunology , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Cell Proliferation/genetics , Cell Proliferation/physiology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Endocrine Glands/cytology , Endocrine Glands/metabolism , Endocrine System/cytology , Endocrine System/metabolism , Female , Humans , Immunologic Surveillance/genetics , Male , Mutation , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
2.
EMBO J ; 42(10): e114050, 2023 05 15.
Article in English | MEDLINE | ID: mdl-37051718

ABSTRACT

Newly formed malignant cells must escape immunosurveillance to generate progressing neoplastic lesions of clinical relevance. Recent data indicate that the immunogenicity of nascent cancer cells, at least in some settings, is dictated by inherent epigenetic mechanisms rather than by immunoediting and the consequent Darwinian selection of poorly immunogenic phenotypes.


Subject(s)
Neoplasms , Humans , Neoplasms/genetics , Monitoring, Immunologic , Immunologic Surveillance/genetics , Epigenomics , Tumor Escape/genetics , Immunotherapy
3.
Immunity ; 49(6): 1132-1147.e7, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30552022

ABSTRACT

Serrated adenocarcinoma, an alternative pathway for colorectal cancer (CRC) development, accounts for 15%-30% of all CRCs and is aggressive and treatment resistant. We show that the expression of atypical protein kinase C ζ (PKCζ) and PKCλ/ι was reduced in human serrated tumors. Simultaneous inactivation of the encoding genes in the mouse intestinal epithelium resulted in spontaneous serrated tumorigenesis that progressed to advanced cancer with a strongly reactive and immunosuppressive stroma. Whereas epithelial PKCλ/ι deficiency led to immunogenic cell death and the infiltration of CD8+ T cells, which repressed tumor initiation, PKCζ loss impaired interferon and CD8+ T cell responses, which resulted in tumorigenesis. Combined treatment with a TGF-ß receptor inhibitor plus anti-PD-L1 checkpoint blockade showed synergistic curative activity. Analysis of human samples supported the relevance of these kinases in the immunosurveillance defects of human serrated CRC. These findings provide insight into avenues for the detection and treatment of this poor-prognosis subtype of CRC.


Subject(s)
Intestinal Mucosa/immunology , Intestinal Neoplasms/immunology , Isoenzymes/immunology , Protein Kinase C/immunology , Adult , Aged , Aged, 80 and over , Animals , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Female , Humans , Immunologic Surveillance/genetics , Immunologic Surveillance/immunology , Intestinal Mucosa/enzymology , Intestinal Mucosa/pathology , Intestinal Neoplasms/enzymology , Intestinal Neoplasms/genetics , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Mice, Knockout , Mice, Transgenic , Middle Aged , Protein Kinase C/genetics , Protein Kinase C/metabolism , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism
4.
Genome Res ; 29(2): 270-280, 2019 02.
Article in English | MEDLINE | ID: mdl-30670627

ABSTRACT

Aberrant DNA methylation is a distinguishing feature of cancer. Yet, how methylation affects immune surveillance and tumor metastasis remains ambiguous. We introduce a novel method, Guide Positioning Sequencing (GPS), for precisely detecting whole-genome DNA methylation with cytosine coverage as high as 96% and unbiased coverage of GC-rich and repetitive regions. Systematic comparisons of GPS with whole-genome bisulfite sequencing (WGBS) found that methylation difference between gene body and promoter is an effective predictor of gene expression with a correlation coefficient of 0.67 (GPS) versus 0.33 (WGBS). Moreover, Methylation Boundary Shift (MBS) in promoters or enhancers is capable of modulating expression of genes associated with immunity and tumor metabolism. Furthermore, aberrant DNA methylation results in tissue-specific enhancer switching, which is responsible for altering cell identity during liver cancer development. Altogether, we demonstrate that GPS is a powerful tool with improved accuracy and efficiency over WGBS in simultaneously detecting genome-wide DNA methylation and genomic variation. Using GPS, we show that aberrant DNA methylation is associated with altering cell identity and immune surveillance networks, which may contribute to tumorigenesis and metastasis.


Subject(s)
DNA Methylation , Gene Expression Regulation, Neoplastic , Sequence Analysis, DNA/methods , Carcinogenesis/genetics , Cell Line, Tumor , Enhancer Elements, Genetic , Genome, Human , Humans , Immunologic Surveillance/genetics , Liver/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Neoplasm Metastasis , Promoter Regions, Genetic , Ribosomal Proteins/genetics
5.
Immunol Rev ; 283(1): 54-76, 2018 05.
Article in English | MEDLINE | ID: mdl-29664571

ABSTRACT

A large proportion of memory T cells disseminated throughout the body are non-recirculating cells whose maintenance and function is regulated by tissue-specific environmental cues. These sessile cells are referred to as tissue-resident memory T (TRM ) cells and similar populations of non-recirculating cells also exist among unconventional T cells and innate lymphocyte cells. The pool of TRM cells is highly diverse with respect to anatomical positioning, phenotype, molecular regulation and effector function. Nevertheless, certain transcriptional programs are shared and appear as important unifying features for the overall population of TRM cells and tissue-resident lymphocytes. It is now widely appreciated that TRM cells are a critical component of our immune defense by acting as peripheral sentinels capable of rapidly mobilizing protective tissue immunity upon pathogen recognition. This function is of particular importance in anatomical sites that are not effectively surveilled by blood-borne memory T cells in absence of inflammation, such as neuronal tissues or epithelial compartments in skin and mucosae. Focusing on the well-characterized subtype of CD8+  CD69+  CD103+ TRM cells, we will review current concepts on the generation, persistence and function of TRM cells and will summarize commonly used tools to study these cells. Furthermore, we will discuss accumulating data that emphasize localized TRM responses as an important determinant of tissue homeostasis and immune defense in the context of microbiota-immune interactions, persistent infections and cancer surveillance.


Subject(s)
Homeostasis , Immunologic Memory , Immunologic Surveillance , Infections/immunology , Neoplasms/immunology , T-Lymphocyte Subsets/immunology , Animals , Biomarkers , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immunologic Surveillance/genetics , Infections/genetics , Lymphocyte Count , Mice , Microbiota/immunology , Models, Biological , Neoplasms/genetics , Organ Specificity/genetics , Organ Specificity/immunology , Phenotype , T-Lymphocyte Subsets/metabolism , Transcriptome
6.
J Cell Mol Med ; 25(5): 2333-2341, 2021 03.
Article in English | MEDLINE | ID: mdl-33605033

ABSTRACT

Autophagy is frequently induced in the hypoxic tumour microenvironment. Accumulating evidence reveals important functions of autophagy at the tumour-immune interface. Herein, we propose an update on the roles of autophagy in modulating tumour immunity. Autophagy promotes adaptive resistance of established tumours to the cytotoxic effects of natural killer cells (NKs), macrophages and effector T cells. Increased autophagic flux in tumours dampen their immunogenicity and inhibits the expansion of cytotoxic T lymphocytes (CTLs) by suppressing the activation of STING type I interferon signalling (IFN-I) innate immune sensing pathway. Autophagy in suppressive tumour-infiltrating immune subsets maintains their survival through metabolic remodelling. On the other hand, autophagy is involved in the antigen processing and presentation process, which is essential for anti-tumour immune responses. Genetic deletion of autophagy induces spontaneous tumours in some models. Thus, the role of autophagy is context-dependent. In summary, our review has revealed the dichotomous roles of autophagy in modulating tumour immunity. Broad targeting of autophagy may not yield maximal benefits. The characterization of specific genes regulating tumour immunogenicity and innovation in targeted delivery of autophagy inhibitors into certain tumours are among the most urgent tasks to sensitize cold cancers to immunotherapy.


Subject(s)
Autophagy , Immunity , Neoplasms/etiology , Neoplasms/metabolism , Tumor Microenvironment/immunology , Adaptive Immunity , Animals , Antigen Presentation , Antigens, Neoplasm , Autophagy/genetics , Autophagy/immunology , Biomarkers , Disease Susceptibility , Gene Expression Regulation, Neoplastic , Humans , Immunity, Innate , Immunologic Surveillance/genetics , Immunologic Surveillance/immunology , Neoplasms/pathology , Signal Transduction , Tumor Microenvironment/genetics
7.
Eur J Immunol ; 50(3): 439-444, 2020 03.
Article in English | MEDLINE | ID: mdl-31729760

ABSTRACT

It is well established that therapeutic impairment of Foxp3+ Treg in mice and humans favors immune rejection of solid tumors. Less explored is the impact Foxp3 allelic variants may have on tumor incidence, progression and therapy. In this work, we tested and demonstrate that the Foxp3fgfp reporter allele, found previously to either enhance or reduce Treg function in specific autoimmunity settings, confers increased anti-tumor immunity. Our conclusions stem out of the analysis of three tumor models of different tissue origin, in two murine genetic backgrounds. When compared to wild type animals, mice carrying the Foxp3fgfp allele spontaneously delay, reduce or prevent primary tumor growth, decrease metastasis growth, and potentiate the response to anti-CTLA4 monotherapy. These findings suggest allelic variances at the Foxp3 locus may serve as predictive indicators for personalized therapy and prognostics, and point at possible new therapeutic targets.


Subject(s)
Forkhead Transcription Factors/immunology , Immunologic Surveillance/genetics , Neoplasms, Experimental/immunology , Alleles , Animals , Forkhead Transcription Factors/genetics , Immunologic Surveillance/immunology , Mice , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/immunology
8.
Proc Natl Acad Sci U S A ; 115(50): 12565-12572, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30455304

ABSTRACT

Endogenous retroviruses (ERVs) are integrated retroviral elements that make up 8% of the human genome. However, the impact of ERVs on human health and disease is not well understood. While select ERVs have been implicated in diseases, including autoimmune disease and cancer, the lack of tools to analyze genome-wide, locus-specific expression of proviral autonomous ERVs has hampered the progress in the field. Here we describe a method called ERVmap, consisting of an annotated database of 3,220 human proviral ERVs and a pipeline that allows for locus-specific genome-wide identification of proviral ERVs that are transcribed based on RNA-sequencing data, and provide examples of the utility of this tool. Using ERVmap, we revealed cell-type-specific ERV expression patterns in commonly used cell lines as well as in primary cells. We identified 124 unique ERV loci that are significantly elevated in the peripheral blood mononuclear cells of patients with systemic lupus erythematosus that represent an IFN-independent signature. Finally, we identified additional tumor-associated ERVs that correlate with cytolytic activity represented by granzyme and perforin expression in breast cancer tissue samples. The open-source code of ERVmap and the accompanied web tool are made publicly available to quantify proviral ERVs in RNA-sequencing data with ease. Use of ERVmap across a range of diseases and experimental conditions has the potential to uncover novel disease-associated antigens and effectors involved in human health that is currently missed by focusing on protein-coding sequences.


Subject(s)
Endogenous Retroviruses/genetics , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/virology , Cell Line , Chromosome Mapping , Computational Biology , Databases, Nucleic Acid , Endogenous Retroviruses/immunology , Endogenous Retroviruses/physiology , Female , Genome, Human , Genome, Viral , Humans , Immunologic Surveillance/genetics , Leukocytes, Mononuclear/virology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/virology , Proviruses/genetics , Sequence Analysis, RNA , Transcription, Genetic
9.
J Cell Mol Med ; 24(24): 14583-14595, 2020 12.
Article in English | MEDLINE | ID: mdl-33185980

ABSTRACT

Long-term subjection to shift work increases the risk of cancer. The purpose of the present study was to explore the mechanism by which chronic circadian disruption impairs natural killer (NK) cell immunosurveillance. Mice were subjected to light-dark reverse every 4 days for 12 weeks to disrupt normal circadian rhythm. NK cell development and function were evaluated by flow cytometry. The mRNA and protein levels of period 1 (per1) and per2 were suppressed, while circadian locomotor output cycle kaput (CLOCK) was increased in the shifted mice, indicating successful generation of the circadian rhythm disruption mouse model. Chronic shift-lag promoted NK cell ageing, which is likely due to the reduction in Ly49 family receptor expression in shifted NK. We further studied the effects of circadian rhythm disruption on NK cell function. Chronic shift-lag inhibited NK cell secretion of granular CD107a and interferon gamma. Moreover, chronic shift-lag attenuated the clearance of MHC-I-deficient tumour cells by NK cells in vivo and promoted lung metastasis of B16F10 melanomas. Furthermore, chronic shift-lag reduced NK cell killing function, which may be due to the suppression of Eomes transcription factor expression, which inhibiting the transcription of CD122. In conclusion, our findings suggest that chronic circadian disruption attenuates NK cell cytolytic activity by decreasing the expression of CD122.


Subject(s)
Cellular Senescence , Circadian Rhythm/genetics , Immunologic Surveillance , Interleukin-2 Receptor beta Subunit/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Shift Work Schedule/adverse effects , Animals , Apoptosis/genetics , Biomarkers , Cell Proliferation , Cellular Senescence/genetics , Gene Expression Regulation , Immunologic Surveillance/genetics , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-2 Receptor beta Subunit/metabolism , Lysosomal Membrane Proteins/genetics , Lysosomal Membrane Proteins/metabolism , Male , Mice , Spleen/immunology , Spleen/metabolism
10.
Cancer Sci ; 111(8): 3032-3044, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32449240

ABSTRACT

Tumor location and immunity play important roles in the progression of colorectal cancer (CRC). This study aimed to investigate the differences in the immunosurveillance pattern between right- and left-sided CRC and analyze their association with clinicopathologic features, including mismatch repair (MMR) status. We included surgically resected stage II/III CRC cases and evaluated the immunohistochemical findings of HLA class I, HLA class II, programmed cell death-ligand 1 (PD-L1), PD-1, CTLA-4, CD3, CD4, CD8, TIA-1, T-bet, GATA3, RORγT, Foxp3, and CD163. A total of 117 patients were included in the analyses; of these, 30 and 87 had right- and left-sided cancer, respectively. Tumor immunity varied according to the tumor location in the overall cohort. Analysis of the tumors excluding those with DNA mismatch repair (MMR) deficiency also revealed that tumor immunity differed according to the tumor location. In right-sided colon cancer (CC), high expression of Foxp3 (P = .0055) and TIA-1 (P = .0396) were associated with significantly better disease-free survival (DFS). High CD8 (P = .0808) and CD3 (P = .0863) expression tended to have better DFS. Furthermore, in left-sided CRC, only high PD-L1 expression in the stroma (P = .0426) was associated with better DFS. In multivariate analysis, high Foxp3 expression in right-sided CC was an independent prognostic factor for DFS (hazard ratio, 7.6445; 95% confidence interval, 1.2091-150.35; P = .0284). In conclusion, the immunosurveillance pattern differs between right- and left-sided CRC, even after adjusting for MMR deficiency.


Subject(s)
Biomarkers, Tumor/metabolism , Colorectal Neoplasms/immunology , DNA Mismatch Repair/immunology , Immunologic Surveillance/genetics , Adult , Aged , Aged, 80 and over , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , Biomarkers, Tumor/immunology , CD3 Complex/immunology , CD3 Complex/metabolism , CD8 Antigens/immunology , CD8 Antigens/metabolism , Colon/immunology , Colon/pathology , Colon/surgery , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Colorectal Neoplasms/surgery , Disease-Free Survival , Female , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Humans , Male , Middle Aged , Rectum/immunology , Rectum/pathology , Rectum/surgery , T-Cell Intracellular Antigen-1/immunology , T-Cell Intracellular Antigen-1/metabolism
11.
Gastroenterology ; 156(6): 1877-1889.e4, 2019 05.
Article in English | MEDLINE | ID: mdl-30710528

ABSTRACT

BACKGROUND & AIMS: Inflammation in the liver provokes fibrosis, but inflammation is also important for tumor surveillance. Inhibitors of chemokine pathways, such as CXCL16 and CXCR6 regulation of lymphocyte trafficking, are being tested as antifibrotic agents, but their effects on the development of hepatocellular carcinoma (HCC) are unclear. We assessed the roles of CXCR6-dependent immune mechanisms in hepatocarcinogenesis. METHODS: C57BL/6J wild-type (WT) mice and CXCR6-deficient mice (Cxcr6eGfp/eGfp) were given injections of diethylnitrosamine (DEN) to induce liver cancer and α-galactosylceramide to activate natural killer T (NKT) cells. We also performed studies in mice with conditional, hepatocyte-specific deletion of NEMO, which develop inflammation-associated liver tumors (NemoLPC-KO and NemoLPC-KOCxcr6eGfp/eGfp mice). We collected liver tissues from patients with cirrhosis (n = 43), HCC (n = 35), and neither of these diseases (control individuals, n = 25). Human and mouse liver tissues were analyzed by histology, immunohistochemistry, flow cytometry, RNA expression arrays (from sorted hepatic lymphocytes), and matrix-assisted laser desorption/ionization imaging. Bone marrow was transferred from Cxcr6eGfp/eGfp or WT mice to irradiated C57BL/6J mice, and spleen and liver cells were analyzed by flow cytometry. CD4+ T cells or NKT cells were isolated from the spleen and liver of CD45.1+ WT mice and transferred into CXCR6-deficient mice after DEN injection. RESULTS: After DEN injection, CXCR6-deficient mice had a significantly higher tumor burden than WT mice and increased tumor progression, characterized by reduced intrahepatic numbers of invariant NKT and CD4+ T cells that express tumor necrosis factor and interferon gamma. Livers of NemoLPC-KOCxcr6eGfp/eGfp mice had significantly more senescent hepatocytes than livers of NemoLPC-KO mice. In studies of bone-marrow chimeras, adoptive cell transfer experiments, and analyses of NemoLPC-KO mice, we found that NKT and CD4 T cells promote the removal of senescent hepatocytes to prevent hepatocarcinogenesis, and that this process required CXCR6. Injection of WT with α-galactosylceramide increased removal of senescent hepatocytes by NKT cells. We observed peritumoral accumulation of CXCR6-associated lymphocytes in human HCC, which appeared reduced compared with cirrhosis tissues. CONCLUSIONS: In studies of mice with liver tumors, we found that CXCR6 mediated NKT-cell and CD4+ T-cell removal of senescent hepatocytes. Antifibrotic strategies to reduce CXCR6 activity in liver, or to reduce inflammation or modulate the immune response, should be tested for their effects on hepatocarcinogenesis.


Subject(s)
Carcinogenesis/immunology , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Natural Killer T-Cells/immunology , Receptors, CXCR6/genetics , Receptors, CXCR6/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , Carcinogenesis/genetics , Carcinoma, Hepatocellular/metabolism , Cellular Senescence , Diethylnitrosamine , Disease Progression , Galactosylceramides/pharmacology , Hepatocytes/physiology , Humans , Immunologic Surveillance/genetics , Interferon-gamma/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Liver Cirrhosis/pathology , Liver Neoplasms/chemically induced , Liver Neoplasms/metabolism , Lymphocyte Activation/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/metabolism , Receptors, CXCR6/metabolism , Tumor Burden/genetics , Tumor Necrosis Factor-alpha/metabolism
12.
BMC Cancer ; 20(1): 588, 2020 Jun 23.
Article in English | MEDLINE | ID: mdl-32576165

ABSTRACT

BACKGROUND: Prostate Cancer (PCa) is the second most common cancer in men where advancements have been made for early detection using imaging techniques, however these are limited by lesion size. Immune surveillance has emerged as an effective approach for early detection and to monitor disease progression. In recent studies, we have shown that host peripheral blood immune cells undergo changes in DNA methylation in liver and breast cancer. METHODS: In the current study, we examined the DNA methylation status of peripheral blood T cells of men with positive biopsy for PCa versus men with negative biopsy having benign prostate tissue, defined as controls. T cells DNA was isolated and subjected to Illumina Infinium methylation EPIC array and validated using Illumina amplicon sequencing and pyrosequencing platforms. RESULTS: Differential methylation of 449 CG sites between control and PCa T cell DNA showed a correlation with Gleason score (p < 0.05). Two hundred twenty-three differentially methylated CGs between control and PCa (Ƨ +/- 10%, p < 0.05), were enriched in pathways involved in immune surveillance system. Three CGs which were found differentially methylated following DMP (Differentially methylated probes) analysis of ChAMP remained significant after BH (Benjamini-Hochberg) correction, of which, 2 CGs were validated. Predictive ability of combination of these 3 CGs (polygenic methylation score, PMS) to detect PCa had high sensitivity, specificity and overall accuracy. PMS also showed strong positive correlation with Gleason score and tumor volume of PCa patients. CONCLUSIONS: Results from the current study provide for the first-time a potential role of DNA methylation changes in peripheral T cells in PCa. This non-invasive methodology may allow for early intervention and stratification of patients into different prognostic groups to reduce PCa associated morbidity from repeat invasive prostate biopsies and design therapeutic strategy to reduce PCa associated mortality.


Subject(s)
DNA Methylation/immunology , Epigenomics/methods , Immunologic Surveillance/genetics , Prostatic Neoplasms/diagnosis , T-Lymphocytes/immunology , Biopsy , Case-Control Studies , Epigenome/immunology , Healthy Volunteers , Humans , Male , Middle Aged , Neoplasm Grading , Prognosis , Prostate/pathology , Prostatic Neoplasms/blood , Prostatic Neoplasms/genetics , Prostatic Neoplasms/immunology , Sensitivity and Specificity , Sequence Analysis, DNA , Tumor Burden
13.
Mol Biol Rep ; 47(3): 2265-2277, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31925644

ABSTRACT

Recent decades, there is significant progress in understanding the mechanisms of tumor progression and immune evasion. The newly discovered protein NLRC5 is demonstrated to participate in regulating cancer immune escape through enhancing MHC class I genes expression in certain tumors. Nevertheless, increasing evidence has revealed that NLRC5 is up-regulated in some other tumors and promote tumor development and progression. The purpose of this review is to describe the role of NLRC5 in tumors and discuss whether NLRC5 can be a potential target in cancer treatment.


Subject(s)
Disease Susceptibility , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasms/etiology , Neoplasms/metabolism , Animals , Biomarkers, Tumor , Disease Progression , Gene Expression Regulation, Neoplastic , Genes, MHC Class I , Humans , Immunologic Surveillance/genetics , Intracellular Signaling Peptides and Proteins/chemistry , Neoplasms/drug therapy , Neoplasms/pathology , Organ Specificity , Signal Transduction , Structure-Activity Relationship
14.
Proc Natl Acad Sci U S A ; 114(7): E1178-E1187, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28137869

ABSTRACT

Memory CD8+ T cells confer long-term immunity against tumors, and anticancer vaccines therefore should maximize their generation. Multiple memory CD8+ T-cell subsets with distinct functional and homing characteristics exist, but the signaling pathways that regulate their development are ill defined. Here we examined the role of the serine/threonine kinase Akt in the generation of protective immunity by CD8+ T cells. Akt is known to be activated by the T-cell antigen receptor and the cytokine IL-2, but its role in T-cell immunity in vivo has not been explored. Using CD8+ T cells from pdk1K465E/K465E knockin mice, we found that decreased Akt activity inhibited the survival of T cells during the effector-to-memory cell transition and abolished their differentiation into C-X-C chemokine receptor 3 (CXCR3)loCD43lo effector-like memory cells. Consequently, antitumor immunity by CD8+ T cells that display defective Akt signaling was substantially diminished during the memory phase. Reduced memory T-cell survival and altered memory cell differentiation were associated with up-regulation of the proapoptotic protein Bim and the T-box transcription factor eomesodermin, respectively. These findings suggest an important role for effector-like memory CD8+ T cells in tumor immune surveillance and identify Akt as a key signaling node in the development of protective memory CD8+ T-cell responses.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Immunologic Surveillance/immunology , Neoplasms, Experimental/immunology , Proto-Oncogene Proteins c-akt/immunology , Animals , Binding Sites/genetics , CD8-Positive T-Lymphocytes/metabolism , CX3C Chemokine Receptor 1/immunology , CX3C Chemokine Receptor 1/metabolism , Cell Line, Tumor , Immunologic Surveillance/genetics , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Neoplasms, Experimental/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, CXCR3/immunology , Receptors, CXCR3/metabolism , Signal Transduction/immunology
15.
J Allergy Clin Immunol ; 143(1): 276-291.e6, 2019 01.
Article in English | MEDLINE | ID: mdl-29800648

ABSTRACT

BACKGROUND: Germline gain-of function (GOF) mutations in PIK3CD, encoding the catalytic p110δ subunit of phosphoinositide 3-kinase (PI3K), result in hyperactivation of the PI3K-AKT-mechanistic target of rapamycin pathway and underlie a novel inborn error of immunity. Affected subjects exhibit perturbed humoral and cellular immunity, manifesting as recurrent infections, autoimmunity, hepatosplenomegaly, uncontrolled EBV and/or cytomegalovirus infection, and increased incidence of B-cell lymphoproliferation, lymphoma, or both. Mechanisms underlying disease pathogenesis remain unknown. OBJECTIVE: Understanding the cellular and molecular mechanisms underpinning inefficient surveillance of EBV-infected B cells is required to understand disease in patients with PIK3CD GOF mutations, identify key molecules required for cell-mediated immunity against EBV, and develop immunotherapeutic interventions for the treatment of this and other EBV-opathies. METHODS: We studied the consequences of PIK3CD GOF mutations on the generation, differentiation, and function of CD8+ T cells and natural killer (NK) cells, which are implicated in host defense against infection with herpesviruses, including EBV. RESULTS: PIK3CD GOF total and EBV-specific CD8+ T cells were skewed toward an effector phenotype, with exaggerated expression of markers associated with premature immunosenescence/exhaustion and increased susceptibility to reactivation-induced cell death. These findings were recapitulated in a novel mouse model of PI3K GOF mutations. NK cells in patients with PIK3CD GOF mutations also exhibited perturbed expression of differentiation-associated molecules. Both CD8+ T and NK cells had reduced capacity to kill EBV-infected B cells. PIK3CD GOF B cells had increased expression of CD48, programmed death ligand 1/2, and CD70. CONCLUSIONS: PIK3CD GOF mutations aberrantly induce exhaustion, senescence, or both and impair cytotoxicity of CD8+ T and NK cells. These defects might contribute to clinical features of affected subjects, such as impaired immunity to herpesviruses and tumor surveillance.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Class I Phosphatidylinositol 3-Kinases , Epstein-Barr Virus Infections , Gain of Function Mutation , Genetic Diseases, Inborn/immunology , Herpesvirus 4, Human/immunology , Killer Cells, Natural/immunology , Adolescent , Adult , Aged , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Differentiation/genetics , Cellular Senescence/genetics , Cellular Senescence/immunology , Child , Child, Preschool , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/immunology , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/pathology , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/pathology , Humans , Immunologic Surveillance/genetics , Killer Cells, Natural/pathology , Male , Middle Aged
16.
Int J Mol Sci ; 21(2)2020 Jan 17.
Article in English | MEDLINE | ID: mdl-31963413

ABSTRACT

The immune system plays a dual role in tumor evolution-it can identify and control nascent tumor cells in a process called immunosurveillance and can promote tumor progression through immunosuppression via various mechanisms. Thus, bilateral host-protective and tumor-promoting actions of immunity are integrated as cancer immunoediting. In this decade, immune checkpoint inhibitors, specifically programmed cell death 1 (PD-1) pathway inhibitors, have changed the treatment paradigm of advanced non-small cell lung cancer (NSCLC). These agents are approved for the treatment of patients with NSCLC and demonstrate impressive clinical activity and durable responses in some patients. However, for many NSCLC patients, the efficacy of immune checkpoint inhibitors is limited. To optimize the full utility of the immune system for eradicating cancer, a broader understanding of cancer immunosurveillance and immunoediting is essential. In this review, we discuss the fundamental knowledge of the phenomena and provide an overview of the next-generation immunotherapies in the pipeline.


Subject(s)
Genetic Variation , Immunologic Surveillance/immunology , Immunotherapy/methods , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Monitoring, Immunologic/methods , Animals , Humans , Immunologic Surveillance/genetics , Lung Neoplasms/genetics
17.
Semin Cancer Biol ; 53: 189-200, 2018 12.
Article in English | MEDLINE | ID: mdl-30261276

ABSTRACT

Tumor lesions comprise multiple subpopulations of cells including those endowed with "stemness" properties. The latter cells are responsible of tumor initiation, metastasis formation, resistance to conventional therapies and disease recurrence. These relatively rare cells denominated cancer stem cells (CSCs) or cancer initiating cells (CICs) are defined based on self-renewing, multipotency and tumorigenicity. These cells through their immunomodulating features can evade from immunesurveillance, persisting in the form of quiescence and dormancy. They can drive the neoplastic growth and recurrence, even after long latency. Moreover, CSCs/CICs due to their ability to modulate and shape immune responses can represent the component of a tumor causing immunotherapy resistance in cancer patients. In this review a general overview of immunological properties of CSCs/CICs is provided, with a special focus on the mechanisms of modulation of T cell mediated responses. The need to further dissect the mechanisms regulating the immunological profile of CSCs/CICs and their interactions with immune cells and tumor microenvironment is discussed. An improved characterization of the immunological properties of CSCs/CICs will contribute to the rationale design of immunotherapeutic interventions which target these cells and may lead to the eradication of malignant diseases.


Subject(s)
Immunologic Surveillance/immunology , Neoplasms/immunology , Neoplastic Stem Cells/immunology , Tumor Microenvironment/immunology , Animals , Gene Expression Regulation, Neoplastic/immunology , Humans , Immunologic Surveillance/genetics , Immunotherapy , MicroRNAs/genetics , MicroRNAs/immunology , Neoplasms/genetics , Neoplasms/therapy , Neoplastic Stem Cells/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Microenvironment/genetics
18.
Proc Natl Acad Sci U S A ; 113(42): E6467-E6475, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27698133

ABSTRACT

Infection with Epstein-Barr virus (EBV) affects most humans worldwide and persists life-long in the presence of robust virus-specific T-cell responses. In both immunocompromised and some immunocompetent people, EBV causes several cancers and lymphoproliferative diseases. EBV transforms B cells in vitro and encodes at least 44 microRNAs (miRNAs), most of which are expressed in EBV-transformed B cells, but their functions are largely unknown. Recently, we showed that EBV miRNAs inhibit CD4+ T-cell responses to infected B cells by targeting IL-12, MHC class II, and lysosomal proteases. Here we investigated whether EBV miRNAs also counteract surveillance by CD8+ T cells. We have found that EBV miRNAs strongly inhibit recognition and killing of infected B cells by EBV-specific CD8+ T cells through multiple mechanisms. EBV miRNAs directly target the peptide transporter subunit TAP2 and reduce levels of the TAP1 subunit, MHC class I molecules, and EBNA1, a protein expressed in most forms of EBV latency and a target of EBV-specific CD8+ T cells. Moreover, miRNA-mediated down-regulation of the cytokine IL-12 decreases the recognition of infected cells by EBV-specific CD8+ T cells. Thus, EBV miRNAs use multiple, distinct pathways, allowing the virus to evade surveillance not only by CD4+ but also by antiviral CD8+ T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/immunology , Immunologic Surveillance/genetics , MicroRNAs/genetics , RNA, Viral/genetics , Antigen Presentation , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/virology , CD8-Positive T-Lymphocytes/metabolism , Cell Line , Cell Survival/immunology , Cytokines/metabolism , Cytotoxicity, Immunologic , Epitopes, T-Lymphocyte/metabolism , Epstein-Barr Virus Infections/metabolism , Gene Expression Regulation, Viral , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Immune Evasion , Receptors, Cytokine/metabolism
19.
Int J Mol Sci ; 20(18)2019 Sep 17.
Article in English | MEDLINE | ID: mdl-31533245

ABSTRACT

Extracellular heat shock proteins (ex-HSPs) have been found in exosomes, oncosomes, membrane surfaces, as well as free HSP in cancer and various pathological conditions, also known as alarmins. Such ex-HSPs include HSP90 (α, ß, Gp96, Trap1), HSP70, and large and small HSPs. Production of HSPs is coordinately induced by heat shock factor 1 (HSF1) and hypoxia-inducible factor 1 (HIF-1), while matrix metalloproteinase 3 (MMP-3) and heterochromatin protein 1 are novel inducers of HSPs. Oncosomes released by tumor cells are a major aspect of the resistance-associated secretory phenotype (RASP) by which immune evasion can be established. The concepts of RASP are: (i) releases of ex-HSP and HSP-rich oncosomes are essential in RASP, by which molecular co-transfer of HSPs with oncogenic factors to recipient cells can promote cancer progression and resistance against stresses such as hypoxia, radiation, drugs, and immune systems; (ii) RASP of tumor cells can eject anticancer drugs, targeted therapeutics, and immune checkpoint inhibitors with oncosomes; (iii) cytotoxic lipids can be also released from tumor cells as RASP. ex-HSP and membrane-surface HSP (mHSP) play immunostimulatory roles recognized by CD91+ scavenger receptor expressed by endothelial cells-1 (SREC-1)+ Toll-like receptors (TLRs)+ antigen-presenting cells, leading to antigen cross-presentation and T cell cross-priming, as well as by CD94+ natural killer cells, leading to tumor cytolysis. On the other hand, ex-HSP/CD91 signaling in cancer cells promotes cancer progression. HSPs in body fluids are potential biomarkers detectable by liquid biopsies in cancers and tissue-damaged diseases. HSP-based vaccines, inhibitors, and RNAi therapeutics are also reviewed.


Subject(s)
Extracellular Space/metabolism , Heat-Shock Proteins/metabolism , Immune Evasion , Immunologic Surveillance , Animals , Biomarkers , Extracellular Vesicles/metabolism , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/genetics , Humans , Immune Evasion/genetics , Immune System/immunology , Immune System/metabolism , Immunologic Surveillance/genetics , Immunomodulation , Liquid Biopsy , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/etiology , Neoplasms/metabolism , Neoplasms/pathology , Receptors, Cell Surface/metabolism
20.
Ann Oncol ; 29(11): 2232-2239, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30203045

ABSTRACT

Background: Little is known about how the immune microenvironment of breast cancer evolves during disease progression. Patients and methods: We compared tumor infiltrating lymphocyte (TIL) count, programmed death-ligand 1 (PD-L1) protein expression by immunohistochemistry and mRNA levels of 730 immune-related genes using Nanostring technology in primary and metastatic cancer samples. Results: TIL counts and PD-L1 positivity were significantly lower in metastases. Immune cell metagenes corresponding to CD8, T-helper, T-reg, Cytotoxic T, Dendritic and Mastoid cells, and expression of 13 of 29 immuno-oncology therapeutic targets in clinical development including PD1, PD-L1, and CTLA4 were significantly lower in metastases. There was also coordinated down regulation of chemoattractant ligand/receptor pairs (CCL19/CCR7, CXCL9/CXCR3, IL15/IL15R), interferon regulated genes (STAT1, IRF-1,-4,-7, IFI-27,-35), granzyme/granulysin, MHC class I and immune proteasome (PSMB-8,-9,-10) expression in metastases. Immunotherapy response predictive signatures were also lower. The expression of macrophage markers (CD163, CCL2/CCR2, CSF1/CSFR1, CXCR4/CXCL12), protumorigenic toll-like receptor pathway genes (CD14/TLR-1,-2,-4,-5,-6/MyD88), HLA-E, ecto-nuclease CD73/NT5E and inhibitory complement receptors (CD-59,-55,-46) remained high in metastases and represent potential therapeutic targets. Conclusions: Metastatic breast cancers are immunologically more inert than the corresponding primary tumors but some immune-oncology targets and macrophage and angiogenesis signatures show preserved expression and suggest therapeutic combinations for clinical testing.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Biomarkers, Tumor/immunology , Breast Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating , Tumor Microenvironment/immunology , Adolescent , Adult , Aged , Antineoplastic Agents, Immunological/pharmacology , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Biopsy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/secondary , Disease Progression , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/immunology , Female , Gene Expression Regulation/immunology , Humans , Immunologic Surveillance/genetics , Immunologic Surveillance/immunology , Lymphocyte Count , Middle Aged , Mutation Rate , Tumor Escape/genetics , Tumor Escape/immunology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL