1
|
Liu H, Zhao Y, Du H, Hao P, Tian H, Wang K, Qiu Y, Dong H, Du Q, Tong D, Huang Y. IL-10 upregulates SOCS3 to inhibit type I interferon signaling to promote PoRVA replication in intestinal epithelial cells. Vet Microbiol 2024; 298:110259. [PMID: 39332165 DOI: 10.1016/j.vetmic.2024.110259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 09/29/2024]
Abstract
Porcine group A rotavirus (PoRVA) is one of the common enteric viruses causing severe diarrhea in piglets. Although PoRVA infection has been identified to promote IL-10 production, the role of IL-10 during viral infection remains unclear. In this study, we found that elevated IL-10 levels during PoRVA infection promote viral replication by inhibiting type I interferon production and response. IL-10 treatment upregulated the expression of SOCS3 in PoRVA-infected IPEC-J2 cells, which inhibited IFN-I production by preventing the degradation of IκB and nuclear translocation of NF-κB, thereby significantly promoting PoRVA replication. Furthermore, we determined that SOCS3 also inhibited type Ⅰ interferon signaling pathway, which led to a significantly reduced ISGs after IFN-α stimulation. In PoRVA-infected cells, overexpression of SOCS3 significantly inhibits phosphorylation and heterodimerization of STAT1, thereby promoting viral replication. Finally, we demonstrated the effect of IL-10 on PoRVA replication in vivo by murine models of PoRVA infection. PoRVA replication levels were lower in the ileum of IL-10 knockout (IL-10-/-) mice than that in PoRVA-infected wild-type mice, but PoRVA replication levels were higher in the ileum of IFNAR knockout (IFNAR-/-) mice than that in PoRVA-infected wild-type mice. Taken together, our findings provide information to understand the strategies of PoRVA to evade host innate antiviral immunity.
Collapse
Affiliation(s)
- Haixin Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yongpan Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, China; Shaanxi Animal Husbandry Experimental and Demonstration Center, China
| | - Huimin Du
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Pengcheng Hao
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Haolun Tian
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Kun Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yudong Qiu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Haiying Dong
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Qian Du
- College of Veterinary Medicine, Northwest A&F University, Yangling, China; Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education, Yangling, China; Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Yangling, China; Engineering Research Center of Efficient New Vaccines for Animals, Universities of Shaanxi Province, Yangling, China
| | - Dewen Tong
- College of Veterinary Medicine, Northwest A&F University, Yangling, China; Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education, Yangling, China; Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Yangling, China; Engineering Research Center of Efficient New Vaccines for Animals, Universities of Shaanxi Province, Yangling, China.
| | - Yong Huang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China; Engineering Research Center of Efficient New Vaccines for Animals, Ministry of Education, Yangling, China; Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Yangling, China; Engineering Research Center of Efficient New Vaccines for Animals, Universities of Shaanxi Province, Yangling, China.
| |
Collapse
|
2
|
Lu B, Liu Y, Yao Y, Zhu D, Zhang X, Dong K, Xu X, Lv D, Zhao Z, Zhang H, Yang X, Fu W, Huang R, Cao J, Chu J, Pan X, Cui X. Unveiling the unique role of TSPAN7 across tumors: a pan-cancer study incorporating retrospective clinical research and bioinformatic analysis. Biol Direct 2024; 19:72. [PMID: 39175035 PMCID: PMC11340126 DOI: 10.1186/s13062-024-00516-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 08/08/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND TSPAN7 is an important factor in tumor progression. However, the precise function of TSPAN7 and its role in pan-cancer are not clear. METHODS Based on Xinhua cohort incorporating 370 patients with kidney neoplasm, we conducted differential expression analysis by immunohistochemistry between tumor and normal tissues, and explored correlations of TSPAN7 with patients' survival. Subsequently, we conducted a pan-cancer study, and successively employed differential expression analysis, competing endogenous RNA (ceRNA) analysis, protein-protein interaction (PPI) analysis, correlation analysis of TSPAN7 with clinical characteristics, tumor purity, tumor genomics, tumor immunity, and drug sensitivity. Last but not least, gene set enrichment analysis was applied to identify enriched pathways of TSPAN7. RESULTS In Xinhua cohort, TSPAN7 expression was significantly up-regulated (P-value = 0.0019) in tumor tissues of kidney neoplasm patients. High TSPAN7 expression was associated with decreases in overall survival (OS) (P-value = 0.009) and progression-free survival (P-value = 0.009), and it was further revealed as an independent risk factor for OS (P-value = 0.0326, HR = 5.66, 95%CI = 1.155-27.8). In pan-cancer analysis, TSPAN7 expression was down-regulated in most tumors, and it was associated with patients' survival, tumor purity, tumor genomics, tumor immunity, and drug sensitivity. The ceRNA network and PPI network of TSPAN7 were also constructed. Last but not least, the top five enriched pathways of TSPAN7 in various tumors were identified. CONCLUSION TSPAN7 served as a promising biomarker of various tumors, especially kidney neoplasms, and it was closely associated with tumor purity, tumor genomics, tumor immunology, and drug sensitivity in pan-cancer level.
Collapse
Affiliation(s)
- Bingnan Lu
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China
| | - Yifan Liu
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China
| | - Yuntao Yao
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China
| | - Dawei Zhu
- Department of Urology, the Second People's Hospital of Pinghu, Zhejiang, 314200, China
| | - Xiangmin Zhang
- Department of Urology, Shanghai Baoshan Luodian Hospital, Shanghai, 201908, China
| | - Keqin Dong
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China
| | - Xiao Xu
- Department of Urology, the Second People's Hospital of Pinghu, Zhejiang, 314200, China
| | - Donghao Lv
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China
| | - Zihui Zhao
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China
| | - Haoyu Zhang
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China
| | - Xinyue Yang
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China
| | - Wenjia Fu
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China
| | - Runzhi Huang
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China.
| | - Jianwei Cao
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China.
- Department of Urology, the Second People's Hospital of Pinghu, Zhejiang, 314200, China.
| | - Jian Chu
- Department of Urology, Shanghai Baoshan Luodian Hospital, Shanghai, 201908, China.
| | - Xiuwu Pan
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China.
| | - Xingang Cui
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China.
| |
Collapse
|
3
|
Xiong Y, Chen C, He C, Yang X, Cheng W. Identification of shared gene signatures and biological mechanisms between preeclampsia and polycystic ovary syndrome. Heliyon 2024; 10:e29225. [PMID: 38638956 PMCID: PMC11024567 DOI: 10.1016/j.heliyon.2024.e29225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 03/24/2024] [Accepted: 04/03/2024] [Indexed: 04/20/2024] Open
Abstract
Preeclampsia (PE) is one of the most common complications of pregnancy and polycystic ovary syndrome (PCOS) is a prevalent metabolic and endocrinopathy disorder in women of reproductive age. Identifying the shared genetic signatures and molecular mechanisms between PCOS and PE was the objective of this study. The intersections of WGCNA module genes, PPI module genes, and PPI hub genes revealed that 8 immunity-related genes might be shared causative genes of PE and PCOS. Further, qRT-PCR results showed that TSIX/miR-223-3p/DDX58 might play a crucial role in immune dysregulation in PE and PCOS and Spearman rank correlation analysis results illustrated the potential of DDX58 as a novel diagnostic and therapeutic target for PE and PCOS. Our study demonstrated a common disease pathway model TSIX/miR-223-3p/DDX58, illustrating that immune dysregulation may be a possible mechanism of PE and PCOS, and revealed that DDX58 might be a novel predictive target for PE and PCOS.
Collapse
Affiliation(s)
- Yaoxi Xiong
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 200030, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, 200030, Shanghai, China
| | - Chao Chen
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 200030, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, 200030, Shanghai, China
| | - Chengrong He
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 200030, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, 200030, Shanghai, China
| | - Xingyu Yang
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 200030, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, 200030, Shanghai, China
| | - Weiwei Cheng
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 200030, Shanghai, China
| |
Collapse
|
4
|
Rady T, Erb S, Deddouche-Grass S, Morales R, Chaubet G, Cianférani S, Basse N, Wagner A. Targeted delivery of immune-stimulating bispecific RNA, inducing apoptosis and anti-tumor immunity in cancer cells. iScience 2024; 27:109068. [PMID: 38380254 PMCID: PMC10877685 DOI: 10.1016/j.isci.2024.109068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 07/18/2023] [Accepted: 01/25/2024] [Indexed: 02/22/2024] Open
Abstract
Double-stranded RNAs (dsRNA)-based strategies appeared as promising therapies to induce an inflammation in the tumor microenvironment. However, currently described systems generally lack active targeting of tissues, and their clinical translation is thus limited to intratumoral injection. Herein, we developed an antibody-siRNA-5'triphosphate conjugate with multiple modes of action, combining cell surface EphA2-specific internalization, leading to a simultaneous gene silencing and activation of the receptor retinoic acid-inducible gene I (RIG-I). Recognition of cytosolic siRNA-5'triphosphate by RIG-I triggers the expression of interferons and pro-inflammatory cytokines, inducing an inflammation of the tumor environment and activating neighboring immune cells. In addition, these RIG-I-specific effects synergized with siRNA-mediated PLK1 silencing to promote cancer cell death by apoptosis. Altogether, such immune-stimulating antibody-RNA conjugate opens a novel modality to overcome some limitations encountered by dsRNA molecules currently in clinical trials.
Collapse
Affiliation(s)
- Tony Rady
- Bio-Functional Chemistry (UMR 7199), LabEx Medalis, University of Strasbourg, 74 Route du Rhin, 67400 Illkirch-Graffenstaden, France
- Sanofi, 13 Quai Jules Guesde, 94400 Vitry-sur-Seine, France
| | - Stéphane Erb
- Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
- Infrastructure Nationale de Protéomique ProFI – FR2048, 67087 Strasbourg, France
| | | | - Renaud Morales
- Sanofi, 13 Quai Jules Guesde, 94400 Vitry-sur-Seine, France
| | - Guilhem Chaubet
- Bio-Functional Chemistry (UMR 7199), LabEx Medalis, University of Strasbourg, 74 Route du Rhin, 67400 Illkirch-Graffenstaden, France
| | - Sarah Cianférani
- Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
- Infrastructure Nationale de Protéomique ProFI – FR2048, 67087 Strasbourg, France
| | - Nicolas Basse
- Sanofi, 13 Quai Jules Guesde, 94400 Vitry-sur-Seine, France
| | - Alain Wagner
- Bio-Functional Chemistry (UMR 7199), LabEx Medalis, University of Strasbourg, 74 Route du Rhin, 67400 Illkirch-Graffenstaden, France
| |
Collapse
|
5
|
Chen Y, Jiang Q, Qing F, Xue J, Xiao Q, He W, Sui L, Liu Z. MDA5 Enhances Invasive Candida albicans Infection by Regulating Macrophage Apoptosis and Phagocytosis/Killing Functions. Inflammation 2024; 47:191-208. [PMID: 37740789 DOI: 10.1007/s10753-023-01903-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/24/2023] [Accepted: 09/08/2023] [Indexed: 09/25/2023]
Abstract
Candida albicans is a common opportunistic pathogenic fungus. The innate immune system provides the first-line host defense against fungal infection. Innate immune receptors and downstream molecules have been shown to play various roles during fungal infection. The innate immune receptor MDA5, encoded by the gene Ifih1, enhances host resistance against viral and Aspergillus fumigatus infection by inducing the production of interferons (IFNs). However, the role of MDA5 in C. albicans infection is still unclear. Here, we found that the gene expression levels of IFIH1 were significantly increased in innate immune cells after C. albicans stimulation through human bioinformatics analysis or mouse experiments. Through in vivo study, MDA5 was shown to enhance host susceptibility to C. albicans infection independent of IFN production. Instead, MDA5 exerted its influence on macrophages and kidneys by modulating the expression of Noxa, Bcl2, and Bax, thereby promoting apoptosis. Additionally, MDA5 compromised killing capabilities of macrophage by inhibition iNOS expression. The introduction of the apoptosis inducer PAC1 further impaired macrophage functions, mimicking the enhancing effect of MDA5 on C. albicans infection. Furthermore, the administration of macrophage scavengers increased the susceptibility of Ifih1-/- mice to C. albicans. The founding suggests that MDA5 promote host susceptibility to invasive C. albicans by enhancing cell apoptosis and compromising macrophage functions, making MDA5 a target to treat candidiasis.
Collapse
Affiliation(s)
- Yayun Chen
- School of Graduate, China Medical University, Shenyang, Liaoning, China
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Qian Jiang
- School of Graduate, China Medical University, Shenyang, Liaoning, China
- School of Nursing, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Furong Qing
- School of Graduate, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Junxia Xue
- School of Graduate, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Qiuxiang Xiao
- Department of Pathology, The First Affiliated Hospital, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Wenji He
- School of Graduate, China Medical University, Shenyang, Liaoning, China
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Lina Sui
- School of Graduate, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Zhiping Liu
- School of Graduate, China Medical University, Shenyang, Liaoning, China.
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China.
- Center for Scientific Research, Gannan Medical University, Ganzhou, Jiangxi, 341000, China.
- Center for Immunology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, 341000, China.
| |
Collapse
|
6
|
Song H, Liu H, Wang X, Yang Y, Zhao X, Jiang WG, Sui L, Song X. Death-associated protein 3 in cancer-discrepant roles of DAP3 in tumours and molecular mechanisms. Front Oncol 2024; 13:1323751. [PMID: 38352299 PMCID: PMC10862491 DOI: 10.3389/fonc.2023.1323751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 12/30/2023] [Indexed: 02/16/2024] Open
Abstract
Cancer, ranks as the secondary cause of death, is a group of diseases that are characterized by uncontrolled tumor growth and distant metastasis, leading to increased mortality year-on-year. To date, targeted therapy to intercept the aberrant proliferation and invasion is crucial for clinical anticancer treatment, however, mutant expression of target genes often leads to drug resistance. Therefore, it is essential to identify more molecules that can be targeted to facilitate combined therapy. Previous studies showed that death associated protein 3 (DAP3) exerts a pivotal role in regulating apoptosis signaling of tumors, meanwhile, aberrant DAP3 expression is associated with the tumorigenesis and disease progression of various cancers. This review provides an overview of the molecule structure of DAP3 and the discrepant roles played by DAP3 in various types of tumors. Considering the molecular mechanism of DAP3-regulated cancer development, new potential treatment strategies might be developed in the future.
Collapse
Affiliation(s)
- Hao Song
- The Second Medical College, Binzhou Medical University, Yantai, China
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Huifang Liu
- The Second Medical College, Binzhou Medical University, Yantai, China
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Xiufeng Wang
- Department of Nursing, Zhaoyuan People's Hospital, Yantai, China
| | - Yuteng Yang
- The Second Medical College, Binzhou Medical University, Yantai, China
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Xiangkun Zhao
- The Second Medical College, Binzhou Medical University, Yantai, China
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Wen G. Jiang
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Laijian Sui
- Department of Orthopedics, Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Xicheng Song
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| |
Collapse
|
7
|
Icard P, Alifano M, Simula L. The potential for citrate to reinforce epigenetic therapy by promoting apoptosis. Trends Endocrinol Metab 2023; 34:586-589. [PMID: 37550099 DOI: 10.1016/j.tem.2023.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/04/2023] [Accepted: 07/18/2023] [Indexed: 08/09/2023]
Abstract
Epigenetic drugs induce ATP depletion, promoting a glycolysis-to-oxidative phosphorylation (OXPHOS) shift which sometimes favors tumor growth by promoting necroptosis over apoptosis. To restore effective apoptosis in tumors, we propose that the administration of citrate could inhibit ATP production, activate caspase-8 (a key necroptosis inhibitor), and downregulate key anti-apoptotic proteins (Bcl-xL and MCL1).
Collapse
Affiliation(s)
- Philippe Icard
- Normandie University, UNICAEN, INSERM U1086 Interdisciplinary Research Unit for Cancer Prevention and Treatment, Caen, France; Service de Chirurgie Thoracique, Hôpital Cochin, Hôpitaux Universitaires Paris Centre, APHP, Université Paris-Descartes, Paris, France.
| | - Marco Alifano
- Service de Chirurgie Thoracique, Hôpital Cochin, Hôpitaux Universitaires Paris Centre, APHP, Université Paris-Descartes, Paris, France; INSERM U1138, Integrative Cancer Immunology, University of Paris, 75006 Paris, France
| | - Luca Simula
- Cochin Institute, INSERM U1016, CNRS UMR8104, University of Paris-Cité, Paris, 75014, France
| |
Collapse
|
8
|
Li J, Wan C, Li X, Quan C, Li X, Wu X. Characterization of tumor microenvironment and tumor immunology based on the double-stranded RNA-binding protein related genes in cervical cancer. J Transl Med 2023; 21:647. [PMID: 37735483 PMCID: PMC10515034 DOI: 10.1186/s12967-023-04505-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 09/05/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND Cervical cancer is one of the most common gynecological cancers threatening women's health worldwide. Double-stranded RNA-binding proteins (dsRBPs) regulate innate immunity and are therefore believed to be involved in virus-related malignancies, however, their role in cervical cancer is not well known. METHODS We performed RNA-seq of tumor samples from cervical cancer patients in local cohort and also assessed the RNA-seq and clinical data derived from public datasets. By using single sample Gene Set Enrichment Analysis (ssGSEA) and univariate Cox analysis, patients were stratified into distinct dsRBP clusters. Stepwise Cox and CoxBoost were performed to construct a risk model based on optimal dsRBPs clusters-related differentially expressed genes (DEGs), and GSE44001 and CGCI-HTMCP-CC were employed as two external validation cohorts. Single cell RNA sequencing data from GSE168652 and Scissor algorithm were applied to evaluated the signature-related cell population. RESULTS The expression of dsRBP features was found to be associated with HPV infection and carcinogenesis in CESC. However, only Adenosine deaminases acting on RNA (ADAR) and Dicer, Drosha, and Argonautes (DDR) exhibited significant correlations with the overall survival (OS) of CESC patients. Based on these findings, CESC patients were divided into three dsRBP clusters. Cluster 3 showed superior OS but lower levels of ADAR and DDR. Additionally, Cluster 3 demonstrated enhanced innate immunity, with significantly higher activity in cancer immunity cycles, immune scores, and levels of tumor-infiltrating immune cells, particularly CD8+ T cells. Furthermore, a risk model based on nine dsRBP cluster-related DEGs was established. The accuracy of survival prediction for 1 to 5 years was consistently above 0.78, and this model's robust predictive capacity was confirmed by two external validation sets. The low-risk group exhibited significantly higher levels of immune checkpoints, such as PDCD1 and CTLA4, as well as a higher abundance of CD8+ T cells. Analysis of single-cell sequencing data revealed a significant association between the dsRBP signature and glycolysis. Importantly, low-risk patients showed improved OS and a higher response rate to immunotherapy, along with enduring clinical benefits from concurrent chemoradiotherapy. CONCLUSIONS dsRBP played a crucial role in the regulation of prognosis and tumor immunology in cervical cancer, and its prognostic signature provides a strategy for risk stratification and immunotherapy evaluation.
Collapse
Affiliation(s)
- Jin Li
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, No. 270 Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Chong Wan
- Precision Medicine Center, Yangtze Delta Region Institute of Tsinghua University, Jiaxing, China
| | - Xiaoqi Li
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, No. 270 Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Chenlian Quan
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, No. 270 Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xiaoqiu Li
- Department of Pathology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xiaohua Wu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, No. 270 Dong'an Road, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
9
|
Marek J, Hanesch L, Krabbe T, El Khawanky N, Heidegger S, Altomonte J. Oncolytic virotherapy with chimeric VSV-NDV synergistically supports RIG-I-dependent checkpoint inhibitor immunotherapy. Mol Ther Oncolytics 2023; 30:117-131. [PMID: 37654972 PMCID: PMC10465858 DOI: 10.1016/j.omto.2023.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 08/03/2023] [Indexed: 09/02/2023] Open
Abstract
Unraveling the complexities of the tumor microenvironment (TME) and its correlation with responsiveness to immunotherapy has become a main focus in overcoming resistance to such treatments. Targeting tumor-intrinsic retinoic acid-inducible gene-I (RIG-I), a sensor for viral RNA, was shown to transform the TME from an immunogenically "cold" state to an inflamed, "hot" lesion, which we demonstrated previously to be a crucial mediator of the efficacy of immune checkpoint inhibition with anti-cytotoxic T lymphocyte-associated protein 4 (CTLA-4). In this study, we focus on the chimeric oncolytic virus vesicular stomatitis virus (VSV)-Newcastle disease virus (NDV), comprised of genetic components of VSV and NDV, and we investigate its utility to support tumor-intrinsic RIG-I-dependent therapy with anti-CTLA-4. Overall, we demonstrate that treatment with VSV-NDV efficiently delays tumor growth and significantly prolongs survival in a murine model of malignant melanoma, which was further enhanced in combination with anti-CTLA-4. Although the direct oncolytic and pro-inflammatory effects of VSV-NDV therapy were independent of RIG-I activation, the synergism with anti-CTLA-4 therapy and associated activation of tumor-specific T cells was critically dependent on active RIG-I signaling in tumor cells. This work highlights the therapeutic value of utilizing an immune-stimulatory oncolytic virus to sensitize tumors to immune checkpoint inhibition.
Collapse
Affiliation(s)
- Janina Marek
- Department of Internal Medicine II, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Lorenz Hanesch
- Department of Internal Medicine II, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Teresa Krabbe
- Department of Internal Medicine II, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Nadia El Khawanky
- Department of Medicine III, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
| | - Simon Heidegger
- Department of Medicine III, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
| | - Jennifer Altomonte
- Department of Internal Medicine II, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| |
Collapse
|
10
|
Chen D, Ji Q, Liu J, Cheng F, Zheng J, Ma Y, He Y, Zhang J, Song T. MicroRNAs in the Regulation of RIG-I-like Receptor Signaling Pathway: Possible Strategy for Viral Infection and Cancer. Biomolecules 2023; 13:1344. [PMID: 37759744 PMCID: PMC10526236 DOI: 10.3390/biom13091344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 08/30/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
The retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs) play a crucial role as pattern-recognition receptors within the innate immune system. These receptors, present in various cell and tissue types, serve as essential sensors for viral infections, enhancing the immune system's capacity to combat infections through the induction of type I interferons (IFN-I) and inflammatory cytokines. RLRs are involved in a variety of physiological and pathological processes, including viral infections, autoimmune disorders, and cancer. An increasing body of research has examined the possibility of RLRs or microRNAs as therapeutic targets for antiviral infections and malignancies, despite the fact that few studies have focused on the regulatory function of microRNAs on RLR signaling. Consequently, our main emphasis in this review is on elucidating the role of microRNAs in modulating the signaling pathways of RLRs in the context of cancer and viral infections. The aim is to establish a robust knowledge base that can serve as a basis for future comprehensive investigations into the interplay between microRNAs and RIG-I, while also facilitating the advancement of therapeutic drug development.
Collapse
Affiliation(s)
- Dengwang Chen
- Department of Immunology, Zunyi Medical University, Zunyi 563002, China; (D.C.); (J.L.); (F.C.); (J.Z.); (Y.M.)
| | - Qinglu Ji
- School of Pharmacy, Zunyi Medical University, Zunyi 563002, China; (Q.J.); (Y.H.)
| | - Jing Liu
- Department of Immunology, Zunyi Medical University, Zunyi 563002, China; (D.C.); (J.L.); (F.C.); (J.Z.); (Y.M.)
| | - Feng Cheng
- Department of Immunology, Zunyi Medical University, Zunyi 563002, China; (D.C.); (J.L.); (F.C.); (J.Z.); (Y.M.)
| | - Jishan Zheng
- Department of Immunology, Zunyi Medical University, Zunyi 563002, China; (D.C.); (J.L.); (F.C.); (J.Z.); (Y.M.)
| | - Yunyan Ma
- Department of Immunology, Zunyi Medical University, Zunyi 563002, China; (D.C.); (J.L.); (F.C.); (J.Z.); (Y.M.)
| | - Yuqi He
- School of Pharmacy, Zunyi Medical University, Zunyi 563002, China; (Q.J.); (Y.H.)
| | - Jidong Zhang
- Department of Immunology, Zunyi Medical University, Zunyi 563002, China; (D.C.); (J.L.); (F.C.); (J.Z.); (Y.M.)
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi 563002, China
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563002, China
| | - Tao Song
- Department of Immunology, Zunyi Medical University, Zunyi 563002, China; (D.C.); (J.L.); (F.C.); (J.Z.); (Y.M.)
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi 563002, China
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563002, China
| |
Collapse
|
11
|
Schweibenz BD, Solotchi M, Hanpude P, Devarkar S, Patel S. RIG-I recognizes metabolite-capped RNAs as signaling ligands. Nucleic Acids Res 2023; 51:8102-8114. [PMID: 37326006 PMCID: PMC10450190 DOI: 10.1093/nar/gkad518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 06/12/2023] [Indexed: 06/17/2023] Open
Abstract
The innate immune receptor RIG-I recognizes 5'-triphosphate double-stranded RNAs (5' PPP dsRNA) as pathogenic RNAs. Such RNA-ends are present in viral genomes and replication intermediates, and they activate the RIG-I signaling pathway to produce a potent interferon response essential for viral clearance. Endogenous mRNAs cap the 5' PPP-end with m7G and methylate the 2'-O-ribose to evade RIG-I, preventing aberrant immune responses deleterious to the cell. Recent studies have identified RNAs in cells capped with metabolites such as NAD+, FAD and dephosphoCoA. Whether RIG-I recognizes these metabolite-capped RNAs has not been investigated. Here, we describe a strategy to make metabolite-capped RNAs free from 5' PPP dsRNA contamination, using in vitro transcription initiated with metabolites. Mechanistic studies show that metabolite-capped RNAs have a high affinity for RIG-I, stimulating the ATPase activity at comparable levels to 5' PPP dsRNA. Cellular signaling assays show that the metabolite-capped RNAs potently stimulate the innate antiviral immune response. This demonstrates that RIG-I can tolerate diphosphate-linked, capped RNAs with bulky groups at the 5' RNA end. This novel class of RNAs that stimulate RIG-I signaling may have cellular roles in activating the interferon response and may be exploited with proper functionalities for RIG-I-related RNA therapeutics.
Collapse
Affiliation(s)
- Brandon D Schweibenz
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
- Graduate School of Biomedical Sciences at the Robert Wood Johnson Medical School of Rutgers University, USA
| | - Mihai Solotchi
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
- Graduate School of Biomedical Sciences at the Robert Wood Johnson Medical School of Rutgers University, USA
| | - Pranita Hanpude
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Swapnil C Devarkar
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
- Graduate School of Biomedical Sciences at the Robert Wood Johnson Medical School of Rutgers University, USA
| | - Smita S Patel
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
12
|
Du G, Xing Z, Zhou J, Cui C, Liu C, Liu Y, Li Z. Retinoic acid-inducible gene-I like receptor pathway in cancer: modification and treatment. Front Immunol 2023; 14:1227041. [PMID: 37662910 PMCID: PMC10468571 DOI: 10.3389/fimmu.2023.1227041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 07/28/2023] [Indexed: 09/05/2023] Open
Abstract
Retinoic acid-inducible gene-I (RIG-I) like receptor (RLR) pathway is one of the most significant pathways supervising aberrant RNA in cells. In predominant conditions, the RLR pathway initiates anti-infection function via activating inflammatory effects, while recently it is discovered to be involved in cancer development as well, acting as a virus-mimicry responder. On one hand, the product IFNs induces tumor elimination. On the other hand, the NF-κB pathway is activated which may lead to tumor progression. Emerging evidence demonstrates that a wide range of modifications are involved in regulating RLR pathways in cancer, which either boost tumor suppression effect or prompt tumor development. This review summarized current epigenetic modulations including DNA methylation, histone modification, and ncRNA interference, as well as post-transcriptional modification like m6A and A-to-I editing of the upstream ligand dsRNA in cancer cells. The post-translational modulations like phosphorylation and ubiquitylation of the pathway's key components were also discussed. Ultimately, we provided an overview of the current therapeutic strategies targeting the RLR pathway in cancers.
Collapse
Affiliation(s)
- Guangyuan Du
- NHC Key Laboratory of Carcinogenesis, National Clinical Research Center for Geriatric Disorders, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Clinical Medicine, Xingya School of Medicine of Central South University, Changsha, China
| | - Zherui Xing
- NHC Key Laboratory of Carcinogenesis, National Clinical Research Center for Geriatric Disorders, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Clinical Medicine, Xingya School of Medicine of Central South University, Changsha, China
| | - Jue Zhou
- NHC Key Laboratory of Carcinogenesis, National Clinical Research Center for Geriatric Disorders, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Clinical Medicine, Xingya School of Medicine of Central South University, Changsha, China
| | - Can Cui
- NHC Key Laboratory of Carcinogenesis, National Clinical Research Center for Geriatric Disorders, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Clinical Medicine, Xingya School of Medicine of Central South University, Changsha, China
| | - Chenyuan Liu
- NHC Key Laboratory of Carcinogenesis, National Clinical Research Center for Geriatric Disorders, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Clinical Medicine, Xingya School of Medicine of Central South University, Changsha, China
| | - Yiping Liu
- NHC Key Laboratory of Carcinogenesis, National Clinical Research Center for Geriatric Disorders, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Clinical Medicine, Xingya School of Medicine of Central South University, Changsha, China
| | - Zheng Li
- NHC Key Laboratory of Carcinogenesis, National Clinical Research Center for Geriatric Disorders, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| |
Collapse
|
13
|
Thierry S, Maadadi S, Berton A, Dimier L, Perret C, Vey N, Ourfali S, Saccas M, Caron S, Boucard-Jourdin M, Colombel M, Werle B, Bonnin M. TL-532, a novel specific Toll-like receptor 3 agonist rationally designed for targeting cancers: discovery process and biological characterization. MICROBIAL CELL (GRAZ, AUSTRIA) 2023; 10:117-132. [PMID: 37275475 PMCID: PMC10236204 DOI: 10.15698/mic2023.06.797] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/22/2023] [Accepted: 04/12/2023] [Indexed: 06/07/2023]
Abstract
Toll-like receptor 3 (TLR3) is an innate immune receptor that recognizes double-stranded RNA (dsRNA) and induces inflammation in immune and normal cells to initiate anti-microbial responses. TLR3 acts also as a death receptor only in cancer cells but not in their normal counterparts, making it an attractive target for cancer therapies. To date, all of the TLR3-activating dsRNAs used at preclinical or clinical stages have major drawbacks such as structural heterogeneity, toxicity, and lack of specificity and/or efficacy. We conducted the discovery process of a new family of TLR3 agonists that are chemically manufactured on solid-phase support and perfectly defined in terms of sequence and size. A stepwise discovery process was performed leading to the identification of TL-532, a 70 base pair dsRNA that is potent without transfection reagent and is highly specific for TLR3 without activating other innate nucleic sensors such as RIG-I/MDA5, TLR7, TLR8, and TLR9. TL-532 induces inflammation in murine RAW264.7 myeloid macrophages, in human NCI-H292 lung cancer cells, and it promotes immunogenic apoptosis in tumor cells in vitro and ex vivo without toxicity towards normal primary cells. In conclusion, we identified a novel TLR3 agonist called TL-532 that has promising anticancer properties.
Collapse
Affiliation(s)
- Sylvain Thierry
- TOLLYS SAS, 60F avenue Rockefeller, Lyon, France; Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France
| | - Sarah Maadadi
- TOLLYS SAS, 60F avenue Rockefeller, Lyon, France; Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France
| | - Aurore Berton
- TOLLYS SAS, 60F avenue Rockefeller, Lyon, France; Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France
| | - Laura Dimier
- TOLLYS SAS, 60F avenue Rockefeller, Lyon, France; Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France
| | - Clémence Perret
- TOLLYS SAS, 60F avenue Rockefeller, Lyon, France; Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France
| | - Nelly Vey
- TOLLYS SAS, 60F avenue Rockefeller, Lyon, France; Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France
| | - Saïd Ourfali
- Service d'Urologie et Chirurgie de la Transplantation, Hospices Civils de Lyon, Lyon, France. Université Claude Bernard Lyon 1; TOLLYS SAS, 60F avenue Rockefeller, Lyon, France; Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France
| | - Mathilde Saccas
- TOLLYS SAS, 60F avenue Rockefeller, Lyon, France; Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France
| | - Solène Caron
- TOLLYS SAS, 60F avenue Rockefeller, Lyon, France; Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France
| | - Mathilde Boucard-Jourdin
- TOLLYS SAS, 60F avenue Rockefeller, Lyon, France; Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France
| | - Marc Colombel
- Service d'Urologie et Chirurgie de la Transplantation, Hospices Civils de Lyon, Lyon, France; Univ Lyon, Université Claude Bernard Lyon 1
| | - Bettina Werle
- TOLLYS SAS, 60F avenue Rockefeller, Lyon, France; Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France
| | - Marc Bonnin
- TOLLYS SAS, 60F avenue Rockefeller, Lyon, France; Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France
| |
Collapse
|
14
|
Yao Y, Kong W, Yang L, Ding Y, Cui H. Immunity and Immune Evasion Mechanisms of Epstein-Barr Virus. Viral Immunol 2023; 36:303-317. [PMID: 37285188 DOI: 10.1089/vim.2022.0200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023] Open
Abstract
Epstein-Barr virus (EBV) is the first human oncogenic virus to be identified, which evades the body's immune surveillance through multiple mechanisms that allow long-term latent infection. Under certain pathological conditions, EBVs undergo a transition from the latent phase to the lytic phase and cause targeted dysregulation of the host immune system, leading to the development of EBV-related diseases. Therefore, an in-depth understanding of the mechanism of developing an immune response to EBV and the evasion of immune recognition by EBV is important for the understanding of the pathogenesis of EBV, which is of great significance for finding strategies to prevent EBV infection, and developing a therapy to treat EBV-associated diseases. In this review, we will discuss the molecular mechanisms of host immunological responses to EBV infection and the mechanisms of EBV-mediated immune evasion during chronic active infection.
Collapse
Affiliation(s)
- Yanqing Yao
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Weijing Kong
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Lijun Yang
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yingxue Ding
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Hong Cui
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
15
|
Zheng J, Shi W, Yang Z, Chen J, Qi A, Yang Y, Deng Y, Yang D, Song N, Song B, Luo D. RIG-I-like receptors: Molecular mechanism of activation and signaling. Adv Immunol 2023; 158:1-74. [PMID: 37453753 DOI: 10.1016/bs.ai.2023.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
During RNA viral infection, RIG-I-like receptors (RLRs) recognize the intracellular pathogenic RNA species derived from viral replication and activate antiviral innate immune response by stimulating type 1 interferon expression. Three RLR members, namely, RIG-I, MDA5, and LGP2 are homologous and belong to a subgroup of superfamily 2 Helicase/ATPase that is preferably activated by double-stranded RNA. RLRs are significantly different in gene architecture, RNA ligand preference, activation, and molecular functions. As switchable macromolecular sensors, RLRs' activities are tightly regulated by RNA ligands, ATP, posttranslational modifications, and cellular cofactors. We provide a comprehensive review of the structure and function of the RLRs and summarize the molecular understanding of sensing and signaling events during the RLR activation process. The key roles RLR signaling play in both anti-infection and immune disease conditions highlight the therapeutic potential in targeting this important molecular pathway.
Collapse
Affiliation(s)
- Jie Zheng
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
| | - Wenjia Shi
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Ziqun Yang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jin Chen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Ao Qi
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yulin Yang
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Ying Deng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Dongyuan Yang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Ning Song
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Bin Song
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Dahai Luo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore; NTU Institute of Structural Biology, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
16
|
Li L, Lv L, Xu JC, He Q, Chang N, Cui YY, Tao ZC, Zhu T, Qian LT. RIG-I Promotes Tumorigenesis and Confers Radioresistance of Esophageal Squamous Cell Carcinoma by Regulating DUSP6. Int J Mol Sci 2023; 24:ijms24065586. [PMID: 36982663 PMCID: PMC10052926 DOI: 10.3390/ijms24065586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/07/2023] [Accepted: 03/12/2023] [Indexed: 03/17/2023] Open
Abstract
We investigated the expression and biological function of retinoic acid inducible gene I (RIG-I) in esophageal squamous cell carcinoma (ESCC). Materials and methods: An immunohistochemical analysis was performed on 86 pairs of tumor tissue and adjacent normal tissue samples of patients with ESCC. We generated RIG-I-overexpressing ESCC cell lines KYSE70 and KYSE450, and RIG-I- knockdown cell lines KYSE150 and KYSE510. Cell viability, migration and invasion, radioresistance, DNA damage, and cell cycle were evaluated using CCK-8, wound-healing and transwell assay, colony formation, immunofluorescence, and flow cytometry and Western blotting, respectively. RNA sequencing was performed to determine the differential gene expression between controls and RIG-I knockdown. Tumor growth and radioresistance were assessed in nude mice using xenograft models. RIG-I expression was higher in ESCC tissues compared with that in matched non-tumor tissues. RIG-I overexpressing cells had a higher proliferation rate than RIG-I knockdown cells. Moreover, the knockdown of RIG-I slowed migration and invasion rates, whereas the overexpression of RIG-I accelerated migration and invasion rates. RIG-I overexpression induced radioresistance and G2/M phase arrest and reduced DNA damage after exposure to ionizing radiations compared with controls; however, it silenced the RIG-I enhanced radiosensitivity and DNA damage, and reduced the G2/M phase arrest. RNA sequencing revealed that the downstream genes DUSP6 and RIG-I had the same biological function; silencing DUSP6 can reduce the radioresistance caused by the overexpression of RIG-I. RIG-I knockdown depleted tumor growth in vivo, and radiation exposure effectively delayed the growth of xenograft tumors compared with the control group. RIG-I enhances the progression and radioresistance of ESCC; therefore, it may be a new potential target for ESCC-targeted therapy.
Collapse
Affiliation(s)
- Lu Li
- Department of Oncology Radiotherapy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230031, China; (L.L.)
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Lei Lv
- Department of Oncology Radiotherapy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230031, China; (L.L.)
| | - Jun-Chao Xu
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Qing He
- Department of Oncology Radiotherapy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230031, China; (L.L.)
| | - Na Chang
- Department of Oncology Radiotherapy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230031, China; (L.L.)
| | - Ya-Yun Cui
- Department of Oncology Radiotherapy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230031, China; (L.L.)
| | - Zhen-Chao Tao
- Department of Oncology Radiotherapy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230031, China; (L.L.)
| | - Tao Zhu
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
- Correspondence: (T.Z.); (L.-T.Q.)
| | - Li-Ting Qian
- Department of Oncology Radiotherapy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230031, China; (L.L.)
- Correspondence: (T.Z.); (L.-T.Q.)
| |
Collapse
|
17
|
Li S, Fan G, Li X, Cai Y, Liu R. Modulation of type I interferon signaling by natural products in the treatment of immune-related diseases. Chin J Nat Med 2023; 21:3-18. [PMID: 36641230 DOI: 10.1016/s1875-5364(23)60381-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Indexed: 01/15/2023]
Abstract
Type I interferon (IFN) is considered as a bridge between innate and adaptive immunity. Proper activation or inhibition of type I IFN signaling is essential for host defense against pathogen invasion, tumor cell proliferation, and overactive immune responses. Due to intricate and diverse chemical structures, natural products and their derivatives have become an invaluable source inspiring innovative drug discovery. In addition, some natural products have been applied in clinical practice for infection, cancer, and autoimmunity over thousands of years and their promising curative effects and safety have been well-accepted. However, whether these natural products are primarily targeting type I IFN signaling and specific molecular targets involved are not fully elucidated. In the current review, we thoroughly summarize recent advances in the pharmacology researches of natural products for their type I IFN activity, including both agonism/activation and antagonism/inhibition, and their potential application as therapies. Furthermore, the source and chemical nature of natural products with type I IFN activity are highlighted and their specific molecular targets in the type I IFN pathway and mode of action are classified. In conclusion, natural products possessing type I IFN activity represent promising therapeutic strategies and have a bright prospect in the treatment of infection, cancer, and autoimmune diseases.
Collapse
Affiliation(s)
- Shuo Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Guifang Fan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiaojiaoyang Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yajie Cai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Runping Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
18
|
Hufbauer M, Rattay S, Hagen C, Quaas A, Pfister H, Hartmann G, Coch C, Akgül B. Poly(I:C) Treatment Prevents Skin Tumor Formation in the Preclinical HPV8 Transgenic Mouse Model. J Invest Dermatol 2022:S0022-202X(22)02900-1. [PMID: 36584911 DOI: 10.1016/j.jid.2022.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/21/2022] [Accepted: 12/09/2022] [Indexed: 12/28/2022]
Abstract
Actinic keratoses and cutaneous squamous cell carcinomas are associated with infections with human papillomavirus of genus beta (betaHPV) in immunosuppressed patients. To date, targeted therapy against betaHPV-associated skin cancer does not exist because of the large number of betaHPV without defined high-risk types. In this study, we hypothesized that the activation of innate antiviral immunity in the skin, asymptomatically infected with betaHPV, induces an antitumor response by in situ autovaccination and prevents the formation of betaHPV-associated skin cancer. To test this, we used the preclinical keratin-14-HPV8 transgenic mouse model, which develops skin tumors after mechanical wounding. Remarkably, treatment with the antiviral immune response activating polyinosinic-polycytidylic acid (poly[I:C]) completely prevented cutaneous tumor growth. The induction of the IFN-induced genes Cxcl10 and Ifit1 by poly(I:C) depended on MDA5 activation. Increased numbers of total and activated CD4 and CD8 T cells were detected in poly(I:C)-treated skin. T cells were found in the skin of poly(I:C)-treated mice but not in the skin tumors of untreated mice. T-cell depletion showed a predominant role of CD4 T cells in poly(I:C)-mediated tumor prevention. Our findings identify the MDA5 ligand poly(I:C) as a promising candidate for in situ autovaccination approaches, which might serve as a treatment strategy against betaHPV-related skin diseases.
Collapse
Affiliation(s)
- Martin Hufbauer
- Institute of Virology, University of Cologne, Medical Faculty and University Hospital Cologne, Cologne, Germany
| | - Stephanie Rattay
- Institute of Virology, University of Cologne, Medical Faculty and University Hospital Cologne, Cologne, Germany
| | - Christian Hagen
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital, University of Bonn, Bonn, Germany
| | - Alexander Quaas
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Herbert Pfister
- Institute of Virology, University of Cologne, Medical Faculty and University Hospital Cologne, Cologne, Germany
| | - Gunther Hartmann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital, University of Bonn, Bonn, Germany
| | - Christoph Coch
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital, University of Bonn, Bonn, Germany; nextevidence GmbH, Munich, Germany
| | - Baki Akgül
- Institute of Virology, University of Cologne, Medical Faculty and University Hospital Cologne, Cologne, Germany.
| |
Collapse
|
19
|
Gu Z, Yu C. Harnessing bioactive nanomaterials in modulating tumor glycolysis-associated metabolism. J Nanobiotechnology 2022; 20:528. [PMID: 36510194 PMCID: PMC9746179 DOI: 10.1186/s12951-022-01740-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Glycolytic reprogramming is emerging as a hallmark of various cancers and a promising therapeutic target. Nanotechnology is revolutionizing the anti-tumor therapeutic approaches associated with glycolysis. Finely controlled chemical composition and nanostructure provide nanomaterials unique advantages, enabling an excellent platform for integrated drug delivery, biochemical modulation and combination therapy. Recent studies have shown promising potential of nanotherapeutic strategies in modulating tumor glycolytic metabolism alone or in combination with other treatments such as chemotherapy, radiotherapy and immunotherapy. To foster more innovation in this cutting-edge and interdisciplinary field, this review summarizes recent understandings of the origin and development of tumor glycolysis, then provides the latest advances in how nanomaterials modulate tumor glycolysis-related metabolism. The interplay of nanochemistry, metabolism and immunity is highlighted. Ultimately, the challenges and opportunities are presented.
Collapse
Affiliation(s)
- Zhengying Gu
- grid.22069.3f0000 0004 0369 6365School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241 People’s Republic of China
| | - Chengzhong Yu
- grid.22069.3f0000 0004 0369 6365School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241 People’s Republic of China ,grid.1003.20000 0000 9320 7537Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072 Australia
| |
Collapse
|
20
|
Liu J, Ji Q, Cheng F, Chen D, Geng T, Huang Y, Zhang J, He Y, Song T. The lncRNAs involved in regulating the RIG-I signaling pathway. Front Cell Infect Microbiol 2022; 12:1041682. [PMID: 36439216 PMCID: PMC9682092 DOI: 10.3389/fcimb.2022.1041682] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/21/2022] [Indexed: 09/23/2023] Open
Abstract
Understanding the targets and interactions of long non-coding RNAs (lncRNAs) related to the retinoic acid-inducible gene-I (RIG-I) signaling pathway is essential for developing interventions, which would enable directing the host inflammatory response regulation toward protective immunity. In the RIG-I signaling pathway, lncRNAs are involved in the important processes of ubiquitination, phosphorylation, and glycolysis, thus promoting the transport of the interferon regulatory factors 3 and 7 (IRF3 and IRF7) and the nuclear factor kappa B (NF-κB) into the nucleus, and activating recruitment of type I interferons (IFN-I) and inflammatory factors to the antiviral action site. In addition, the RIG-I signaling pathway has recently been reported to contain the targets of coronavirus disease-19 (COVID-19)-related lncRNAs. The molecules in the RIG-I signaling pathway are directly regulated by the lncRNA-microRNAs (miRNAs)-messenger RNA (mRNA) axis. Therefore, targeting this axis has become a novel strategy for the diagnosis and treatment of cancer. In this paper, the studies on the regulation of the RIG-I signaling pathway by lncRNAs during viral infections and cancer are comprehensively analyzed. The aim is to provide a solid foundation of information for conducting further detailed studies on lncRNAs and RIG-I in the future and also contribute to clinical drug development.
Collapse
Affiliation(s)
- Jing Liu
- Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Qinglu Ji
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Feng Cheng
- Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Dengwang Chen
- Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Tingting Geng
- Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Yueyue Huang
- Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Jidong Zhang
- Department of Immunology, Zunyi Medical University, Zunyi, China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, China
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Yuqi He
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Tao Song
- Department of Immunology, Zunyi Medical University, Zunyi, China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, China
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi, China
| |
Collapse
|
21
|
Wu CG, Casanova R, Mairinger F, Soltermann A. Lung adenocarcinoma patients with malignant pleural effusions in hot adaptive immunity status have a longer overall survival. Front Oncol 2022; 12:1031094. [PMID: 36267973 PMCID: PMC9577289 DOI: 10.3389/fonc.2022.1031094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Malignant pleural effusion (MPE) is a common complication of lung adenocarcinoma (LADC) which is associated with a dismal prognosis. We investigated the prognostic role of PD-L1 and other immunomodulators expression in the immune compartment of MPE immune composition. MPE cytologic cell blocks of 83 LADC patients were analysed for the mRNA expression of 770 cancer-immune genes by the NanoString nCounter platform. The expression of relevant immune cell lineage markers was validated by immunohistochemistry (IHC) using quantitative pathology. The mRNA immune profiling identified four MPE patient clusters (C). C1/2 (adaptive+, hot) showed better overall survival (OS) than C3/4 (adaptive-, cold). Additionally, cold immunity profiles (adaptive-), C4 (innate+) were associated with worse OS than C3 (innate-). High PD-L1 expression was linked to the regulation of T cell activation and interferon signalling pathways. Genes of pattern recognition receptor and type I interferon signalling pathways were specifically upregulated in the long-survival (≥90 days) patient group. Moreover, immunomodulators were co-activated and highly expressed in hot adaptive immunity patient clusters, whereas CD274 (PD-L1), TNFRSF9 (4-1BB), VEGFA (VEGF-A) and CD276 (B7-H3) were upregulated in the groups referred as cold. The patient cluster, age and PD-L1 expression were independent prognosticators for LADC MPE patients (p-value < 0.05). Our study sheds light on the variances of immune contexture regarding different PD-L1 expression and survival conditions. It revealed four distinct prognostic patient clusters with specific immune cell components and immunomodulator expression profiles, which, collectively, is supportive for future therapeutic and prognosis for cancer management.
Collapse
Affiliation(s)
- Cheng-Guang Wu
- Institute of Pathology, University Hospital Zurich, Zurich, Switzerland
- *Correspondence: Cheng-Guang Wu, ; Alex Soltermann,
| | - Ruben Casanova
- Institute of Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Fabian Mairinger
- Institute of Pathology, University Hospital Essen, Essen, Germany
| | - Alex Soltermann
- Facharzt Foederatio Medicorum Helveticorum (FMH) Pathologie, Pathologie Länggasse, Ittigen, Switzerland
- *Correspondence: Cheng-Guang Wu, ; Alex Soltermann,
| |
Collapse
|
22
|
Solstad A, Hogaboam O, Forero A, Hemann EA. RIG-I-like Receptor Regulation of Immune Cell Function and Therapeutic Implications. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:845-854. [PMID: 36130131 PMCID: PMC9512390 DOI: 10.4049/jimmunol.2200395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 06/30/2022] [Indexed: 01/04/2023]
Abstract
Retinoic acid-inducible gene I-like receptors (RLRs) are cytosolic RNA sensors critical for initiation of antiviral immunity. Activation of RLRs following RNA recognition leads to production of antiviral genes and IFNs for induction of broad antiviral immunity. Although the RLRs are ubiquitously expressed, much of our understanding of these molecules comes from their study in epithelial cells and fibroblasts. However, RLR activation is critical for induction of immune function and long-term protective immunity. Recent work has focused on the roles of RLRs in immune cells and their contribution to programming of effective immune responses. This new understanding of RLR function in immune cells and immune programming has led to the development of vaccines and therapeutics targeting the RLRs. This review covers recent advances in our understanding of the contribution of RLRs to immune cell function during infection and the emerging RLR-targeting strategies for induction of immunity against cancer and viral infection.
Collapse
Affiliation(s)
- Abigail Solstad
- Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, OH
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH
| | - Octavia Hogaboam
- Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, OH
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH
| | - Adriana Forero
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH
- Infectious Diseases Institute, The Ohio State University, Columbus, OH; and
| | - Emily A Hemann
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH;
- Infectious Diseases Institute, The Ohio State University, Columbus, OH; and
| |
Collapse
|
23
|
Ong HH, Andiappan AK, Duan K, Lum J, Liu J, Tan KS, Howland S, Lee B, Ong YK, Thong M, Chow VT, Wang DY. Transcriptomics of rhinovirus persistence reveals sustained expression of RIG-I and interferon-stimulated genes in nasal epithelial cells in vitro. Allergy 2022; 77:2778-2793. [PMID: 35274302 DOI: 10.1111/all.15280] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 02/19/2022] [Accepted: 02/21/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND Human rhinoviruses (HRVs) are frequently associated with asthma exacerbations, and have been found in the airways of asthmatic patients. While HRV-induced acute infection is well-documented, it is less clear whether the nasal epithelium sustains prolonged HRV infections along with the associated activation of host immune responses. OBJECTIVE To investigate sustainably regulated host responses of human nasal epithelial cells (hNECs) during HRV persistence. METHODS Using a time-course study, HRV16 persistence and viral replication dynamics were established using an in vitro infection model of hNECs. RNA sequencing was performed on hNECs in the early and late stages of infection at 3 and 14 days post-infection (dpi), respectively. The functional enrichment of differentially expressed genes (DEGs) was evaluated using gene ontology (GO) and Ingenuity pathway analysis. RESULTS HRV RNA and protein expression persisted throughout prolonged infections, even after decreased production of infectious virus progeny. GO analysis of unique DEGs indicated altered regulation of pathways related to ciliary function and airway remodeling at 3 dpi and serine-type endopeptidase activity at 14 dpi. The functional enrichment of shared DEGs between the two time-points was related to interferon (IFN) and cytoplasmic pattern recognition receptor (PRR) signaling pathways. Validation of the sustained regulation of candidate genes confirmed the persistent expression of RIG-I and revealed its close co-regulation with interferon-stimulated genes (ISGs) during HRV persistence. CONCLUSIONS The persistence of HRV RNA does not necessarily indicate an active infection during prolonged infection. The sustained expression of RIG-I and ISGs in response to viral RNA persistence highlights the importance of assessing how immune-activating host factors can change during active HRV infection and the immune regulation that persists thereafter.
Collapse
Affiliation(s)
- Hsiao Hui Ong
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Anand Kumar Andiappan
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Kaibo Duan
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Josephine Lum
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jing Liu
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kai Sen Tan
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Biosafety level 3 Core Facility, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore, Singapore
| | - Shanshan Howland
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Bernett Lee
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yew Kwang Ong
- Department of Otolaryngology - Head & Neck Surgery, National University Health System, Singapore, Singapore
| | - Mark Thong
- Department of Otolaryngology - Head & Neck Surgery, National University Health System, Singapore, Singapore
| | - Vincent T Chow
- Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - De-Yun Wang
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
24
|
Song J, Li M, Li C, Liu K, Zhu Y, Zhang H. Friend or foe: RIG- I like receptors and diseases. Autoimmun Rev 2022; 21:103161. [PMID: 35926770 PMCID: PMC9343065 DOI: 10.1016/j.autrev.2022.103161] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 07/29/2022] [Indexed: 12/22/2022]
Abstract
Retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs), which are pivotal sensors of RNA virus invasions, mediate the transcriptional induction of genes encoding type I interferons (IFNs) and proinflammatory cytokines, successfully establishing host antiviral immune response. A few excellent reviews have elaborated on the structural biology of RLRs and the antiviral mechanisms of RLR activation. In this review, we give a basic understanding of RLR biology and summarize recent findings of how RLR signaling cascade is strictly controlled by host regulatory mechanisms, which include RLR-interacting proteins, post-translational modifications and microRNAs (miRNAs). Furthermore, we pay particular attention to the relationship between RLRs and diseases, especially how RLRs participate in SARS-CoV-2, malaria or bacterial infections, how single-nucleotide polymorphisms (SNPs) or mutations in RLRs and antibodies against RLRs lead to autoinflammatory diseases and autoimmune diseases, and how RLRs are involved in anti-tumor immunity. These findings will provide insights and guidance for antiviral and immunomodulatory therapies targeting RLRs.
Collapse
Affiliation(s)
- Jie Song
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha City, Hunan Province, China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha City, Hunan Province, China
| | - Muyuan Li
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha City, Hunan Province, China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha City, Hunan Province, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha City, Hunan Province, China
| | - Caiyan Li
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha City, Hunan Province, China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha City, Hunan Province, China
| | - Ke Liu
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha City, Hunan Province, China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha City, Hunan Province, China
| | - Yaxi Zhu
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha City, Hunan Province, China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha City, Hunan Province, China.
| | - Huali Zhang
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha City, Hunan Province, China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha City, Hunan Province, China.
| |
Collapse
|
25
|
Xie R, Xie M, Zhu L, Chiu JWY, Lam W, Yap DYH. The Relationship of Pyroptosis-Related Genes, Patient Outcomes, and Tumor-Infiltrating Cells in Bladder Urothelial Carcinoma (BLCA). Front Pharmacol 2022; 13:930951. [PMID: 35928267 PMCID: PMC9343957 DOI: 10.3389/fphar.2022.930951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/16/2022] [Indexed: 11/17/2022] Open
Abstract
Introduction: The role of pyroptosis and its effects on tumor-infiltrating cells (TICs) in the pathogenesis and treatment outcomes of patients with bladder urothelial carcinoma (BLCA) remains unclear. Methods: We conducted a bioinformatics analysis on the pyroptosis-related genes (PRGs) and TICs using data from public domains, and evaluated their impact on the pathogenesis and clinical outcomes of BLCA patients. A risk score based on PRGs and a prognostic risk model that incorporated patient demographics, tumor characteristics, and differentially expressed genes (DEGs) were developed. Results: Twenty-three DEGs of 52 PRGs were identified in BLCA and normal samples from the TCGA database. Missense mutations and single nucleotide polymorphisms in PRGs are the most common genetic abnormalities. Patients with high PRG risk scores showed an inferior survival compared to those with low risk scores. The prognostic risk model based on patient demographics, tumor characteristics, and DEGs showed good predictive values for patient survival at 1, 3, and 5 years in BLCA patients. Caspase-8 (CASP8) was the only intersection gene of the prognostic genes, DEGs, and different genes expressed in tissue. Patients with a high CASP8 expression had improved survival, and an increased CASP8 expression level was observed in activated CD4 memory T cells, follicular T helper cells, resting NK cells, M0 macrophages, and activated dendritic cells. CASP8 expression also showed a positive correlation with the IL7R expression—a key cell marker of CD4 memory T cells. CASP8 expression also showed correlations with immune checkpoints (PDCD1, CD274, and CTLA4) and response to immune checkpoint inhibitors. Conclusion: Our data suggest that PRGs, especially CASP8, showed strong associations with patient outcomes and TICs in BLCA. If validated, these results could potentially aid in the prognostication and guide treatment in BLCA patients.
Collapse
Affiliation(s)
- Ruiyan Xie
- Division of Nephrology, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Ming Xie
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Litong Zhu
- Division of Nephrology, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Joanne W. Y. Chiu
- Division of Haematology, Medical Oncology and Haemopoietic Stem Cell Transplantation, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Wayne Lam
- Division of Urology, Department of Surgery, Queen Mary Hospital, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Desmond Y. H. Yap
- Division of Nephrology, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- *Correspondence: Desmond Y. H. Yap,
| |
Collapse
|
26
|
Li Z, Mei Z, Ding S, Chen L, Li H, Feng K, Huang T, Cai YD. Identifying Methylation Signatures and Rules for COVID-19 With Machine Learning Methods. Front Mol Biosci 2022; 9:908080. [PMID: 35620480 PMCID: PMC9127386 DOI: 10.3389/fmolb.2022.908080] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 04/27/2022] [Indexed: 11/13/2022] Open
Abstract
The occurrence of coronavirus disease 2019 (COVID-19) has become a serious challenge to global public health. Definitive and effective treatments for COVID-19 are still lacking, and targeted antiviral drugs are not available. In addition, viruses can regulate host innate immunity and antiviral processes through the epigenome to promote viral self-replication and disease progression. In this study, we first analyzed the methylation dataset of COVID-19 using the Monte Carlo feature selection method to obtain a feature list. This feature list was subjected to the incremental feature selection method combined with a decision tree algorithm to extract key biomarkers, build effective classification models and classification rules that can remarkably distinguish patients with or without COVID-19. EPSTI1, NACAP1, SHROOM3, C19ORF35, and MX1 as the essential features play important roles in the infection and immune response to novel coronavirus. The six significant rules extracted from the optimal classifier quantitatively explained the expression pattern of COVID-19. Therefore, these findings validated that our method can distinguish COVID-19 at the methylation level and provide guidance for the diagnosis and treatment of COVID-19.
Collapse
Affiliation(s)
- Zhandong Li
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, China
| | - Zi Mei
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Shijian Ding
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai, China
| | - Hao Li
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, China
| | - Kaiyan Feng
- Department of Computer Science, Guangdong AIB Polytechnic College, Guangzhou, China
| | - Tao Huang
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- *Correspondence: Tao Huang, ; Yu-Dong Cai,
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai, China
- *Correspondence: Tao Huang, ; Yu-Dong Cai,
| |
Collapse
|
27
|
Bianconi A, Aruta G, Rizzo F, Salvati LF, Zeppa P, Garbossa D, Cofano F. Systematic Review on Tumor Microenvironment in Glial Neoplasm: From Understanding Pathogenesis to Future Therapeutic Perspectives. Int J Mol Sci 2022; 23:4166. [PMID: 35456984 PMCID: PMC9029619 DOI: 10.3390/ijms23084166] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/04/2022] [Accepted: 04/06/2022] [Indexed: 02/04/2023] Open
Abstract
Despite the multidisciplinary management in the treatment of glioblastomas, the average survival of GBM patients is still 15 months. In recent years, molecular biomarkers have gained more and more importance both in the diagnosis and therapy of glial tumors. At the same time, it has become clear that non neoplastic cells, which constitute about 30% of glioma mass, dramatically influence tumor growth, spread, and recurrence. This is the main reason why, in recent years, scientific research has been focused on understanding the function and the composition of tumor microenvironment and its role in gliomagenesis and recurrence. The aim of this review is to summarize the most recent discovery about resident microglia, tumor-associated macrophages, lymphocytes, and the role of extracellular vesicles and their bijective interaction with glioma cells. Moreover, we reported the most recent updates about new therapeutic strategies targeting immune system receptors and soluble factors. Understanding how glioma cells interact with non-neoplastic cells in tumor microenvironment is an essential step to comprehend mechanisms at the base of disease progression and to find new therapeutic strategies for GBM patients. However, no significant results have yet been obtained in studies targeting single molecules/pathways; considering the complex microenvironment, it is likely that only by using multiple therapeutic agents acting on multiple molecular targets can significant results be achieved.
Collapse
Affiliation(s)
- Andrea Bianconi
- Department of Neuroscience “Rita Levi Montalcini”, University of Turin, 10126 Turin, Italy; (G.A.); (F.R.); (P.Z.); (D.G.); (F.C.)
| | - Gelsomina Aruta
- Department of Neuroscience “Rita Levi Montalcini”, University of Turin, 10126 Turin, Italy; (G.A.); (F.R.); (P.Z.); (D.G.); (F.C.)
| | - Francesca Rizzo
- Department of Neuroscience “Rita Levi Montalcini”, University of Turin, 10126 Turin, Italy; (G.A.); (F.R.); (P.Z.); (D.G.); (F.C.)
| | | | - Pietro Zeppa
- Department of Neuroscience “Rita Levi Montalcini”, University of Turin, 10126 Turin, Italy; (G.A.); (F.R.); (P.Z.); (D.G.); (F.C.)
| | - Diego Garbossa
- Department of Neuroscience “Rita Levi Montalcini”, University of Turin, 10126 Turin, Italy; (G.A.); (F.R.); (P.Z.); (D.G.); (F.C.)
| | - Fabio Cofano
- Department of Neuroscience “Rita Levi Montalcini”, University of Turin, 10126 Turin, Italy; (G.A.); (F.R.); (P.Z.); (D.G.); (F.C.)
- Spine Surgery Unit, Humanitas Gradeningo, 10100 Turin, Italy
| |
Collapse
|
28
|
Expressed prognostic biomarkers for primary prostate cancer independent of multifocality and transcriptome heterogeneity. Cancer Gene Ther 2022; 29:1276-1284. [PMID: 35194199 DOI: 10.1038/s41417-022-00444-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/28/2022] [Accepted: 02/08/2022] [Indexed: 12/17/2022]
Abstract
The majority of prostate cancer patients are diagnosed with multiple primary malignant foci. The distinct foci are exceptionally heterogeneous with regard to DNA mutations, but whether this is recapitulated at the transcriptome level remains unknown. In this study, inter- and intrafocal heterogeneity has been assessed by whole-transcriptome sequencing of 87 tissue samples from 23 patients with localized prostate cancer treated with radical prostatectomy. From each patient, multiple samples were taken from one or more malignant foci, in addition to one sample from benign prostate tissue. Transcriptomic profiles of different malignant foci from the same patient showed a similar level of heterogeneity as tumors from different patients. This applies to expression of genes, fusion genes, and somatic mutations. Within-patient pair-wise analyses identified expression patterns linked to ETS status and extraprostatic extension. A set of 62 genes were found with low intrapatient heterogeneity and high interpatient heterogeneity, retaining stable expression profiles across foci within the same patient. Among these, 16 genes are associated with biochemical recurrence in a separately published study and are therefore nominated as biomarkers with prognostic value regardless of which malignant focus is sampled. In conclusion, an extensive heterogeneity in multifocal prostate cancer is confirmed at the gene expression level. Diagnostic biomarkers were identified for ETS positive samples and samples from extraprostatic extensions. Finally, prognostic biomarkers independent of multifocal heterogeneity were found.
Collapse
|
29
|
Bacterial Translocation in Gastrointestinal Cancers and Cancer Treatment. Biomedicines 2022; 10:biomedicines10020380. [PMID: 35203589 PMCID: PMC8962358 DOI: 10.3390/biomedicines10020380] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 11/16/2022] Open
Abstract
In recent years, there has been increasing evidence that gut microbiota is associated with the onset and exacerbation of various diseases, such as gastrointestinal cancer. For instance, it is well known that local inflammation of the intestinal tract in colorectal cancer that is caused by the increased number of Fusobacterium, due to changes in the intestinal bacterial flora, is involved in carcinogenesis. In contrast, gut bacteria or their products, pathogen-associated molecular patterns, not only cause intestinal inflammation but also invade the bloodstream through dysbiosis and gut barrier dysfunction, thereby leading to systemic inflammation, namely bacterial translocation. The involvement of bacterial translocation in the carcinogenesis of gastrointestinal cancers and their prognosis is increasingly being recognized. The Toll-like receptor signaling pathways plays an important role in the carcinogenesis of such cancers. In addition, bacterial translocation influences the treatment of cancers such as surgery and chemotherapy. In this review, we outline the concept of bacterial translocation, summarize the current knowledge on the relationship between gut bacteria and gastrointestinal cancer, and provide future perspectives of this field.
Collapse
|
30
|
Sales Conniff A, Encalada G, Patel S, Bhandary M, Al-Takrouri F, Heller L. Poly(I:C) transfection induces a pro-inflammatory cascade in murine mammary carcinoma and fibrosarcoma cells. RNA Biol 2022; 19:841-851. [PMID: 35737804 PMCID: PMC9235898 DOI: 10.1080/15476286.2022.2084861] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Germline-encoded pattern recognition receptors [PRRs] in mammalian cells function in the detection of molecular patterns associated with pathogen invasion or cellular damage. A PRR subset is activated by the atypical presence and location of double-stranded RNA [dsRNA] or its synthetic analogue polyinosinic-polycytidylic acid [poly(I:C)], triggering pro-inflammatory signalling and death in many cell types. Poly(I:C) has been tested as a sole or combination cancer therapy in preclinical studies and clinical trials. The purpose of this study was to evaluate the effects of poly(I:C) transfection via electroporation on cell lines from a cancer of epithelial origin, 4T1 mammary carcinoma, and a cancer of mesenchymal origin, WEHI 164 fibrosarcoma. The effects of the poly(I:C) delivery on cell metabolism implicate the induction of cell death. A pro-inflammatory response was demonstrated by mRNA upregulation and the secretion of Type I interferon and several cytokines and chemokines. The mRNAs of dsRNA sensor DExD/H-box helicase 58/retinoic acid-inducible gene I protein [Ddx58/RIG-I] and sensor/co-sensor DEAH-box helicase 9 [Dhx9] were not regulated, but the mRNAs of RNA sensors toll-like receptor 3 [TLR3], interferon-induced with helicase C domain 1/melanoma differentiation-associated protein 5 [Ifih1/MDA5] and Z-DNA binding protein 1 [Zbp1] and co-sensors DEAD (Asp-Glu-Ala-Asp) box polypeptide 60 [Ddx60] and interferon-inducible protein 204 [Ifi204] were upregulated in both cell lines. The mRNAs encoding signalling pathways components were present or upregulated in both cell types. These data demonstrate that RNA sensing effects can be amplified by electroporation delivery, potentially expanding the practicality of this immunotherapeutic approach.
Collapse
Affiliation(s)
- A Sales Conniff
- Department of Medical Engineering, University of South Florida, Tampa, FL, USA
| | - G Encalada
- Department of Medical Engineering, University of South Florida, Tampa, FL, USA
| | - S Patel
- Department of Medical Engineering, University of South Florida, Tampa, FL, USA
| | - M Bhandary
- Department of Medical Engineering, University of South Florida, Tampa, FL, USA
| | - F Al-Takrouri
- Department of Medical Engineering, University of South Florida, Tampa, FL, USA
| | - L Heller
- Department of Medical Engineering, University of South Florida, Tampa, FL, USA
| |
Collapse
|
31
|
Ye Y, Xu C, Chen F, Liu Q, Cheng N. Targeting Innate Immunity in Breast Cancer Therapy: A Narrative Review. Front Immunol 2021; 12:771201. [PMID: 34899721 PMCID: PMC8656691 DOI: 10.3389/fimmu.2021.771201] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/08/2021] [Indexed: 01/07/2023] Open
Abstract
Although breast cancer has been previously considered "cold" tumors, numerous studies are currently conducted to explore the great potentials of immunotherapies in improving breast cancer patient outcomes. In addition to the focus on stimulating adaptive immunity for antitumor responses, growing evidence showed the importance of triggering host innate immunity to eradicate established tumors and/or control tumor metastasis of breast cancer. In this review, we first briefly introduce the breast tumor immune microenvironment. We also discuss innate immune targets and pathways and mechanisms of their synergy with the adaptive antitumor response and other treatment strategies. Lastly, we review clinical trials targeting innate immune pathways for breast cancer therapies.
Collapse
Affiliation(s)
- Yanqi Ye
- Zenomics. Inc. Magnify at California NanoSystems Institute, Los Angeles, CA, United States
| | - Chun Xu
- School of Dentistry, The University of Queensland, Brisbane, QLD, Australia
| | - Fengqian Chen
- School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Qi Liu
- School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Ning Cheng
- Department of Otolaryngology - Head and Neck Surgery, University of California at San Francisco, San Francisco, CA, United States
| |
Collapse
|
32
|
Development and Validation of Tumor Immunogenicity Based Gene Signature for Skin Cancer Risk Stratification. Int J Mol Sci 2021; 22:ijms222112025. [PMID: 34769455 PMCID: PMC8584987 DOI: 10.3390/ijms222112025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 12/13/2022] Open
Abstract
Melanoma is one of the most aggressive types of skin cancer, with significant heterogeneity in overall survival. Currently, tumor-node-metastasis (TNM) staging is insufficient to provide accurate survival prediction and appropriate treatment decision making for several types of tumors, such as those in melanoma patients. Therefore, the identification of more reliable prognosis biomarkers is urgently essential. Recent studies have shown that low immune cells infiltration is significantly associated with unfavorable clinical outcome in melanoma patients. Here we constructed a prognostic-related gene signature for melanoma risk stratification by quantifying the levels of several cancer hallmarks and identify the Wnt/β-catenin activation pathway as a primary risk factor for low tumor immunity. A series of bioinformatics and statistical methods were combined and applied to construct a Wnt-immune-related prognosis gene signature. With this gene signature, we computed risk scores for individual patients that can predict overall survival. To evaluate the robustness of the result, we validated the signature in multiple independent GEO datasets. Finally, an overall survival-related nomogram was established based on the gene signature and clinicopathological features. The Wnt-immune-related prognostic risk score could better predict overall survival compared with standard clinicopathological features. Our results provide a comprehensive map of the oncogene-immune-related gene signature that can serve as valuable biomarkers for better clinical decision making.
Collapse
|
33
|
Vedagiri D, Gupta D, Mishra A, Krishna G, Bhaskar M, Sah V, Basu A, Nayak D, Kalia M, Valiya Veettil M, Harshan KH. Retinoic Acid-Inducible Gene I-Like Receptors Activate Snail To Limit RNA Viral Infections. J Virol 2021; 95:e0121621. [PMID: 34379517 PMCID: PMC8513471 DOI: 10.1128/jvi.01216-21] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 08/06/2021] [Indexed: 11/20/2022] Open
Abstract
Retinoic acid-inducible gene I-like receptors (RLRs) are important cytosolic pattern recognition receptors (PRRs) that sense viral RNA before mounting a response leading to the activation of type I IFNs. Several viral infections induce epithelial-mesenchymal transition (EMT), even as its significance remains unclear. Here, we show that EMT or an EMT-like process is a general response to viral infections. Our studies identify a previously unknown mechanism of regulation of an important EMT-transcription factor (EMT-TF) Snail during RNA viral infections and describe its possible implication. RNA viral infections, poly(I·C) transfection, and ectopic expression of RLR components induced Snail levels, indicating that RLR pathway could regulate its expression. Detailed examination using mitochondrial antiviral signaling protein knockout (MAVS-KO) cells established that MAVS is essential in this regulation. We identified two interferon-stimulated response elements (ISREs) in the SNAI1 promoter region and demonstrated that they are important in its transcriptional activation by phosphorylated IRF3. Increasing the levels of Snail activated RLR pathway and dramatically limited replication of the RNA viruses dengue virus, Japanese encephalitis virus (JEV), and vesicular stomatitis virus, pointing to their antiviral functions. Knockdown of Snail resulted in a considerable increase in the JEV titer, validating its antiviral functions. Finally, transforming growth factor β-mediated IFNB activation was dependent on Snail levels, confirming its important role in type I IFN activation. Thus, EMT-TF Snail is transcriptionally coregulated with type I IFN by RLRs and, in turn, promotes the RLR pathway, further strengthening the antiviral state in the cell. Our work identified an interesting mechanism of regulation of Snail that demonstrates potential coregulation of multiple innate antiviral pathways triggered by RLRs. Identification of antiviral functions of Snail also provides an opportunity to expand the sphere of RLR signaling. IMPORTANCE RLRs sense viral genomic RNA or the double-stranded RNA intermediates and trigger the activation of type I IFNs. Snail transcription factor, commonly associated with epithelial-mesenchymal transition (EMT), has been reported to facilitate EMT in several viral infections. Many of these reports are based on oncoviruses, leading to the speculation that EMT induced during infection is an important factor in the oncogenesis triggered by these infections. However, our studies reveal that EMT or EMT-like processes during viral infections have important functions in antiviral response. We have characterized a new mechanism of transcriptional regulation of Snail by IRF3 through interferon-stimulated response elements in their promoters, and this finding could have importance in nonviral contexts as well. We also identify that EMT-TF Snail promotes antiviral status of the infected cells through the RLR pathway. This study characterizes a new regulatory mechanism of activation of Snail and establishes its unidentified function in antiviral response.
Collapse
Affiliation(s)
- Dhiviya Vedagiri
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
- Academy for Scientific and Innovative Research, Ghaziabad, India
| | - Divya Gupta
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Anurag Mishra
- Discipline of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, India
| | - Gayathri Krishna
- Virology Laboratory, Department of Biotechnology, Cochin University of Science and Technology, Cochin, Kerala, India
| | | | - Vishal Sah
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
- Academy for Scientific and Innovative Research, Ghaziabad, India
| | - Anirban Basu
- National Brain Research Centre, Manesar, Haryana, India
| | - Debasis Nayak
- Discipline of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, India
| | - Manjula Kalia
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Mohanan Valiya Veettil
- Virology Laboratory, Department of Biotechnology, Cochin University of Science and Technology, Cochin, Kerala, India
| | - Krishnan Harinivas Harshan
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
- Academy for Scientific and Innovative Research, Ghaziabad, India
| |
Collapse
|
34
|
Thoresen D, Wang W, Galls D, Guo R, Xu L, Pyle AM. The molecular mechanism of RIG-I activation and signaling. Immunol Rev 2021; 304:154-168. [PMID: 34514601 PMCID: PMC9293153 DOI: 10.1111/imr.13022] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 08/10/2021] [Accepted: 08/17/2021] [Indexed: 12/25/2022]
Abstract
RIG‐I is our first line of defense against RNA viruses, serving as a pattern recognition receptor that identifies molecular features common among dsRNA and ssRNA viral pathogens. RIG‐I is maintained in an inactive conformation as it samples the cellular space for pathogenic RNAs. Upon encounter with the triphosphorylated terminus of blunt‐ended viral RNA duplexes, the receptor changes conformation and releases a pair of signaling domains (CARDs) that are selectively modified and interact with an adapter protein (MAVS), thereby triggering a signaling cascade that stimulates transcription of interferons. Here, we describe the structural determinants for specific RIG‐I activation by viral RNA, and we describe the strategies by which RIG‐I remains inactivated in the presence of host RNAs. From the initial RNA triggering event to the final stages of interferon expression, we describe the experimental evidence underpinning our working knowledge of RIG‐I signaling. We draw parallels with behavior of related proteins MDA5 and LGP2, describing evolutionary implications of their collective surveillance of the cell. We conclude by describing the cell biology and immunological investigations that will be needed to accurately describe the role of RIG‐I in innate immunity and to provide the necessary foundation for pharmacological manipulation of this important receptor.
Collapse
Affiliation(s)
- Daniel Thoresen
- Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Wenshuai Wang
- Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Drew Galls
- Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Rong Guo
- Chemistry, Yale University, New Haven, CT, USA
| | - Ling Xu
- Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Anna Marie Pyle
- Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA.,Chemistry, Yale University, New Haven, CT, USA.,Howard Hughes Medical Institute, Yale University, New Haven, CT, USA
| |
Collapse
|
35
|
Musella M, Galassi C, Manduca N, Sistigu A. The Yin and Yang of Type I IFNs in Cancer Promotion and Immune Activation. BIOLOGY 2021; 10:856. [PMID: 34571733 PMCID: PMC8467547 DOI: 10.3390/biology10090856] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 12/22/2022]
Abstract
Type I Interferons (IFNs) are key regulators of natural and therapy-induced host defense against viral infection and cancer. Several years of remarkable progress in the field of oncoimmunology have revealed the dual nature of these cytokines. Hence, Type I IFNs may trigger anti-tumoral responses, while leading immune dysfunction and disease progression. This dichotomy relies on the duration and intensity of the transduced signaling, the nature of the unleashed IFN stimulated genes, and the subset of responding cells. Here, we discuss the role of Type I IFNs in the evolving relationship between the host immune system and cancer, as we offer a view of the therapeutic strategies that exploit and require an intact Type I IFN signaling, and the role of these cytokines in inducing adaptive resistance. A deep understanding of the complex, yet highly regulated, network of Type I IFN triggered molecular pathways will help find a timely and immune"logical" way to exploit these cytokines for anticancer therapy.
Collapse
Affiliation(s)
- Martina Musella
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (C.G.); (N.M.)
| | - Claudia Galassi
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (C.G.); (N.M.)
| | - Nicoletta Manduca
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (C.G.); (N.M.)
| | - Antonella Sistigu
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (C.G.); (N.M.)
- Tumor Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| |
Collapse
|
36
|
Wang Z, Zou W, Wang F, Zhang G, Chen K, Hu M, Liu R. Identification of the immune cell infiltration landscape in pancreatic cancer to assist immunotherapy. Future Oncol 2021; 17:4131-4143. [PMID: 34346253 DOI: 10.2217/fon-2021-0495] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background: A malignant tumor's immune environment, including infiltrating immune cell status, can be critical to patient outcomes. Recent studies have shown that immune cell infiltration (ICI) in pancreatic cancer (PC) is highly correlated with the response to immunotherapy and patient prognosis. Therefore, we aimed to create an ICI score that accurately predicts patient outcomes and immunotherapeutic efficacy. Methods: The ICI statuses of patients with PC were estimated from the publicly available The Cancer Genome Atlas (TCGA) pancreatic ductal adenocarcinoma and GSE57495 gene expression datasets using two computational algorithms (CIBERSORT and ESTIMATE). ICI and transcriptome subsets were defined using a clustering algorithm, and survival analysis was also performed. Principal component analysis was used to calculate the novel ICI score, and gene set enrichment analysis was performed to identify the pathways underlying the defined clusters. The tumor mutational burden (TMB) was further explored in TCGA cohort, and survival analysis was used to assess the capability of the ICI and TMB scores to predict overall survival. Additionally, common driver gene mutations and their differential expression in the different ICI score group were investigated. Results: The ICI landscapes of 240 patients were generated using the devised algorithm, revealing three ICI and three gene clusters whose use improved the prediction of overall survival (p = 0.019 and p < 0.001, respectively). Crucial immune checkpoint genes were differentially expressed among these subtypes; the RIG-I-LIKE and NOD-LIKE receptor signaling pathways were enriched in samples with low ICI scores (p < 0.05). We also found that the TMB scores could predict survival outcomes, whereas the ICI scores also could predict prognoses independent of TMB. Notably, ICI scores could effectively predict responses to immunotherapy. KRAS, TP53, CDKN2A, SMAD4 and TTN remained the most commonly mutated genes in PC; moreover, KRAS and TP53 mutation rates were significantly different between the two ICI score groups. Conclusions: We developed a novel ICI score that could independently predict the response to immunotherapy and survival of patients with PC. Evaluation of the ICI landscape in a larger cohort could clarify the interactions between these infiltrating cells, the tumor microenvironment and response to immunotherapy.
Collapse
Affiliation(s)
- Zizheng Wang
- Faculty of Hepato-Pancreato-Biliary Surgery, Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of Chinese PLA, & Key Laboratory of Digital Hepetobiliary Surgery, PLA, Beijing, China
| | - Wenbo Zou
- Faculty of Hepato-Pancreato-Biliary Surgery, Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of Chinese PLA, & Key Laboratory of Digital Hepetobiliary Surgery, PLA, Beijing, China.,Medical School of Chinese PLA, Beijing, China
| | - Fei Wang
- Faculty of Hepato-Pancreato-Biliary Surgery, Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of Chinese PLA, & Key Laboratory of Digital Hepetobiliary Surgery, PLA, Beijing, China
| | - Gong Zhang
- Faculty of Hepato-Pancreato-Biliary Surgery, Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of Chinese PLA, & Key Laboratory of Digital Hepetobiliary Surgery, PLA, Beijing, China.,Medical School of Chinese PLA, Beijing, China
| | - Kuang Chen
- Faculty of Hepato-Pancreato-Biliary Surgery, Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of Chinese PLA, & Key Laboratory of Digital Hepetobiliary Surgery, PLA, Beijing, China.,Medical School of Chinese PLA, Beijing, China
| | - Minggen Hu
- Faculty of Hepato-Pancreato-Biliary Surgery, Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of Chinese PLA, & Key Laboratory of Digital Hepetobiliary Surgery, PLA, Beijing, China.,Medical School of Chinese PLA, Beijing, China
| | - Rong Liu
- Faculty of Hepato-Pancreato-Biliary Surgery, Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of Chinese PLA, & Key Laboratory of Digital Hepetobiliary Surgery, PLA, Beijing, China.,Medical School of Chinese PLA, Beijing, China
| |
Collapse
|
37
|
Batool M, Kim MS, Choi S. Structural insights into the distinctive RNA recognition and therapeutic potentials of RIG-I-like receptors. Med Res Rev 2021; 42:399-425. [PMID: 34287999 DOI: 10.1002/med.21845] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 06/11/2021] [Accepted: 07/04/2021] [Indexed: 12/12/2022]
Abstract
RNA viruses, including the coronavirus, develop a unique strategy to evade the host immune response by interrupting the normal function of cytosolic retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs). RLRs rapidly detect atypical nucleic acids, thereby triggering the antiviral innate immune signaling cascade and subsequently activates the interferons transcription and induction of other proinflammatory cytokines and chemokines. Nonetheless, these receptors are manipulated by viral proteins to subvert the host immune system and sustain the infectivity and replication potential of the virus. RIG-I senses the single-stranded, double-stranded, and short double-stranded RNAs and recognizes the key signature, a 5'-triphosphate moiety, at the blunt end of the viral RNA. Meanwhile, the melanoma differentiation-associated gene 5 (MDA5) is triggered by longer double stranded RNAs, messenger RNAs lacking 2'-O-methylation in their 5'-cap, and RNA aggregates. Therefore, structural insights into the nucleic-acid-sensing and downstream signaling mechanisms of these receptors hold great promise for developing effective antiviral therapeutic interventions. This review highlights the critical roles played by RLRs in viral infections as well as their ligand recognition mechanisms. In addition, we highlight the crosstalk between the toll-like receptors and RLRs and provide a comprehensive overview of RLR-associated diseases as well as the therapeutic potential of RLRs for the development of antiviral-drugs. Moreover, we believe that these RLR-based antivirals will serve as a step toward countering the recent coronavirus disease 2019 pandemic.
Collapse
Affiliation(s)
- Maria Batool
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
- S&K Therapeutics, Campus Plaza 418, Ajou University, Suwon, Korea
| | - Moon Suk Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
- S&K Therapeutics, Campus Plaza 418, Ajou University, Suwon, Korea
| |
Collapse
|
38
|
de Sousa E, Lérias JR, Beltran A, Paraschoudi G, Condeço C, Kamiki J, António PA, Figueiredo N, Carvalho C, Castillo-Martin M, Wang Z, Ligeiro D, Rao M, Maeurer M. Targeting Neoepitopes to Treat Solid Malignancies: Immunosurgery. Front Immunol 2021; 12:592031. [PMID: 34335558 PMCID: PMC8320363 DOI: 10.3389/fimmu.2021.592031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 05/07/2021] [Indexed: 12/26/2022] Open
Abstract
Successful outcome of immune checkpoint blockade in patients with solid cancers is in part associated with a high tumor mutational burden (TMB) and the recognition of private neoantigens by T-cells. The quality and quantity of target recognition is determined by the repertoire of ‘neoepitope’-specific T-cell receptors (TCRs) in tumor-infiltrating lymphocytes (TIL), or peripheral T-cells. Interferon gamma (IFN-γ), produced by T-cells and other immune cells, is essential for controlling proliferation of transformed cells, induction of apoptosis and enhancing human leukocyte antigen (HLA) expression, thereby increasing immunogenicity of cancer cells. TCR αβ-dependent therapies should account for tumor heterogeneity and availability of the TCR repertoire capable of reacting to neoepitopes and functional HLA pathways. Immunogenic epitopes in the tumor-stroma may also be targeted to achieve tumor-containment by changing the immune-contexture in the tumor microenvironment (TME). Non protein-coding regions of the tumor-cell genome may also contain many aberrantly expressed, non-mutated tumor-associated antigens (TAAs) capable of eliciting productive anti-tumor immune responses. Whole-exome sequencing (WES) and/or RNA sequencing (RNA-Seq) of cancer tissue, combined with several layers of bioinformatic analysis is commonly used to predict possible neoepitopes present in clinical samples. At the ImmunoSurgery Unit of the Champalimaud Centre for the Unknown (CCU), a pipeline combining several tools is used for predicting private mutations from WES and RNA-Seq data followed by the construction of synthetic peptides tailored for immunological response assessment reflecting the patient’s tumor mutations, guided by MHC typing. Subsequent immunoassays allow the detection of differential IFN-γ production patterns associated with (intra-tumoral) spatiotemporal differences in TIL or peripheral T-cells versus TIL. These bioinformatics tools, in addition to histopathological assessment, immunological readouts from functional bioassays and deep T-cell ‘adaptome’ analyses, are expected to advance discovery and development of next-generation personalized precision medicine strategies to improve clinical outcomes in cancer in the context of i) anti-tumor vaccination strategies, ii) gauging mutation-reactive T-cell responses in biological therapies and iii) expansion of tumor-reactive T-cells for the cellular treatment of patients with cancer.
Collapse
Affiliation(s)
- Eric de Sousa
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Joana R Lérias
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Antonio Beltran
- Department of Pathology, Champalimaud Clinical Centre, Lisbon, Portugal
| | | | - Carolina Condeço
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Jéssica Kamiki
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | | | - Nuno Figueiredo
- Digestive Unit, Champalimaud Clinical Centre, Lisbon, Portugal
| | - Carlos Carvalho
- Digestive Unit, Champalimaud Clinical Centre, Lisbon, Portugal
| | | | - Zhe Wang
- Jiangsu Industrial Technology Research Institute (JITRI), Applied Adaptome Immunology Institute, Nanjing, China
| | - Dário Ligeiro
- Lisbon Centre for Blood and Transplantation, Instituto Português do Sangue e Transplantação (IPST), Lisbon, Portugal
| | - Martin Rao
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Markus Maeurer
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal.,I Medical Clinic, Johannes Gutenberg University of Mainz, Mainz, Germany
| |
Collapse
|
39
|
Bufalieri F, Basili I, Di Marcotullio L, Infante P. Harnessing the Activation of RIG-I Like Receptors to Inhibit Glioblastoma Tumorigenesis. Front Mol Neurosci 2021; 14:710171. [PMID: 34305530 PMCID: PMC8295747 DOI: 10.3389/fnmol.2021.710171] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 06/16/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GB) is an incurable form of brain malignancy in an adult with a median survival of less than 15 months. The current standard of care, which consists of surgical resection, radiotherapy, and chemotherapy with temozolomide, has been unsuccessful due to an extensive inter- and intra-tumoral genetic and molecular heterogeneity. This aspect represents a serious obstacle for developing alternative therapeutic options for GB. In the last years, immunotherapy has emerged as an effective treatment for a wide range of cancers and several trials have evaluated its effects in GB patients. Unfortunately, clinical outcomes were disappointing particularly because of the presence of tumor immunosuppressive microenvironment. Recently, anti-cancer approaches aimed to improve the expression and the activity of RIG-I-like receptors (RLRs) have emerged. These innovative therapeutic strategies attempt to stimulate both innate and adaptive immune responses against tumor antigens and to promote the apoptosis of cancer cells. Indeed, RLRs are important mediators of the innate immune system by triggering the type I interferon (IFN) response upon recognition of immunostimulatory RNAs. In this mini-review, we discuss the functions of RLRs family members in the control of immune response and we focus on the potential clinical application of RLRs agonists as a promising strategy for GB therapy.
Collapse
Affiliation(s)
| | - Irene Basili
- Department of Molecular Medicine, University La Sapienza, Rome, Italy
| | - Lucia Di Marcotullio
- Department of Molecular Medicine, University La Sapienza, Rome, Italy
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Paola Infante
- Center For Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| |
Collapse
|
40
|
Yarza R, Bover M, Agulló-Ortuño MT, Iglesias-Docampo LC. Current approach and novel perspectives in nasopharyngeal carcinoma: the role of targeting proteasome dysregulation as a molecular landmark in nasopharyngeal cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:202. [PMID: 34154654 PMCID: PMC8215824 DOI: 10.1186/s13046-021-02010-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 06/07/2021] [Indexed: 12/15/2022]
Abstract
Nasopharyngeal carcinoma (NPC) represents a molecularly paradigmatic tumor given the complex diversity of environmental as well as host dependent factors that are closely implicated in tissue transformation and carcinogenesis. Epstein Barr Virus (EBV) plays a key role in tissue invasion, hyperplasia and malignant transformation. Therefore, EBV related oncoviral proteins such as Latent Membrane Protein family (LMP1, LMP2), Epstein Barr Nuclear Antigen 1 (EBNA1) and EBV related glycoprotein B (gB) are responsible for inducing intracellular signalling aberrations leading to sustained proliferation and further acquisition of NPC related invasive nature and metastatic potential.Dysregulation of proteasome signaling seems to be centrally implicated in oncoviral protein stabilization as well as in modulating tumor microenvironment. Different studies in vitro and in vivo suggest a potential role of proteasome inhibitors in the therapeutic setting of NPC. Furthermore, alterations affecting proteasome signalling in NPC have been associated to tumor growth and invasion, distant metastasis, immune exclusion and resistance as well as to clinical poor prognosis. So on, recent studies have shown the efficacy of immunotherapy as a suitable therapeutic approach to NPC. Nevertheless, novel strategies seem to look for combinatorial regimens aiming to potentiate immune recognition as well as to restore both primary and acquired immune resistance.In this work, our goal is to thoroughly review the molecular implications of proteasome dysregulation in the molecular pathogenesis of NPC, together with their direct relationship with EBV related oncoviral proteins and their role in promoting immune evasion and resistance. We also aim to hypothesize about the feasibility of the use of proteasome inhibitors as part of immunotherapy-including combinatorial regimens for their potential role in reversing immune resistance and favouring tumor recognition and eventual tumor death.
Collapse
Affiliation(s)
- Ramon Yarza
- Medical Oncology Division, Hospital Universitarioss 12 de Octubre, Avda. Córdoba s/n, E-28041, Madrid, Spain. .,Clinical and Translational Laboratory, Instituto de Investigación Hospital 12 de Octubre (I+12), Madrid, Spain.
| | - Mateo Bover
- Medical Oncology Division, Hospital Universitarioss 12 de Octubre, Avda. Córdoba s/n, E-28041, Madrid, Spain.,Clinical and Translational Laboratory, Instituto de Investigación Hospital 12 de Octubre (I+12), Madrid, Spain
| | - Maria Teresa Agulló-Ortuño
- Clinical and Translational Laboratory, Instituto de Investigación Hospital 12 de Octubre (I+12), Madrid, Spain. .,Lung Cancer Group, Clinical Research Program (H12O-CNIO), Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain. .,Biomedical Research Networking Centre: Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain. .,Facultad de Fisioterapia y Enfermería, Universidad de Castilla La Mancha (UCLM), Toledo, Spain.
| | - Lara Carmen Iglesias-Docampo
- Medical Oncology Division, Hospital Universitarioss 12 de Octubre, Avda. Córdoba s/n, E-28041, Madrid, Spain.,Clinical and Translational Laboratory, Instituto de Investigación Hospital 12 de Octubre (I+12), Madrid, Spain.,Lung Cancer Group, Clinical Research Program (H12O-CNIO), Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| |
Collapse
|
41
|
Constanzo J, Faget J, Ursino C, Badie C, Pouget JP. Radiation-Induced Immunity and Toxicities: The Versatility of the cGAS-STING Pathway. Front Immunol 2021; 12:680503. [PMID: 34079557 PMCID: PMC8165314 DOI: 10.3389/fimmu.2021.680503] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 04/26/2021] [Indexed: 12/20/2022] Open
Abstract
In the past decade, radiation therapy (RT) entered the era of personalized medicine, following the striking improvements in radiation delivery and treatment planning optimization, and in the understanding of the cancer response, including the immunological response. The next challenge is to identify the optimal radiation regimen(s) to induce a clinically relevant anti-tumor immunity response. Organs at risks and the tumor microenvironment (e.g. endothelial cells, macrophages and fibroblasts) often limit the radiation regimen effects due to adverse toxicities. Here, we reviewed how RT can modulate the immune response involved in the tumor control and side effects associated with inflammatory processes. Moreover, we discussed the versatile roles of tumor microenvironment components during RT, how the innate immune sensing of RT-induced genotoxicity, through the cGAS-STING pathway, might link the anti-tumor immune response, radiation-induced necrosis and radiation-induced fibrosis, and how a better understanding of the switch between favorable and deleterious events might help to define innovative approaches to increase RT benefits in patients with cancer.
Collapse
Affiliation(s)
- Julie Constanzo
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
| | - Julien Faget
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
| | - Chiara Ursino
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
| | - Christophe Badie
- Cancer Mechanisms and Biomarkers Group, Radiation Effects Department, Centre for Radiation, Chemical & Environmental Hazards Public Health England Chilton, Didcot, United Kingdom
| | - Jean-Pierre Pouget
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
| |
Collapse
|
42
|
Lederer M, Müller S, Glaß M, Bley N, Ihling C, Sinz A, Hüttelmaier S. Oncogenic Potential of the Dual-Function Protein MEX3A. BIOLOGY 2021; 10:415. [PMID: 34067172 PMCID: PMC8151450 DOI: 10.3390/biology10050415] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 04/26/2021] [Accepted: 05/05/2021] [Indexed: 12/23/2022]
Abstract
MEX3A belongs to the MEX3 (Muscle EXcess) protein family consisting of four members (MEX3A-D) in humans. Characteristic for MEX3 proteins is their domain structure with 2 HNRNPK homology (KH) domains mediating RNA binding and a C-terminal really interesting new gene (RING) domain that harbors E3 ligase function. In agreement with their domain composition, MEX3 proteins were reported to modulate both RNA fate and protein ubiquitination. MEX3 paralogs exhibit an oncofetal expression pattern, they are severely downregulated postnatally, and re-expression is observed in various malignancies. Enforced expression of MEX3 proteins in various cancers correlates with poor prognosis, emphasizing their oncogenic potential. The latter is supported by MEX3A's impact on proliferation, self-renewal as well as migration of tumor cells in vitro and tumor growth in xenograft studies.
Collapse
Affiliation(s)
- Marcell Lederer
- Charles Tanford Protein Center, Faculty of Medicine, Institute of Molecular Medicine, Section for Molecular Cell Biology, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str. 3a, 06120 Halle, Germany; (S.M.).; (M.G.).; (N.B.); (S.H.)
| | - Simon Müller
- Charles Tanford Protein Center, Faculty of Medicine, Institute of Molecular Medicine, Section for Molecular Cell Biology, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str. 3a, 06120 Halle, Germany; (S.M.).; (M.G.).; (N.B.); (S.H.)
| | - Markus Glaß
- Charles Tanford Protein Center, Faculty of Medicine, Institute of Molecular Medicine, Section for Molecular Cell Biology, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str. 3a, 06120 Halle, Germany; (S.M.).; (M.G.).; (N.B.); (S.H.)
| | - Nadine Bley
- Charles Tanford Protein Center, Faculty of Medicine, Institute of Molecular Medicine, Section for Molecular Cell Biology, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str. 3a, 06120 Halle, Germany; (S.M.).; (M.G.).; (N.B.); (S.H.)
| | - Christian Ihling
- Center for Structural Mass Spectrometry, Department of Pharmaceutical Chemistry & Bioanalytics, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str. 3, 06120 Halle (Saale), Germany; (C.I.); (A.S.)
| | - Andrea Sinz
- Center for Structural Mass Spectrometry, Department of Pharmaceutical Chemistry & Bioanalytics, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str. 3, 06120 Halle (Saale), Germany; (C.I.); (A.S.)
| | - Stefan Hüttelmaier
- Charles Tanford Protein Center, Faculty of Medicine, Institute of Molecular Medicine, Section for Molecular Cell Biology, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str. 3a, 06120 Halle, Germany; (S.M.).; (M.G.).; (N.B.); (S.H.)
| |
Collapse
|
43
|
Sato Y, Yoshino H, Kashiwakura I, Tsuruga E. DAP3 Is Involved in Modulation of Cellular Radiation Response by RIG-I-Like Receptor Agonist in Human Lung Adenocarcinoma Cells. Int J Mol Sci 2021; 22:E420. [PMID: 33401559 PMCID: PMC7795940 DOI: 10.3390/ijms22010420] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 12/28/2020] [Accepted: 12/30/2020] [Indexed: 12/14/2022] Open
Abstract
Retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs) mediate anti-viral response through mitochondria. In addition, RLR activation induces anti-tumor effects on various cancers. We previously reported that the RLR agonist Poly(I:C)-HMW/LyoVec™ (Poly(I:C)) enhanced radiosensitivity and that cotreatment with Poly(I:C) and ionizing radiation (IR) more than additively increased cell death in lung adenocarcinoma cells, indicating that Poly(I:C) modulates the cellular radiation response. However, it remains unclear how mitochondria are involved in the modulation of this response. Here, we investigated the involvement of mitochondrial dynamics and mitochondrial ribosome protein death-associated protein 3 (DAP3) in the modulation of cellular radiation response by Poly(I:C) in A549 and H1299 human lung adenocarcinoma cell lines. Western blotting revealed that Poly(I:C) decreased the expression of mitochondrial dynamics-related proteins and DAP3. In addition, siRNA experiments showed that DAP3, and not mitochondrial dynamics, is involved in the resistance of lung adenocarcinoma cells to IR-induced cell death. Finally, we revealed that a more-than-additive effect of cotreatment with Poly(I:C) and IR on increasing cell death was diluted by DAP3-knockdown because of an increase in cell death induced by IR alone. Together, our findings suggest that RLR agonist Poly(I:C) modulates the cellular radiation response of lung adenocarcinoma cells by downregulating DAP3 expression.
Collapse
Affiliation(s)
| | - Hironori Yoshino
- Department of Radiation Science, Graduate School of Health Sciences, Hirosaki University, Hirosaki, Aomori 036-8564, Japan; (Y.S.); (I.K.); (E.T.)
| | | | | |
Collapse
|
44
|
Chen M, Hu S, Li Y, Jiang TT, Jin H, Feng L. Targeting nuclear acid-mediated immunity in cancer immune checkpoint inhibitor therapies. Signal Transduct Target Ther 2020; 5:270. [PMID: 33214545 PMCID: PMC7677403 DOI: 10.1038/s41392-020-00347-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 09/11/2020] [Accepted: 09/21/2020] [Indexed: 12/16/2022] Open
Abstract
Cancer immunotherapy especially immune checkpoint inhibition has achieved unprecedented successes in cancer treatment. However, there are many patients who failed to benefit from these therapies, highlighting the need for new combinations to increase the clinical efficacy of immune checkpoint inhibitors. In this review, we summarized the latest discoveries on the combination of nucleic acid-sensing immunity and immune checkpoint inhibitors in cancer immunotherapy. Given the critical role of nuclear acid-mediated immunity in maintaining the activation of T cell function, it seems that harnessing the nuclear acid-mediated immunity opens up new strategies to enhance the effect of immune checkpoint inhibitors for tumor control.
Collapse
Affiliation(s)
- Miaoqin Chen
- Laboratory of Cancer Biology, Key lab of Biotherapy in Zhejiang Province, Cancer Institute of Zhejiang University, Sir Run Run Shaw hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
| | - Shiman Hu
- Laboratory of Cancer Biology, Key lab of Biotherapy in Zhejiang Province, Cancer Institute of Zhejiang University, Sir Run Run Shaw hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
| | - Yiling Li
- Laboratory of Cancer Biology, Key lab of Biotherapy in Zhejiang Province, Cancer Institute of Zhejiang University, Sir Run Run Shaw hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
| | - Ting Ting Jiang
- Department of Radiation Oncology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, 310016, China
| | - Hongchuan Jin
- Laboratory of Cancer Biology, Key lab of Biotherapy in Zhejiang Province, Cancer Institute of Zhejiang University, Sir Run Run Shaw hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
| | - Lifeng Feng
- Laboratory of Cancer Biology, Key lab of Biotherapy in Zhejiang Province, Cancer Institute of Zhejiang University, Sir Run Run Shaw hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China.
| |
Collapse
|
45
|
Iurescia S, Fioretti D, Rinaldi M. The Innate Immune Signalling Pathways: Turning RIG-I Sensor Activation Against Cancer. Cancers (Basel) 2020; 12:E3158. [PMID: 33121210 PMCID: PMC7693898 DOI: 10.3390/cancers12113158] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/22/2020] [Accepted: 10/22/2020] [Indexed: 02/06/2023] Open
Abstract
Over the last 15 years, the ability to harness a patient's own immune system has led to significant progress in cancer therapy. For instance, immunotherapeutic strategies, including checkpoint inhibitors or adoptive cell therapy using chimeric antigen receptor T-cell (CAR-T), are specifically aimed at enhancing adaptive anti-tumour immunity. Several research groups demonstrated that adaptive anti-tumour immunity is highly sustained by innate immune responses. Host innate immunity provides the first line of defence and mediates recognition of danger signals through pattern recognition receptors (PRRs), such as cytosolic sensors of pathogen-associated molecular patterns (PAMPs) and damage-associated molecular pattern (DAMP) signals. The retinoic acid-inducible gene I (RIG-I) is a cytosolic RNA helicase, which detects viral double-strand RNA and, once activated, triggers signalling pathways, converging on the production of type I interferons, proinflammatory cytokines, and programmed cell death. Approaches aimed at activating RIG-I within cancers are being explored as novel therapeutic treatments to generate an inflammatory tumour microenvironment and to facilitate cytotoxic T-cell cross-priming and infiltration. Here, we provide an overview of studies regarding the role of RIG-I signalling in the tumour microenvironment, and the most recent preclinical studies that employ RIG-I agonists. Lastly, we present a selection of clinical trials designed to prove the antitumour role of RIG I and that may result in improved therapeutic outcomes for cancer patients.
Collapse
Affiliation(s)
- Sandra Iurescia
- Institute of Translational Pharmacology (IFT), Department of Biomedical Science, National Research Council (CNR), 00133 Rome, Italy;
| | | | - Monica Rinaldi
- Institute of Translational Pharmacology (IFT), Department of Biomedical Science, National Research Council (CNR), 00133 Rome, Italy;
| |
Collapse
|
46
|
Protein phosphatase 1 in tumorigenesis: is it worth a closer look? Biochim Biophys Acta Rev Cancer 2020; 1874:188433. [PMID: 32956763 DOI: 10.1016/j.bbcan.2020.188433] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/26/2020] [Accepted: 09/12/2020] [Indexed: 02/06/2023]
Abstract
Cancer cells take advantage of signaling cascades to meet their requirements for sustained growth and survival. Cell signaling is tightly controlled by reversible protein phosphorylation mechanisms, which require the counterbalanced action of protein kinases and protein phosphatases. Imbalances on this system are associated with cancer development and progression. Protein phosphatase 1 (PP1) is one of the most relevant protein phosphatases in eukaryotic cells. Despite the widely recognized involvement of PP1 in key biological processes, both in health and disease, its relevance in cancer has been largely neglected. Here, we provide compelling evidence that support major roles for PP1 in tumorigenesis.
Collapse
|
47
|
Zhou B, Li C, Yang Y, Wang Z. RIG-I Promotes Cell Death in Hepatocellular Carcinoma by Inducing M1 Polarization of Perineal Macrophages Through the RIG-I/MAVS/NF-κB Pathway. Onco Targets Ther 2020; 13:8783-8794. [PMID: 32982277 PMCID: PMC7493023 DOI: 10.2147/ott.s258450] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 08/16/2020] [Indexed: 12/11/2022] Open
Abstract
Background The development and metastasis of cancer cells are regulated by tumor-associated macrophages (TAMs) present in the surrounding tumor microenvironment. RIG-I is a key pathogen recognition receptor against RNA viruses that regulates innate immunity in cancer progression. Till now, the mechanism of RIG-I regulation of the polarization of TAMs in the progression of hepatocellular carcinoma (HCC) has not been understood. Materials and Methods Levels of RIG-I and the key proteins in the NF-κB pathway in HCC and paired paracancerous tissues were detected by Western blotting. The transfection efficiency of RIG-I was observed by fluorescence microscopy. The M1 and M2 markers were detected by real-time polymerase chain reaction and FACS assays. Apoptosis of RIG-I lentivirus-infected HCC cells was detected by flow cytometry assay. Death of Hepa1-6 and H22 cells was analyzed by lactate dehydrogenase releasing assay. Results The level of RIG-I was decreased in HCC tissues as compared to that in the paired paracancerous tissues. Overexpression of RIG-I in mouse peritoneal macrophages increased the expression of the biomarkers CD16/32 and CD11c associated with M1 macrophages. The relative levels of IL-1β, TNF-α, IL-6, and iNOS were significantly increased in RIG-I lentivirus-infected macrophages, whereas the levels of Arg-1 and IL-10 were not significantly different in RIG-I-overexpressed peritoneal macrophages. Moreover, overexpression of RIG-I in peritoneal macrophages promoted apoptosis of Hepa1-6 and H22 cells. Furthermore, overexpression of RIG-I increased the levels of phosphorylated p65 and p-IκB and decreased the level of IκB in peritoneal macrophages. Importantly, the expression of MAVS and TRAF2 was significantly increased in RIG-I lentivirus-infected macrophages. Conclusion Our results demonstrate that overexpression of RIG-I promoted apoptosis and death of HCC cells. Moreover, RIG-I promoted the polarization of M1 through the RIG-I/MAVS/TRAF2/NF-κB pathway in mice peritoneal macrophages, suggesting that RIG-I may be a novel target in the immunotherapy of HCC.
Collapse
Affiliation(s)
- Bei Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, People's Republic of China
| | - Cuiping Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, People's Republic of China
| | - Yun Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, People's Republic of China
| | - Zhuo Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, People's Republic of China
| |
Collapse
|
48
|
Abstract
Retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs) are key sensors of virus infection, mediating the transcriptional induction of type I interferons and other genes that collectively establish an antiviral host response. Recent studies have revealed that both viral and host-derived RNAs can trigger RLR activation; this can lead to an effective antiviral response but also immunopathology if RLR activities are uncontrolled. In this Review, we discuss recent advances in our understanding of the types of RNA sensed by RLRs in the contexts of viral infection, malignancies and autoimmune diseases. We further describe how the activity of RLRs is controlled by host regulatory mechanisms, including RLR-interacting proteins, post-translational modifications and non-coding RNAs. Finally, we discuss key outstanding questions in the RLR field, including how our knowledge of RLR biology could be translated into new therapeutics.
Collapse
Affiliation(s)
- Jan Rehwinkel
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| | - Michaela U Gack
- Department of Microbiology, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
49
|
Hubert M, Gobbini E, Couillault C, Manh TPV, Doffin AC, Berthet J, Rodriguez C, Ollion V, Kielbassa J, Sajous C, Treilleux I, Tredan O, Dubois B, Dalod M, Bendriss-Vermare N, Caux C, Valladeau-Guilemond J. IFN-III is selectively produced by cDC1 and predicts good clinical outcome in breast cancer. Sci Immunol 2020; 5:5/46/eaav3942. [PMID: 32303573 DOI: 10.1126/sciimmunol.aav3942] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 11/02/2019] [Accepted: 03/26/2020] [Indexed: 12/14/2022]
Abstract
Dendritic cells play a key role in the orchestration of antitumor immune responses. The cDC1 (conventional dendritic cell 1) subset has been shown to be essential for antitumor responses and response to immunotherapy, but its precise role in humans is largely unexplored. Using a multidisciplinary approach, we demonstrate that human cDC1 play an important role in the antitumor immune response through their capacity to produce type III interferon (IFN-λ). By analyzing a large cohort of breast primary tumors and public transcriptomic datasets, we observed specific production of IFN-λ1 by cDC1. In addition, both IFN-λ1 and its receptor were associated with favorable patient outcomes. We show that IFN-III promotes a TH1 microenvironment through increased production of IL-12p70, IFN-γ, and cytotoxic lymphocyte-recruiting chemokines. Last, we showed that engagement of TLR3 is a therapeutic strategy to induce IFN-III production by tumor-associated cDC1. These data provide insight into potential IFN- or cDC1-targeting antitumor therapies.
Collapse
Affiliation(s)
- Margaux Hubert
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69008, France.,Laboratoire d'Immunothérapie des Cancers de Lyon (LICL), Lyon, France
| | - Elisa Gobbini
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69008, France.,CHU Grenoble-Alpes, France
| | - Coline Couillault
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69008, France
| | - Thien-Phong Vu Manh
- Aix Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Anne-Claire Doffin
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69008, France
| | - Justine Berthet
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69008, France.,Laboratoire d'Immunothérapie des Cancers de Lyon (LICL), Lyon, France
| | - Céline Rodriguez
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69008, France.,Laboratoire d'Immunothérapie des Cancers de Lyon (LICL), Lyon, France
| | - Vincent Ollion
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69008, France.,LabEx DEVweCAN, Lyon, France
| | - Janice Kielbassa
- Synergie Lyon Cancer, Plateforme de Bio-informatique 'Gilles Thomas', Lyon, France
| | - Christophe Sajous
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69008, France
| | | | | | - Bertrand Dubois
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69008, France.,Laboratoire d'Immunothérapie des Cancers de Lyon (LICL), Lyon, France
| | - Marc Dalod
- Aix Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Nathalie Bendriss-Vermare
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69008, France.,Laboratoire d'Immunothérapie des Cancers de Lyon (LICL), Lyon, France.,LabEx DEVweCAN, Lyon, France
| | - Christophe Caux
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69008, France.,Laboratoire d'Immunothérapie des Cancers de Lyon (LICL), Lyon, France.,LabEx DEVweCAN, Lyon, France.,Centre Léon Bérard, F-69008 Lyon, France
| | - Jenny Valladeau-Guilemond
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69008, France. .,LabEx DEVweCAN, Lyon, France
| |
Collapse
|
50
|
Wang S, Zhou L, Ling L, Meng X, Chu F, Zhang S, Zhou F. The Crosstalk Between Hippo-YAP Pathway and Innate Immunity. Front Immunol 2020; 11:323. [PMID: 32174922 PMCID: PMC7056731 DOI: 10.3389/fimmu.2020.00323] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/10/2020] [Indexed: 12/12/2022] Open
Abstract
Recognition of pathogen-associated molecular patterns (PAMPs) triggers expression of antiviral interferons and proinflammatory cytokines, which functions as the frontier of host defense against microbial pathogen invasion. Hippo-YAP pathway regulates cell proliferation, survival, differentiation and is involved in diverse life processes, including tissue homeostasis and tumor suppression. Emerging discoveries elucidated that the components of Hippo-YAP pathway, such as MST1/2, NDR1/2, and YAP/TAZ played crucial regulatory roles in innate immunity. Meanwhile the innate immune signaling also exhibited regulatory effect on Hippo-YAP pathway. As for the importance of these two pathways, it would be interesting to figure out the deeper biological implications of their interplays. This review focuses on the regulation between Hippo-YAP pathway and innate immune signaling. We also propose the possible contribution of these interplays to tumor development.
Collapse
Affiliation(s)
- Shuai Wang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Lili Zhou
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Li Ling
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Xuli Meng
- Department of Breast Surgery, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Feng Chu
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Suping Zhang
- Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Pharmacology, Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Fangfang Zhou
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| |
Collapse
|