1
|
Feng X, Feng Q, Abbas Raza SH, Li F, Ma Y. Identification of key factors causing ketosis in dairy cows with low feed intake. Anim Biotechnol 2025; 36:2487089. [PMID: 40184169 DOI: 10.1080/10495398.2025.2487089] [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: 12/16/2024] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
Ketosis is a common metabolic disease in high-yield dairy cows. Key genes affecting ketosis need to be further explored by new methods. The gene expression profiling and clinical data of GSE92398, GSE104079, and GSE4304 were obtained from the gene expression omnibus (GEO) database. Core modules and genes associated with RFI (residual feed intake) and ADF (alternate day fasting) were identified by weighted gene co-expression network analysis (WGCNA). Subsequently, the key genes related to ketosis and RFI were determined by protein-protein interaction (PPI) networks, ROC curves, functional enrichment, and differential expression analysis, respectively. The results showed that the genes of ACACA, ELOVL6 and XPO7 could be used as regulators of ketosis induced by low feed intake in dairy cows. At the same time, three genes (HRFI, STAT3 and IFNAR1) were retained as additional RFI biomarkers that could be considered. We identified three key factors as candidate genes and biomarkers of ketosis and RFI, respectively. These factors may provide a theoretical basis for targeted therapy of ketosis in dairy cows.
Collapse
Affiliation(s)
- Xue Feng
- Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Qi Feng
- School of Computer and Information Engineering, Henan University, Kaifeng, China
| | - Sayed Haidar Abbas Raza
- Research Center for Machining and Safety of Livestock and Poultry Products, South China Agricultural University, Guangzhou, China
| | - Fen Li
- Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Yun Ma
- Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University, Yinchuan, China
| |
Collapse
|
2
|
Liu C, Tang L, Yang W, Gu Y, Xu W, Liang Z, Jiang J. cGAS/STING pathway and gastrointestinal cancer: Mechanisms and diagnostic and therapeutic targets (Review). Oncol Rep 2025; 53:15. [PMID: 39611480 PMCID: PMC11632663 DOI: 10.3892/or.2024.8848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 10/23/2024] [Indexed: 11/30/2024] Open
Abstract
The health of individuals is seriously threatened by intestinal cancer, which includes pancreatic, colorectal, esophageal, gastric and gallbladder cancer. Most gastrointestinal cancers do not have typical and specific early symptoms, and lack specific and effective diagnostic markers and treatment methods. It is critical to understand the etiology of gastrointestinal cancer and develop more efficient methods of diagnosis and treatment. The cyclic GMP‑AMP synthase (cGAS)/stimulator of interferon genes (STING) pathway serves a crucial role in the occurrence, progression and treatment of gastrointestinal cancer. The present review focuses on the latest progress regarding the role and mechanism of the cGAS/STING pathway in gastrointestinal cancer, and discusses treatment approaches and related applications based on the cGAS/STING signaling pathway. In order to improve the knowledge of the connection between the cGAS/STING pathway and gastrointestinal cancer, aid the diagnosis and treatment of gastrointestinal cancer, and lessen the burden on patients and society, the present review also discusses future research directions and existing challenges regarding cGAS/STING in the study of gastrointestinal cancer.
Collapse
Affiliation(s)
- Chang Liu
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, Jiangsu 215600, P.R. China
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Li Tang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Wenhui Yang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Yuning Gu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Wenrong Xu
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, Jiangsu 215600, P.R. China
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Zhaofeng Liang
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, Jiangsu 215600, P.R. China
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Jiajia Jiang
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, Jiangsu 215600, P.R. China
| |
Collapse
|
3
|
Yamamoto G, Tanaka K, Kamata R, Saito H, Yamamori-Morita T, Nakao T, Liu J, Mori S, Yagishita S, Hamada A, Shinno Y, Yoshida T, Horinouchi H, Ohe Y, Watanabe SI, Yatabe Y, Kitai H, Konno S, Kobayashi SS, Ohashi A. WEE1 confers resistance to KRAS G12C inhibitors in non-small cell lung cancer. Cancer Lett 2024; 611:217414. [PMID: 39725152 DOI: 10.1016/j.canlet.2024.217414] [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: 09/30/2024] [Revised: 12/18/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
KRASG12C inhibitors sotorasib and adagrasib have been approved for the treatment of KRASG12C-mutant non-small cell lung cancer (NSCLC). However, the efficacy of single-agent treatments is limited, presumably due to multiple resistance mechanisms. To overcome these therapeutic limitations, combination strategies that potentiate the antitumor efficacy of KRASG12C inhibitors must be developed. Through unbiased high-throughput screening of 1395 kinase inhibitors, we identified adavosertib, a WEE1 inhibitor, as a promising combination partner of sotorasib. The combination of sotorasib and adavosertib exhibited synergistic antiproliferative activities both in vitro and in vivo, irrespective of TP53, STK11, and KEAP1 co-mutation profiles. WEE1 inhibition potentiated MCL-1-mediated apoptosis in sotorasib-treated cancer cells. Mechanistically, the combination downregulated MCL-1 protein levels by attenuating de novo translation and enhancing its degradation. WEE1 overexpression conferred resistance against sotorasib via MCL-1 upregulation. Moreover, cells that acquired sotorasib resistance profoundly upregulated both WEE1 and MCL-1 proteins, highlighting WEE1 as a crucial driver of sotorasib resistance. Importantly, WEE1 inhibition re-sensitized resistant cells to sotorasib treatment. The current findings demonstrate that combined inhibition of KRASG12C and WEE1 not only exhibits synergistic antitumor efficacy but also overcomes resistance to KRASG12C inhibitors, thus representing a novel therapeutic strategy for KRASG12C-mutant NSCLC.
Collapse
Affiliation(s)
- Gaku Yamamoto
- Division of Collaborative Research and Developments, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan; Division of Translational Genomics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan; Division of Translational Informatics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan; Department of Respiratory Medicine, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Kosuke Tanaka
- Division of Translational Genomics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan; Division of Cancer Immunology, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan; Division of Cancer Immunology, National Cancer Center Research Institute, Tokyo, Japan.
| | - Ryo Kamata
- Division of Collaborative Research and Developments, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan; Division of Translational Genomics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Hitoshi Saito
- Division of Collaborative Research and Developments, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan; Division of Translational Genomics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Tomoko Yamamori-Morita
- Division of Collaborative Research and Developments, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan; Division of Translational Genomics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Takehiro Nakao
- Division of Collaborative Research and Developments, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan; Division of Translational Genomics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Jie Liu
- Division of Translational Genomics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Shunta Mori
- Division of Translational Genomics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Shigehiro Yagishita
- Division of Molecular Pharmacology, National Cancer Center Research Institute, Tokyo, Japan
| | - Akinobu Hamada
- Division of Molecular Pharmacology, National Cancer Center Research Institute, Tokyo, Japan
| | - Yuki Shinno
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Tatsuya Yoshida
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hidehito Horinouchi
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yuichiro Ohe
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Shun-Ichi Watanabe
- Division of Thoracic Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Yasushi Yatabe
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
| | - Hidenori Kitai
- Department of Respiratory Medicine, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Satoshi Konno
- Department of Respiratory Medicine, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Susumu S Kobayashi
- Division of Translational Genomics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan; Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Akihiro Ohashi
- Division of Collaborative Research and Developments, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan; Division of Translational Genomics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan.
| |
Collapse
|
4
|
Jia Z, Zhou X, Liu J, De X, Li Y, Yang Z, Duan H, Wang F, Ge J. Immune enhancement of rhamnolipid/manganese calcium phosphate mineralized nanoparticle: A promising subunit antigen delivery system. Int J Biol Macromol 2024; 282:137239. [PMID: 39491710 DOI: 10.1016/j.ijbiomac.2024.137239] [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: 07/13/2023] [Revised: 10/16/2024] [Accepted: 11/01/2024] [Indexed: 11/05/2024]
Abstract
The use of biomimetic mineralization strategy is promising to solve the problem of poor stability and immune effect of subunit antigens. However, non-specifically inducing protein mineralization is still a challenge. we hypothesized that rhamnolipids with both protein and metal binding capacity could be used to develop more functional and biocompatible calcium mineralized nanoparticle (RMCP). The results show that rhamnolipids synergistically enhanced the mineralization of protein with manganese ions and improved 21 % the loading antigens of RMCP compared to manganese calcium phosphate nanoparticles. Transmission electron microscopy (TEM) and Dynamic Light Scattering (DLS) showed particle size of RMCP is 260 ± 12.1 nm with spherical morphology. In vitro experiments have shown that RMCP effectively activate immune cells through the cGAS-STING and NLRP3 pathways and demonstrated a higher level of cytokines in RAW264.7 Macrophages. In vivo, RMCP triggered an increased IgG titer with 16.5-fold IgG2a/IgG1 ratio compared to the aluminum adjuvant which improved the recovery status after challenge in mice. We used biological surfactants for the first time to enhance the biomimetic mineralization process of subunit antigen, which provides a new approach for constructing calcium-based biocompatible antigen delivery vectors, helping to develop a new generation of stable, efficient, and safe subunit vaccines.
Collapse
Affiliation(s)
- Zheng Jia
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China
| | - Xinyao Zhou
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China
| | - Jingjing Liu
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China; State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150086, China
| | - Xinqi De
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China
| | - Yifan Li
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China
| | - Zaixing Yang
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China
| | - Haoyuan Duan
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China
| | - Fang Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150086, China
| | - Junwei Ge
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China; Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China.
| |
Collapse
|
5
|
Manetsch P, Böhi F, Nowak K, Leslie Pedrioli DM, Hottiger MO. PARP7-mediated ADP-ribosylation of FRA1 promotes cancer cell growth by repressing IRF1- and IRF3-dependent apoptosis. Proc Natl Acad Sci U S A 2023; 120:e2309047120. [PMID: 38011562 DOI: 10.1073/pnas.2309047120] [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: 05/31/2023] [Accepted: 10/26/2023] [Indexed: 11/29/2023] Open
Abstract
PARP7 was reported to promote tumor growth in a cell-autonomous manner and by repressing the antitumor immune response. Nevertheless, the molecular mechanism of how PARP7-mediated ADP-ribosylation exerts these effects in cancer cells remains elusive. Here, we identified PARP7 as a nuclear and cysteine-specific mono-ADP-ribosyltransferase that modifies targets critical for regulating transcription, including the AP-1 transcription factor FRA1. Loss of FRA1 ADP-ribosylation via PARP7 inhibition by RBN-2397 or mutation of the ADP-ribosylation site C97 increased FRA1 degradation by the proteasome via PSMC3. The reduction in FRA1 protein levels promoted IRF1- and IRF3-dependent cytokine as well as proapoptotic gene expression, culminating in CASP8-mediated apoptosis. Furthermore, high PARP7 expression was indicative of the PARP7 inhibitor response in FRA1-positive lung and breast cancer cells. Collectively, our findings highlight the connected roles of PARP7 and FRA1 and emphasize the clinical potential of PARP7 inhibitors for FRA1-driven cancers.
Collapse
Affiliation(s)
- Patrick Manetsch
- Department of Molecular Mechanisms of Disease, University of Zurich, 8057 Zurich, Switzerland
- Molecular Life Science Ph.D. Program, Life Science Zurich Graduate School, University of Zurich, 8057 Zurich, Switzerland
| | - Flurina Böhi
- Department of Molecular Mechanisms of Disease, University of Zurich, 8057 Zurich, Switzerland
- Cancer Biology Ph.D. Program, Life Science Zurich Graduate School, University of Zurich, 8057 Zurich, Switzerland
| | - Kathrin Nowak
- Department of Molecular Mechanisms of Disease, University of Zurich, 8057 Zurich, Switzerland
| | - Deena M Leslie Pedrioli
- Department of Molecular Mechanisms of Disease, University of Zurich, 8057 Zurich, Switzerland
| | - Michael O Hottiger
- Department of Molecular Mechanisms of Disease, University of Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
6
|
Zhang X, Cai X, Yan C. Opportunities and challenges in combining immunotherapy and radiotherapy in esophageal cancer. J Cancer Res Clin Oncol 2023; 149:18253-18270. [PMID: 37985502 PMCID: PMC10725359 DOI: 10.1007/s00432-023-05499-z] [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: 10/11/2023] [Accepted: 10/30/2023] [Indexed: 11/22/2023]
Abstract
BACKGROUND Immunotherapy has shown promise in the treatment of esophageal cancer, but using it alone only benefits a small number of patients. Most patients either do not have a significant response or develop secondary drug resistance. The combination of radiotherapy and immunotherapy appears to be a promising approach to treating esophageal cancer. PURPOSE We reviewed milestone clinical trials of radiotherapy combined with immunotherapy for esophageal cancer. We then discussed potential biomarkers for radiotherapy combined with immunotherapy, including programmed cell death-ligand 1 (PD-L1) status, tumor mutation burden (TMB), tumor-infiltrating lymphocytes, ct-DNA, imaging biomarkers, and clinical factors. Furthermore, we emphasize the key mechanisms of radiation therapy-induced immune stimulation and immune suppression in order to propose strategies for overcoming immune resistance in radiation therapy (RT). Lastly, we discussed the emerging role of low-dose radiotherapy (LDRT) , which has become a promising approach to overcome the limitations of high-dose radiotherapy. CONCLUSION Radiotherapy can be considered a triggering factor for systemic anti-tumor immune response and, with the assistance of immunotherapy, can serve as a systemic treatment option and potentially become the standard treatment for cancer patients.
Collapse
Affiliation(s)
- Xinyu Zhang
- Weifang Hospital of Traditional Chinese Medicine, 666 Weizhou Road, Weifang, 261000, Shandong, China
- Shandong University of Traditional Chinese Medicine, Jinan, 250000, Shandong, China
| | - Xinsheng Cai
- Weifang Hospital of Traditional Chinese Medicine, 666 Weizhou Road, Weifang, 261000, Shandong, China
| | - Chaoguang Yan
- Weifang Hospital of Traditional Chinese Medicine, 666 Weizhou Road, Weifang, 261000, Shandong, China.
| |
Collapse
|
7
|
Kong P, Yang M, Wang Y, Yu KN, Wu L, Han W. Ferroptosis triggered by STAT1- IRF1-ACSL4 pathway was involved in radiation-induced intestinal injury. Redox Biol 2023; 66:102857. [PMID: 37611494 PMCID: PMC10466894 DOI: 10.1016/j.redox.2023.102857] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 08/16/2023] [Indexed: 08/25/2023] Open
Abstract
Radiation-induced intestinal injury (RIII), a common gastrointestinal complication caused by radiotherapy on pelvic, abdominal and retroperitoneal tumors, seriously affects the life quality of patients and may result in termination of radiotherapy. At present, the pathogenesis of RIII has not been fully understood. Herein, we demonstrated that ferroptosis played a critical role in RIII occurrence. The RNA sequencing analysis strongly hinted ferroptosis was involved in RIII mice. In line with this, the levels of 4-hydroxynonenal (4-HNE) and malondialdehyde (MDA), markers of lipid peroxidation, remarkably increased in RIII mice. And the ferroptosis inhibitor, Ferrostatin-1 (Fer-1), improved the mice survival and alleviated intestinal fibrosis in vivo. Moreover, our results revealed that arachidonic acid (AA) enhanced ferroptosis in cultured intestinal epithelial cells (IECs) and organoids in vitro after irradiation, and AA gavage aggravated RIII in mice. Mechanistic studies revealed the level of ACSL4 protein significantly increased in mouse jejunums and IECs after irradiation. Radiation-induced ferroptosis in IECs was also prevented following ACSL4 knockdown or with the function inhibitor of ACSL4. Furthermore, we found that transcription of ACSL4 induced by irradiation was regulated by STAT1/IRF1 axis, and AMPK activation triggered by AA negatively regulated radiation-induced ferroptosis. Taken together, our results suggest that ferroptosis mediates RIII and reducing dietary AA intake as well as targeting the STAT1-IRF1-ACSL4 axis or AMPK may be the potential approaches to alleviate RIII.
Collapse
Affiliation(s)
- Peizhong Kong
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, PR China
| | - Miaomiao Yang
- The First Affiliated Hospital of Anhui Medical University, Hefei, 230011, PR China; Anhui Public Health Clinical Center, Hefei, 230011, PR China
| | - Ying Wang
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, PR China
| | - K N Yu
- Department of Physics, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, 999077, Hong Kong, China; State Key Laboratory in Marine Pollution, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, 999077, Hong Kong, China
| | - Lijun Wu
- Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601, PR China
| | - Wei Han
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, PR China; Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, PR China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions and School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou, 215006, PR China.
| |
Collapse
|
8
|
Du J, Kageyama SI, Yamashita R, Tanaka K, Okumura M, Motegi A, Hojo H, Nakamura M, Hirata H, Sunakawa H, Kotani D, Yano T, Kojima T, Hamaya Y, Kojima M, Nakamura Y, Suzuki A, Suzuki Y, Tsuchihara K, Akimoto T. Transposable elements potentiate radiotherapy-induced cellular immune reactions via RIG-I-mediated virus-sensing pathways. Commun Biol 2023; 6:818. [PMID: 37543704 PMCID: PMC10404237 DOI: 10.1038/s42003-023-05080-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 06/28/2023] [Indexed: 08/07/2023] Open
Abstract
Radiotherapy (RT) plus immunotherapy is a promising modality; however, the therapeutic effects are insufficient, and the molecular mechanism requires clarification to further develop combination therapies. Here, we found that the RNA virus sensor pathway dominantly regulates the cellular immune response in NSCLC and ESCC cell lines. Notably, transposable elements (TEs), especially long terminal repeats (LTRs), functioned as key ligands for the RNA virus sensor RIG-I, and the mTOR-LTR-RIG-I axis induced the cellular immune response and dendritic cell and macrophage infiltration after irradiation. Moreover, RIG-I-dependent immune activation was observed in ESCC patient tissue. scRNA sequencing and spatial transcriptome analysis revealed that radiotherapy induced the expression of LTRs, and the RNA virus sensor pathway in immune and cancer cells; this pathway was also found to mediate tumour conversion to an immunological hot state. Here, we report the upstream and ligand of the RNA virus sensor pathway functions in irradiated cancer tissues.
Collapse
Affiliation(s)
- Junyan Du
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Shun-Ichiro Kageyama
- Division of Radiation Oncology and Particle Therapy, National Cancer Center Hospital East, Chiba, Japan.
- Department of Radiation Oncology, National Cancer Center Hospital East, Chiba, Japan.
| | - Riu Yamashita
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Kosuke Tanaka
- Division of Cancer Immunology, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Masayuki Okumura
- Division of Radiation Oncology and Particle Therapy, National Cancer Center Hospital East, Chiba, Japan
| | - Atsushi Motegi
- Division of Radiation Oncology and Particle Therapy, National Cancer Center Hospital East, Chiba, Japan
| | - Hidehiro Hojo
- Division of Radiation Oncology and Particle Therapy, National Cancer Center Hospital East, Chiba, Japan
| | - Masaki Nakamura
- Division of Radiation Oncology and Particle Therapy, National Cancer Center Hospital East, Chiba, Japan
| | - Hidenari Hirata
- Division of Radiation Oncology and Particle Therapy, National Cancer Center Hospital East, Chiba, Japan
| | - Hironori Sunakawa
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital East, Chiba, Japan
| | - Daisuke Kotani
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Tomonori Yano
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital East, Chiba, Japan
| | - Takashi Kojima
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Yamato Hamaya
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Motohiro Kojima
- Division of Pathology, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Yuka Nakamura
- Division of Pathology, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Ayako Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Katsuya Tsuchihara
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Tetsuo Akimoto
- Division of Radiation Oncology and Particle Therapy, National Cancer Center Hospital East, Chiba, Japan
- Department of Radiation Oncology, National Cancer Center Hospital East, Chiba, Japan
| |
Collapse
|
9
|
Fan J, Li Q, Liang J, Chen Z, Chen L, Lai J, Chen Q. Regulation of IFNβ expression: focusing on the role of its promoter and transcription regulators. Front Microbiol 2023; 14:1158777. [PMID: 37396372 PMCID: PMC10309559 DOI: 10.3389/fmicb.2023.1158777] [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: 02/07/2023] [Accepted: 05/23/2023] [Indexed: 07/04/2023] Open
Abstract
IFNβ is a single-copy gene without an intron. Under normal circumstances, it shows low or no expression in cells. It is upregulated only when the body needs it or is stimulated. Stimuli bind to the pattern recognition receptors (PRRs) and pass via various signaling pathways to several basic transcriptional regulators, such as IRFs, NF-кB, and AP-1. Subsequently, the transcriptional regulators enter the nucleus and bind to regulatory elements of the IFNβ promoter. After various modifications, the position of the nucleosome is altered and the complex is assembled to activate the IFNβ expression. However, IFNβ regulation involves a complex network. For the study of immunity and diseases, it is important to understand how transcription factors bind to regulatory elements through specific forms, which elements in cells are involved in regulation, what regulation occurs during the assembly of enhancers and transcription complexes, and the possible regulatory mechanisms after transcription. Thus, this review focuses on the various regulatory mechanisms and elements involved in the activation of IFNβ expression. In addition, we discuss the impact of this regulation in biology.
Collapse
Affiliation(s)
- Jiqiang Fan
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou, China
| | - Qiumei Li
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou, China
| | - Jiadi Liang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou, China
| | - Zhirong Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou, China
| | - Linqin Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou, China
| | - Junzhong Lai
- The Cancer Center, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Qi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou, China
| |
Collapse
|
10
|
Yang L, Zhang X, Huang X, Dong X, Jing S, Zhang Y, Zhao B, Wang Z, Qu H. Correlation between IFNAR1 expression in peripheral blood T lymphocytes and inflammatory cytokines, tumor-infiltrating lymphocytes, and chemosensitivity in patients with colorectal cancer. Cytokine 2022; 159:156008. [PMID: 36063748 DOI: 10.1016/j.cyto.2022.156008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 08/03/2022] [Accepted: 08/06/2022] [Indexed: 11/15/2022]
Abstract
IFN-α receptor (IFNAR) is critical for maintaining the crosstalk between cancer cells and lymphocytes. We investigated IFNAR1 expression in peripheral blood CD4+ and CD8+ T cells and explored their relationships with plasma cytokines, chemosensitivity and infiltrated T cells in the tumor microenvironment (TME) of colorectal cancer (CRC). The levels of IFNAR1, IFN-γ, and PD1 in peripheral T cells were tested using flow cytometry. Immunohistochemical staining of IFNAR1 in CRC tissues was performed. A cytometric bead array was used to determine the plasma concentrations of cytokines. In CRC patients, IFNAR1 levels were significantly increased in peripheral blood T cells, and plasma IL-6 levels were also significantly increased. Pearson correlation analysis revealed that IFNAR1 expression in CD8+ T cells was negatively associated with plasma IL-2, IFN-γ, and TNFα. IFNAR1 expression in CD4+ T cells was positively associated with TME infiltrated levels of CD8+ T cells. The levels of CD8+ T cells with IFNAR1 and plasma IFN-γ were associated with chemosensitivity. Collectively, IFNAR1 levels in CD4+ and CD8+ T cells were significantly upregulated in CRC patients and positively associated with T-cell infiltration. IFNAR1 may be a chemotherapy biomarker for predicting response.
Collapse
Affiliation(s)
- Lei Yang
- Department of General Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Xiaojing Zhang
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Xiaoxi Huang
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Xichen Dong
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Shui Jing
- Department of General Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Yudong Zhang
- Department of General Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Baocheng Zhao
- Department of General Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| | - Zhenjun Wang
- Department of General Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| | - Hao Qu
- Department of General Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| |
Collapse
|