Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 667
1.
In Vitro Cell Dev Biol Anim ; 60(4): 382-396, 2024 Apr.
Article En | MEDLINE | ID: mdl-38625487

It is necessary to explore new targets for the treatment of colon adenocarcinoma (COAD) according to the tumor microenvironment. The expression levels of JAML and CXADR were analyzed by bioinformatics analysis and validation of clinical samples. JAML over-expression CD8+ T cell line was constructed, and the proliferation activity was detected by MTT. The production of inflammatory factors was detected by ELISA. The expression of immune checkpoint PD-1 and TIM-3 was detected by Western blot. The apoptosis level was detected by flow cytometry and apoptosis markers. The AOM/DSS mouse model of colorectal cancer was constructed. The expression levels of JAML, CXADR and PD-1 were detected by PCR and Western blot, and the proportion of CD8+ T cells and exhausted T cells were detected by flow cytometry. The expression levels of JAML and CXADR were significantly decreased in colon cancer tissues. Overexpression of JAML can promote the proliferation of T cells, secrete a variety of inflammatory factors. Overexpression of CXADR can reduce the proliferation of colorectal cancer cells, promote apoptosis, and down-regulate the migration and invasion ability of tumor cells. Both JAML agonists and PD-L1 inhibitors can effectively treat colorectal cancer, and the combined use of JAML agonists and PD-L1 inhibitors can enhance the effect. JAML can promote the proliferation and toxicity of CD8+ T cells and down-regulate the expression of immune checkpoints in colon cancer. CXADR can inhibit the proliferation of cancer cells and promote the apoptosis. JAML agonist can effectively treat colorectal cancer by regulating CD8+ T cells.


Apoptosis , CD8-Positive T-Lymphocytes , Carcinogenesis , Cell Proliferation , Colorectal Neoplasms , Tumor Microenvironment , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Animals , Colorectal Neoplasms/pathology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/genetics , Humans , Cell Proliferation/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Mice , Carcinogenesis/drug effects , Carcinogenesis/immunology , Carcinogenesis/genetics , Apoptosis/drug effects , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Cell Movement/drug effects , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/genetics , Male
2.
EBioMedicine ; 103: 105102, 2024 May.
Article En | MEDLINE | ID: mdl-38614865

BACKGROUND: Cell-cell interaction factors that facilitate the progression of adenoma to sporadic colorectal cancer (CRC) remain unclear, thereby hindering patient survival. METHODS: We performed spatial transcriptomics on five early CRC cases, which included adenoma and carcinoma, and one advanced CRC. To elucidate cell-cell interactions within the tumour microenvironment (TME), we investigated the colocalisation network at single-cell resolution using a deep generative model for colocalisation analysis, combined with a single-cell transcriptome, and assessed the clinical significance in CRC patients. FINDINGS: CRC cells colocalised with regulatory T cells (Tregs) at the adenoma-carcinoma interface. At early-stage carcinogenesis, cell-cell interaction inference between colocalised adenoma and cancer epithelial cells and Tregs based on the spatial distribution of single cells highlighted midkine (MDK) as a prominent signalling molecule sent from tumour epithelial cells to Tregs. Interaction between MDK-high CRC cells and SPP1+ macrophages and stromal cells proved to be the mechanism underlying immunosuppression in the TME. Additionally, we identified syndecan4 (SDC4) as a receptor for MDK associated with Treg colocalisation. Finally, clinical analysis using CRC datasets indicated that increased MDK/SDC4 levels correlated with poor overall survival in CRC patients. INTERPRETATION: MDK is involved in the immune tolerance shown by Tregs to tumour growth. MDK-mediated formation of the TME could be a potential target for early diagnosis and treatment of CRC. FUNDING: Japan Society for the Promotion of Science (JSPS) Grant-in-Aid for Science Research; OITA Cancer Research Foundation; AMED under Grant Number; Japan Science and Technology Agency (JST); Takeda Science Foundation; The Princess Takamatsu Cancer Research Fund.


Colorectal Neoplasms , Single-Cell Analysis , T-Lymphocytes, Regulatory , Tumor Microenvironment , Humans , Colorectal Neoplasms/immunology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Tumor Microenvironment/immunology , Carcinogenesis/genetics , Carcinogenesis/immunology , Gene Expression Profiling , Transcriptome , Cell Communication/immunology , Immune Tolerance , Gene Expression Regulation, Neoplastic , Male , Female
3.
Adv Drug Deliv Rev ; 209: 115319, 2024 Jun.
Article En | MEDLINE | ID: mdl-38643839

The microbiome has emerged as a significant biomarker and modulator in cancer development and treatment response. Recent research highlights the notable role of Fusobacterium nucleatum (F. nucleatum) in various tumor types, including breast, colorectal, esophageal, gastric, pancreatic, and lung cancers. Accumulating evidence suggests that the local microbial community forms an integral component of the tumor microenvironment, with bacterial communities within tumors displaying specificity to tumor types. Mechanistic investigations indicate that tumor-associated microbiota can directly influence tumor initiation, progression, and responses to chemotherapy or immunotherapy. This article presents a comprehensive review of microbial communities especially F. nucleatum in tumor tissue, exploring their roles and underlying mechanisms in tumor development, treatment, and prevention. When the tumor-associated F. nucleatum is killed, the host immune response is activated to recognize tumor cells. Bacteria epitopes restricted by the host antigens, can be identified for future anti-bacteria/tumor vaccine development.


Fusobacterium nucleatum , Neoplasms , Humans , Neoplasms/drug therapy , Neoplasms/immunology , Drug Delivery Systems , Carcinogenesis/immunology , Tumor Microenvironment/immunology , Animals
4.
Cell Rep ; 43(4): 114096, 2024 Apr 23.
Article En | MEDLINE | ID: mdl-38607919

Receptors controlling the cross-presentation of tumor antigens by macrophage subsets in cancer tissues are poorly explored. Here, we show that TIM4+ large peritoneal macrophages efficiently capture and cross-present tumor-associated antigens at early stages of peritoneal infiltration by ovarian cancer cells. The phosphatidylserine (PS) receptor TIM4 promotes maximal uptake of dead cells or PS-coated artificial targets and triggers inflammatory and metabolic gene programs in combination with cytoskeletal remodeling and upregulation of transcriptional signatures related to antigen processing. At the cellular level, TIM4-mediated engulfment induces nucleation of F-actin around nascent phagosomes, delaying the recruitment of vacuolar ATPase, acidification, and cargo degradation. In vivo, TIM4 deletion blunts induction of early anti-tumoral effector CD8 T cells and accelerates the progression of ovarian tumors. We conclude that TIM4-mediated uptake drives the formation of specialized phagosomes that prolong the integrity of ingested antigens and facilitate cross-presentation, contributing to immune surveillance of the peritoneum.


Antigens, Neoplasm , Carcinogenesis , Macrophages, Peritoneal , Animals , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/immunology , Female , Mice , Carcinogenesis/pathology , Carcinogenesis/immunology , Carcinogenesis/metabolism , Humans , Antigens, Neoplasm/metabolism , Antigens, Neoplasm/immunology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Cross-Priming/immunology , Cell Line, Tumor , Phagosomes/metabolism , Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Actins/metabolism
5.
Int Immunopharmacol ; 132: 111918, 2024 May 10.
Article En | MEDLINE | ID: mdl-38537539

BACKGROUND: Sideroflexin 1 (SFXN1), a mitochondrial serine transporter implicated in one-carbon metabolism, is a prognostic biomarker in lung adenocarcinoma (LUAD). However, its role in LUAD progression remains elusive. This study aimed to investigate the functional significance of SFXN1 in LUAD and evaluate its potential as a therapeutic target. METHODS: We analyzed SFXN1 expression and its diagnostic and prognostic value in LUAD using the Pan-cancer TCGA dataset. In vitro assays (CCK-8, cell cycle, EDU, wound-healing, and transwell) were employed to assess the role of SFXN1, complemented by in vivo experiments. RNA sequencing elucidated SFXN1-mediated cellular functions and potential mechanisms. Bulk RNA-seq and scRNA-seq data from TCGA and GEO were used to investigate the correlation between SFXN1 and the tumor immune microenvironment. RT-qPCR, Western blot, and IHC assays validated SFXN1 expression and its impact on the immune microenvironment in LUAD. RESULTS: SFXN1 was upregulated in LUAD tissues and associated with poor prognosis. RNA-seq and scRNA-seq analyses revealed increased SFXN1 expression in tumor cells, accompanied by decreased infiltration of NK and cytotoxic T cells. SFXN1 knockdown significantly reduced cell proliferation and migration, and the inhibition of ERK phosphorylation and CCL20 expression may be the molecular mechanism involved. In vivo, targeting SFXN1 decreased Tregs infiltration and inhibited tumor growth. CONCLUSIONS: Our findings suggest that SFXN1 may be a potential therapeutic target for LUAD treatment.


Adenocarcinoma of Lung , Amino Acid Transport Systems, Neutral , Lung Neoplasms , Lymphocytes, Tumor-Infiltrating , Tumor Microenvironment , Humans , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/immunology , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinogenesis/genetics , Carcinogenesis/immunology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Lung Neoplasms/immunology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Prognosis , Tumor Microenvironment/immunology , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism
6.
Science ; 381(6664): 1287-1288, 2023 09 22.
Article En | MEDLINE | ID: mdl-37733861

Mitochondrial metabolite reduces melanoma growth by boosting antigen presentation.


Antigen Presentation , Melanoma , Mitochondria , Humans , Melanoma/immunology , Melanoma/pathology , Mitochondria/metabolism , Carcinogenesis/immunology , Carcinogenesis/pathology
7.
Int J Cancer ; 152(12): 2615-2628, 2023 06 15.
Article En | MEDLINE | ID: mdl-36912275

Due to a combination of rapid disease progression and the lack of curative treatment options, hepatocellular carcinoma (HCC) is one of the deadliest cancers worldwide. Infiltrated, monocyte-derived, tumor-associated macrophages are known to play a role in HCC pathogenesis, but the involvement of Kupffer cells (KCs) remains elusive. Here, we used the Clec4F-diphteria toxin receptor transgenic mouse model to specifically investigate the effect of KC depletion on HCC initiation, progression and neoplastic growth following liver resection. For this purpose, several HCC mouse models with varying underlying etiologies were used and partial hepatectomy was performed. Our results show that in HCC, developed on a fibrotic or non-alcoholic steatohepatitis background, depletion of embryonic KCs at the onset of HCC induction and the subsequent replacement by monocyte-derived KCs does not affect the tumor burden, tumor microenvironment or the phenotype of isolated KCs at end-stage disease. In non-chronic liver disease-associated diethylnitrosamine-induced HCC, ablation of Clec4F+ KCs did not alter tumor progression or neoplastic growth following liver resection. Our results show that temporal ablation of resident KCs does not impact HCC pathogenesis, neither in the induction phase nor in advanced disease, and indicate that bone marrow-derived KCs are able to swiftly repopulate the available KC niche and adopt their phenotype.


Carcinogenesis , Carcinoma, Hepatocellular , Kupffer Cells , Liver Neoplasms, Experimental , Liver Neoplasms , Tumor-Associated Macrophages , Kupffer Cells/immunology , Disease Progression , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/pathology , Animals , Mice , Liver Neoplasms, Experimental/immunology , Liver Neoplasms, Experimental/pathology , Monocyte-Macrophage Precursor Cells/immunology , Carcinogenesis/immunology , Carcinogenesis/pathology , Mice, Inbred C57BL , Male
8.
Front Immunol ; 14: 942849, 2023.
Article En | MEDLINE | ID: mdl-36825005

Secretory pathway kinase or kinase-like proteins (SPKKPs) are effective in the lumen of the endoplasmic reticulum (ER), Golgi apparatus (GA), and extracellular space. These proteins are involved in secretory signaling pathways and are distinctive from typical protein kinases. Various reports have shown that SPKKPs regulate the tumorigenesis and progression of human cancer via the phosphorylation of various substrates, which is essential in physiological and pathological processes. Emerging evidence has revealed that the expression of SPKKPs in human cancers is regulated by multiple factors. This review summarizes the current understanding of the contribution of SPKKPs in tumorigenesis and the progression of immunity. With the epidemic trend of immunotherapy, targeting SPKKPs may be a novel approach to anticancer therapy. This study briefly discusses the recent advances regarding SPKKPs.


Neoplasms , Phosphotransferases , Secretory Pathway , Humans , Carcinogenesis/immunology , Neoplasms/immunology , Phosphotransferases/immunology , Proteins/immunology , Secretory Pathway/immunology , Signal Transduction/immunology , Disease Progression
9.
Nat Commun ; 13(1): 7243, 2022 11 25.
Article En | MEDLINE | ID: mdl-36433954

Exonic circular RNAs (circRNAs) produce predominantly non-coding RNA species that have been recently profiled in many tumors. However, their functional contribution to cancer progression is still poorly understood. Here, we identify the circRNAs expressed in soft tissue sarcoma cells and explore how the circRNAs regulate sarcoma growth in vivo. We show that circCsnk1g3 and circAnkib1 promote tumor growth by shaping a pro-tumorigenic microenvironment, possibly due to their capabilities to regulate tumor-promoting elements extrinsic to the tumor cells. Accordingly, circCsnk1g3 and circAnkib1 can control the expression of interferon-related genes and pro-inflammatory factors in the sarcoma cells, thus directing immune cell recruitment into the tumor mass, and hence their activation. Mechanistically, circRNAs may repress pro-inflammatory elements by buffering activation of the pathways mediated by RIG-I, the cytosolic viral RNA sensor. The current findings suggest that the targeting of specific circRNAs could augment the efficacy of tumor and immune response to mainstay therapies.


Carcinogenesis , Interferons , RNA, Circular , Sarcoma , Soft Tissue Neoplasms , Tumor Microenvironment , Humans , Carcinogenesis/genetics , Carcinogenesis/immunology , Interferons/genetics , Interferons/immunology , RNA, Circular/genetics , RNA, Circular/immunology , Sarcoma/genetics , Sarcoma/immunology , Soft Tissue Neoplasms/genetics , Soft Tissue Neoplasms/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Casein Kinase I/genetics , Casein Kinase I/immunology
10.
Proc Natl Acad Sci U S A ; 119(29): e2205574119, 2022 07 19.
Article En | MEDLINE | ID: mdl-35858331

Intestinal barrier immunity is essential for controlling gut microbiota without eliciting harmful immune responses, while its defect contributes to the breakdown of intestinal homeostasis and colitis development. Chemerin, which is abundantly expressed in barrier tissues, has been demonstrated to regulate tissue inflammation via CMKLR1, its functional receptor. Several studies have reported the association between increased expression of chemerin-CMKLR1 and disease severity and immunotherapy resistance in inflammatory bowel disease (IBD) patients. However, the pathophysiological role of endogenous chemerin-CMKLR1 signaling in intestinal homeostasis remains elusive. We herein demonstrated that deficiency of chemerin or intestinal epithelial cell (IEC)-specific CMKLR1 conferred high susceptibility to microbiota-driven neutrophilic colon inflammation and subsequent tumorigenesis in mice following epithelial injury. Unexpectedly, we found that lack of chemerin-CMKLR1 signaling specifically reduced expression of lactoperoxidase (LPO), a peroxidase that is predominantly expressed in colonic ECs and utilizes H2O2 to oxidize thiocyanates to the antibiotic compound, thereby leading to the outgrowth and mucosal invasion of gram-negative bacteria and dysregulated CXCL1/2-mediated neutrophilia. Importantly, decreased LPO expression was causally linked to aggravated microbiota-driven colitis and associated tumorigenesis, as LPO supplementation could completely rescue such phenotypes in mice deficient in epithelial chemerin-CMKLR1 signaling. Moreover, epithelial chemerin-CMKLR1 signaling is necessary for early host defense against bacterial infection in an LPO-dependent manner. Collectively, our study reveals that the chemerin-CMKLR1/LPO axis represents an unrecognized immune mechanism that potentiates epithelial antimicrobial defense and restricts harmful colonic neutrophilia and suggests that LPO supplementation may be beneficial for microbiota dysbiosis in IBD patients with a defective innate antimicrobial mechanism.


Carcinogenesis , Chemokines , Colitis , Colon , Gastrointestinal Microbiome , Intercellular Signaling Peptides and Proteins , Lactoperoxidase , Receptors, Chemokine , Animals , Carcinogenesis/immunology , Cell Transformation, Neoplastic , Chemokines/genetics , Chemokines/metabolism , Colitis/immunology , Colitis/microbiology , Colon/immunology , Colon/microbiology , Hydrogen Peroxide/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Lactoperoxidase/metabolism , Mice , Neutrophils/immunology , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism
11.
Front Immunol ; 13: 891268, 2022.
Article En | MEDLINE | ID: mdl-35720407

Since TGF-ß was recognized as an essential secreted cytokine in embryogenesis and adult tissue homeostasis a decade ago, our knowledge of the role of TGF-ß in mammalian development and disease, particularly cancer, has constantly been updated. Mounting evidence has confirmed that TGF-ß is the principal regulator of the immune system, as deprivation of TGF-ß signaling completely abrogates adaptive immunity. However, enhancing TGF-ß signaling constrains the immune response through multiple mechanisms, including boosting Treg cell differentiation and inducing CD8+ T-cell apoptosis in the disease context. The love-hate relationship between TGF-ß signaling and the immune system makes it challenging to develop effective monotherapies targeting TGF-ß, especially for cancer treatment. Nonetheless, recent work on combination therapies of TGF-ß inhibition and immunotherapy have provide insights into the development of TGF-ß-targeted therapies, with favorable outcomes in patients with advanced cancer. Hence, we summarize the entanglement between TGF-ß and the immune system in the developmental and tumor contexts and recent progress on hijacking crucial TGF-ß signaling pathways as an emerging area of cancer therapy.


Neoplasms , Transforming Growth Factor beta , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Carcinogenesis/immunology , Cell Transformation, Neoplastic , Humans , Immunotherapy , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Signal Transduction/immunology , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism
12.
Nature ; 606(7912): 172-179, 2022 06.
Article En | MEDLINE | ID: mdl-35545680

Missense driver mutations in cancer are concentrated in a few hotspots1. Various mechanisms have been proposed to explain this skew, including biased mutational processes2, phenotypic differences3-6 and immunoediting of neoantigens7,8; however, to our knowledge, no existing model weighs the relative contribution of these features to tumour evolution. We propose a unified theoretical 'free fitness' framework that parsimoniously integrates multimodal genomic, epigenetic, transcriptomic and proteomic data into a biophysical model of the rate-limiting processes underlying the fitness advantage conferred on cancer cells by driver gene mutations. Focusing on TP53, the most mutated gene in cancer1, we present an inference of mutant p53 concentration and demonstrate that TP53 hotspot mutations optimally solve an evolutionary trade-off between oncogenic potential and neoantigen immunogenicity. Our model anticipates patient survival in The Cancer Genome Atlas and patients with lung cancer treated with immunotherapy as well as the age of tumour onset in germline carriers of TP53 variants. The predicted differential immunogenicity between hotspot mutations was validated experimentally in patients with cancer and in a unique large dataset of healthy individuals. Our data indicate that immune selective pressure on TP53 mutations has a smaller role in non-cancerous lesions than in tumours, suggesting that targeted immunotherapy may offer an early prophylactic opportunity for the former. Determining the relative contribution of immunogenicity and oncogenic function to the selective advantage of hotspot mutations thus has important implications for both precision immunotherapies and our understanding of tumour evolution.


Carcinogenesis , Evolution, Molecular , Lung Neoplasms , Mutation , Carcinogenesis/genetics , Carcinogenesis/immunology , Datasets as Topic , Genes, p53 , Genetic Fitness , Genomics , Healthy Volunteers , Humans , Immunotherapy , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Mutation/genetics , Mutation, Missense , Reproducibility of Results
13.
Signal Transduct Target Ther ; 7(1): 19, 2022 01 20.
Article En | MEDLINE | ID: mdl-35046386

Interleukin-37b (hereafter called IL-37) was identified as fundamental inhibitor of natural and acquired immunity. The molecular mechanism and function of IL-37 in colorectal cancer (CRC) has been elusive. Here, we found that IL-37 transgenic (IL-37tg) mice were highly susceptible to colitis-associated colorectal cancer (CAC) and suffered from dramatically increased tumor burdens in colon. Nevertheless, IL-37 is dispensable for intestinal mutagenesis, and CRC cell proliferation, apoptosis, and migration. Notably, IL-37 dampened protective cytotoxic T cell-mediated immunity in CAC and B16-OVA models. CD8+ T cell dysfunction is defined by reduced retention and activation as well as failure to proliferate and produce cytotoxic cytokines in IL-37tg mice, enabling tumor evasion of immune surveillance. The dysfunction led by IL-37 antagonizes IL-18-induced proliferation and effector function of CD8+ T cells, which was dependent on SIGIRR (single immunoglobulin interleukin-1 receptor-related protein). Finally, we observed that IL-37 levels were significantly increased in CRC patients, and positively correlated with serum CRC biomarker CEA levels, but negatively correlated with the CD8+ T cell infiltration in CRC patients. Our findings highlight the role of IL-37 in harnessing antitumor immunity by inactivation of cytotoxic T cells and establish a new defined inhibitory factor IL-37/SIGIRR in cancer-immunity cycle as therapeutic targets in CRC.


Carcinogenesis/immunology , Colitis/immunology , Colorectal Neoplasms/immunology , Interleukin-1/immunology , Neoplasm Proteins/immunology , Receptors, Interleukin-1/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Carcinogenesis/genetics , Colitis/genetics , Colitis/pathology , Colorectal Neoplasms/genetics , Interleukin-1/genetics , Mice , Mice, Transgenic , Neoplasm Proteins/genetics , Receptors, Interleukin-1/genetics
14.
Mol Biol Rep ; 49(2): 1529-1535, 2022 Feb.
Article En | MEDLINE | ID: mdl-34981335

Infection processes induce various soluble factors that are carcinogens in humans; therefore, research into the soluble factors of chronic disease released from cells that have been infected with parasites is warranted. Parasitic infections in host cells release high levels of IFNγ. Studies have hypothesised that parasitosis-associated carcinogenesis might be analogous to colorectal cancers developed from inflammatory bowel diseases, whereby various cytokines and chemokines are secreted during chronic inflammation. IL-18 and IL-21 are other factors that might be involved in the development of colorectal cancer in schistosomiasis patients and patients with other infections. IL-21 has profound effects on tumour growth and immunosurveillance of colitis-associated tumourigenesis, thereby emphasising its involvement in the pathogenesis of colorectal cancer. The prominent role of IL-21 in antitumour effects greatly depends on the enhanced cytolytic activity of NK cells and the pathogenic role of IL-21, which is often associated with enhanced risks of cancer and chronic inflammatory processes. As IL-15 is also related to chronic disease, it is believed to also play a role in the antitumour effect of colorectal carcinogenesis. IL-15 generates and maintains long-term CD8+ T cell immunity against T. gondii to control the infection of intracellular pathogens. The lack of IL-15 in mice contributes to the downregulation of the IFNγ-producing CD4+ T cell response against acute T. gondii infection. IL-15 induces hyperplasia and supports the progressive growth of colon cancer via multiple functions. The limited role of IL-15 in the development of NK and CD8+ T cells suggests that there may be other cytokines compensating for the loss of the IL-15 gene.


Colorectal Neoplasms/immunology , Communicable Diseases/immunology , Cytokines/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , Carcinogenesis/immunology , Carcinogenesis/pathology , Colitis , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/physiopathology , Cytokines/immunology , Humans , Inflammation/pathology , Interleukin-15 , Interleukins , Killer Cells, Natural/pathology , Mice , Toxoplasma , Toxoplasmosis/complications , Toxoplasmosis/immunology
15.
Bioengineered ; 13(2): 3221-3239, 2022 02.
Article En | MEDLINE | ID: mdl-35067176

Secreted phosphoprotein 1 (SPP1) is involved in immune regulation, cell survival, and tumor progression. Studies have demonstrated that SPP1 plays an important role in certain individual tumors. However, the expression profile and oncogenic features of SPP1 in diverse cancers are remaining unknown. Therefore, we performed a comprehensive analysis using The Cancer Genome Atlas (TCGA) database. Raw data of 33 cancer types were download from the University of California Santa Cruz (UCSC) Xena website. The expression of SPP1 and its relationship with tumor prognosis, immune invasion, tumor microenvironment, and immunotherapy were analyzed using the R language. The function analysis was conducted using Gene Set Enrichment Analysis (GSEA). The oncogenic features of SPP1 was validated by wound-healing assay and EdU staining assay. SPP1 highly expressed in most cancers. The expression of SPP1 was significant related to prognosis, tumor mutation burden (TMB), microsatellite instability (MSI), and immune checkpoint genes, suggested that SPP1 plays an essential role in the tumor immune microenvironment and immune cell infiltration. The immune/stromal scores correlated positively with the SPP1 expression, and the relationship was affected by tumor heterogeneity and immunotherapy. In addition, SPP1 could predict the response of tumor immunotherapy. Functional analysis revealed the SPP1-associated terms and pathways. Finally, SPP1 significantly elevated cell proliferation and migration in A549, Huh7, HT-29, A2780 tumor cell lines. In conclusion, this study indicated that SPP1 involved in tumorigenesis, tumor progression, and regulated tumor immune microenvironment, revealing SPP1 might be a potential target for evaluating prognosis and immunotherapy in multiple cancers.


Biomarkers, Tumor/immunology , Databases, Nucleic Acid , Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , Osteopontin/immunology , A549 Cells , Biomarkers, Tumor/genetics , Carcinogenesis/genetics , Carcinogenesis/immunology , Female , HT29 Cells , Humans , Male , Neoplasms/diagnosis , Neoplasms/genetics , Osteopontin/genetics
16.
JCI Insight ; 7(2)2022 01 25.
Article En | MEDLINE | ID: mdl-35076024

Besides promoting inflammation by mobilizing lipid mediators, group IIA secreted phospholipase A2 (sPLA2-IIA) prevents bacterial infection by degrading bacterial membranes. Here, we show that, despite the restricted intestinal expression of sPLA2-IIA in BALB/c mice, its genetic deletion leads to amelioration of cancer and exacerbation of psoriasis in distal skin. Intestinal expression of sPLA2-IIA is reduced after treatment with antibiotics or under germ-free conditions, suggesting its upregulation by gut microbiota. Metagenome, transcriptome, and metabolome analyses have revealed that sPLA2-IIA deficiency alters the gut microbiota, accompanied by notable changes in the intestinal expression of genes related to immunity and metabolism, as well as in the levels of various blood metabolites and fecal bacterial lipids, suggesting that sPLA2-IIA contributes to shaping of the gut microbiota. The skin phenotypes in Pla2g2a-/- mice are lost (a) when they are cohoused with littermate WT mice, resulting in the mixing of the microbiota between the genotypes, or (b) when they are housed in a more stringent pathogen-free facility, where Pla2g2a expression in WT mice is low and the gut microbial compositions in both genotypes are nearly identical. Thus, our results highlight a potentially new aspect of sPLA2-IIA as a modulator of gut microbiota, perturbation of which affects distal skin responses.


Gastrointestinal Microbiome/immunology , Group II Phospholipases A2/metabolism , Psoriasis , Skin Neoplasms , Animals , Carcinogenesis/immunology , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation/immunology , Inflammation/microbiology , Mice , Mice, Inbred BALB C , Pathology, Molecular/methods , Psoriasis/immunology , Psoriasis/microbiology , Skin Neoplasms/immunology , Skin Neoplasms/microbiology
17.
Front Immunol ; 13: 957233, 2022.
Article En | MEDLINE | ID: mdl-36591314

Introduction: Colorectal cancer and other adult solid cancers pose a significant challenge for successful treatment because the tumor microenvironment both hinders the action of conventional therapeutics and suppresses the immune activities of infiltrating leukocytes. The immune suppression is largely the effect of enhanced local mediators such as purine nucleosides and eicosanoids. Genetic approaches have the promise of interfering with these mechanisms of local immunosuppression to allow both intrinsic and therapeutic immunological anticancer processes. Bacterial phages offer a novel means of enabling access into tissues for therapeutic genetic manipulations. Methods: We generated spheroids of fibroblastic and CRC cancer cells to model the 3-dimensional stromal and parenchymal components of colorectal tumours. We used these to examine the access and effects of both wildtype (WT) and epidermal growth factor (EGF)-presenting bacteriophage λ (WT- λ and EGF-λ) as a means of delivery of targeted genetic interventions in solid cancers. We used both confocal microscopy of spheroids exposed to AF488-tagged phages, and the recovery of viable phages as measured by plaque-forming assays to evaluate access; and measures of mitochondrial enzyme activity and cellular ATP to evaluate the outcome on the constituent cells. Results: Using flourescence-tagged derivatives of these bacteriophages (AF488-WT-λ and AF488-EGF-λ) we showed that phage entry into these tumour microenvironments was possible and that the EGF ligand enabled efficient and persistent uptake into the cancer cell mass. EGF-λ became localized in the intracellular portion of cancer cells and was subjected to subsequent cellular processing. The targeted λ phage had no independent effect upon mature tumour spheroids, but interfered with the early formation and growth of cancer tissues without the need for addition of a toxic payload, suggesting that it might have beneficial effects by itself in addition to any genetic intervention delivered to the tumour. Interference with spheroid formation persisted over the duration of culture. Discussion: We conclude that targeted phage technology is a feasible strategy to facilitate delivery into colorectal cancer tumour tissue (and by extension other solid carcinomas) and provides an appropriate delivery vehicle for a gene therapeutic that can reduce local immunosuppression and/or deliver an additional direct anticancer activity.


Bacteriophage lambda , Carcinogenesis , Colorectal Neoplasms , Tumor Microenvironment , Humans , Bacteriophage lambda/genetics , Bacteriophage lambda/immunology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Epidermal Growth Factor/genetics , Epidermal Growth Factor/immunology , ErbB Receptors/genetics , ErbB Receptors/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Carcinogenesis/genetics , Carcinogenesis/immunology
18.
Cancer Lett ; 525: 84-96, 2022 01 28.
Article En | MEDLINE | ID: mdl-34740608

Wnt/ß-catenin signaling is a highly conserved pathway that regulates cell proliferation, differentiation, apoptosis, stem cell self-renewal, tissue homeostasis, and wound healing. Dysregulation of the Wnt pathway is intricately involved in almost all stages of tumorigenesis in various cancers. Through direct and/or indirect effects on effector T cells, T-regulatory cells, T-helper cells, dendritic cells, and other cytokine-expressing immune cells, abnormal activation of Wnt/ß-catenin signaling benefits immune exclusion and hinders T-cell-mediated antitumor immune responses. Activation of Wnt signaling results in increased resistance to immunotherapies. In this review, we summarize the process by which Wnt signaling affects cancer and immune surveillance, and the potential for targeting the Wnt-signaling pathway via cancer immunotherapy.


Carcinogenesis/genetics , Immunotherapy , Neoplasms/immunology , Tumor Microenvironment/immunology , Carcinogenesis/immunology , Cell Proliferation/genetics , Cell Self Renewal/immunology , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/therapy , Th1 Cells/immunology , Wnt Signaling Pathway/genetics , Wnt Signaling Pathway/immunology
19.
Semin Cancer Biol ; 79: 91-104, 2022 02.
Article En | MEDLINE | ID: mdl-34280576

Beyond their well-known functions in the acute phases of the immune response, neutrophils play important roles in the various phases of tumor initiation and progression, through the release of their stored or newly synthesized mediators. In addition to reactive oxygen species, cytokines, chemokines, granule proteins and lipid mediators, neutrophil extracellular traps (NETs) can also be released upon neutrophil activation. NET formation can be achieved through a cell-death process or in association with the release of mitochondrial DNA from viable neutrophils. NETs are described as extracellular fibers of DNA and decorating proteins responsible for trapping and killing extracellular pathogens, playing a protective role in the antimicrobial defense. There is increasing evidence, however, that NETs play multiple roles in the scenario of cancer-related inflammation. For instance, NETs directly or indirectly promote tumor growth and progression, fostering tumor spread at distant sites and shielding cancer cells thus preventing the effects of cytotoxic lymphocytes. NETs can also promote tumor angiogenesis and cancer-associated thrombosis. On the other hand, there is some evidence that NETs may play anti-inflammatory and anti-tumorigenic roles. In this review, we focus on the main mechanisms underlying the emerging effects of NETs in cancer initiation and progression.


Carcinogenesis/pathology , Extracellular Traps/immunology , Neoplasms/pathology , Neutrophil Activation/immunology , Neutrophils/immunology , Carcinogenesis/immunology , Cytokines/metabolism , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Neoplasms/immunology , Neovascularization, Pathologic/pathology
20.
Nat Immunol ; 23(1): 99-108, 2022 01.
Article En | MEDLINE | ID: mdl-34937926

Enzymes of the TET family are methylcytosine dioxygenases that undergo frequent mutational or functional inactivation in human cancers. Recurrent loss-of-function mutations in TET proteins are frequent in human diffuse large B cell lymphoma (DLBCL). Here, we investigate the role of TET proteins in B cell homeostasis and development of B cell lymphomas with features of DLBCL. We show that deletion of Tet2 and Tet3 genes in mature B cells in mice perturbs B cell homeostasis and results in spontaneous development of germinal center (GC)-derived B cell lymphomas with increased G-quadruplexes and R-loops. At a genome-wide level, G-quadruplexes and R-loops were associated with increased DNA double-strand breaks (DSBs) at immunoglobulin switch regions. Deletion of the DNA methyltransferase DNMT1 in TET-deficient B cells prevented expansion of GC B cells, diminished the accumulation of G-quadruplexes and R-loops and delayed B lymphoma development, consistent with the opposing functions of DNMT and TET enzymes in DNA methylation and demethylation. Clustered regularly interspaced short palindromic repeats (CRISPR)-mediated depletion of nucleases and helicases that regulate G-quadruplexes and R-loops decreased the viability of TET-deficient B cells. Our studies suggest a molecular mechanism by which TET loss of function might predispose to the development of B cell malignancies.


B-Lymphocytes/immunology , Carcinogenesis/immunology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/immunology , Dioxygenases/immunology , Homeostasis/immunology , R-Loop Structures/immunology , Animals , Cell Differentiation/immunology , DNA Methylation/immunology , G-Quadruplexes , Germinal Center/immunology , Mice , Mice, Inbred C57BL
...