1
|
Khan M, Huang X, Ye X, Zhang D, Wang B, Xu A, Li R, Ren A, Chen C, Song J, Zheng R, Yuan Y, Lin J. Necroptosis-based glioblastoma prognostic subtypes: implications for TME remodeling and therapy response. Ann Med 2024; 56:2405079. [PMID: 39387496 PMCID: PMC11469424 DOI: 10.1080/07853890.2024.2405079] [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: 05/09/2024] [Revised: 08/14/2024] [Accepted: 08/28/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) is an aggressive primary brain tumor with a high recurrence rate and poor prognosis. Necroptosis, a pathological hallmark of GBM, is poorly understood in terms of its role in prognosis, tumor microenvironment (TME) alteration, and immunotherapy. METHODS & RESULTS We assessed the expression of 55 necroptosis-related genes in GBM and normal brain tissues. We identified necroptosis-stratified clusters using Uni-Cox and Least Absolute Shrinkage and Selection Operator (LASSO) regression to establish the 10-gene Glioblastoma Necroptosis Index (GNI). GNI demonstrated significant prognostic efficacy in the TCGA dataset (n = 160) and internal validation dataset (n = 345) and in external validation cohorts (n = 591). The GNI-high subgroup displayed a mesenchymal phenotype, lacking the IDH1 mutation, and MGMT methylation. This subgroup was characterized by significant enrichment in inflammatory and humoral immune pathways with prominent cell adhesion molecules (CD44 and ICAM1), inflammatory cytokines (TGFB1, IL1B, and IL10), and chemokines (CX3CL1, CXCL9, and CCL5). The TME in this subgroup showed elevated infiltration of M0 macrophages, neutrophils, mast cells, and regulatory T cells. GNI-related genes appeared to limit macrophage polarization, as confirmed by immunohistochemistry and flow cytometry. The top 30% high-risk score subset exhibited increased CD8 T cell infiltration and enhanced cytolytic activity. GNI showed promise in predicting responses to immunotherapy and targeted treatment. CONCLUSIONS Our study highlights the role of necroptosis-related genes in glioblastoma (GBM) and their effects on the tumor microenvironment and patient prognosis. TheGNI demonstrates potential as a prognostic marker and provides insights into immune characteristics and treatment responsiveness.
Collapse
Affiliation(s)
- Muhammad Khan
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Xiuting Huang
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Xiaoxin Ye
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Donghui Zhang
- Department of Pathology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Baiyao Wang
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Anan Xu
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Rong Li
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Anbang Ren
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Chengcong Chen
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Jingjing Song
- Department of Pathology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Rong Zheng
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou, People’s Republic of China
- Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors, Fujian Medical University, Fuzhou, People’s Republic of China
- Clinical Research Center for Radiology and Radiotherapy of Fujian Province (Digestive, Hematological and Breast Malignancies), Fuzhou, People’s Republic of China
| | - Yawei Yuan
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Jie Lin
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| |
Collapse
|
2
|
Ding Y, Sun Y, Wang H, Zhao H, Yin R, Zhang M, Pan X, Zhu X. Atherosis-associated lnc_000048 activates PKR to enhance STAT1-mediated polarization of THP-1 macrophages to M1 phenotype. Neural Regen Res 2024; 19:2488-2498. [PMID: 38526285 PMCID: PMC11090429 DOI: 10.4103/nrr.nrr-d-23-01355] [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: 08/13/2023] [Revised: 12/12/2023] [Accepted: 01/20/2024] [Indexed: 03/26/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202419110-00029/figure1/v/2024-03-08T184507Z/r/image-tiff Our previous study has demonstrated that lnc_000048 is upregulated in large-artery atherosclerotic stroke and promotes atherosclerosis in ApoE-/- mice. However, little is known about the role of lnc_000048 in classically activated macrophage (M1) polarization. In this study, we established THP-1-derived testing state macrophages (M0), M1 macrophages, and alternately activated macrophages (M2). Real-time fluorescence quantitative PCR was used to verify the expression of marker genes and the expression of lnc_000048 in macrophages. Flow cytometry was used to detect phenotypic proteins (CD11b, CD38, CD80). We generated cell lines with lentivirus-mediated upregulation or downregulation of lnc_000048. Flow cytometry, western blot, and real-time fluorescence quantitative PCR results showed that down-regulation of lnc_000048 reduced M1 macrophage polarization and the inflammation response, while over-expression of lnc_000048 led to the opposite effect. Western blot results indicated that lnc_000048 enhanced the activation of the STAT1 pathway and mediated the M1 macrophage polarization. Moreover, catRAPID prediction, RNA-pull down, and mass spectrometry were used to identify and screen the protein kinase RNA-activated (PKR), then catRAPID and RPIseq were used to predict the binding ability of lnc_000048 to PKR. Immunofluorescence (IF)-RNA fluorescence in situ hybridization (FISH) double labeling was performed to verify the subcellular colocalization of lnc_000048 and PKR in the cytoplasm of M1 macrophage. We speculate that lnc_000048 may form stem-loop structure-specific binding and activate PKR by inducing its phosphorylation, leading to activation of STAT1 phosphorylation and thereby enhancing STAT1 pathway-mediated polarization of THP-1 macrophages to M1 and inflammatory factor expression. Taken together, these results reveal that the lnc_000048/PKR/STAT1 axis plays a crucial role in the polarization of M1 macrophages and may be a novel therapeutic target for atherosclerosis alleviation in stroke.
Collapse
Affiliation(s)
- Yuanyuan Ding
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Yu Sun
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Hongyan Wang
- Qingdao Cadre Health Care Service Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Hongqin Zhao
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Ruihua Yin
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Meng Zhang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Xudong Pan
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Xiaoyan Zhu
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| |
Collapse
|
3
|
Zhang Z, Zhang F, Xie W, Niu Y, Wang H, Li G, Zhao L, Wang X, Xie W. Induced Necroptosis and Its Role in Cancer Immunotherapy. Int J Mol Sci 2024; 25:10760. [PMID: 39409087 PMCID: PMC11477008 DOI: 10.3390/ijms251910760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 08/30/2024] [Accepted: 10/04/2024] [Indexed: 10/20/2024] Open
Abstract
Necroptosis is a type of regulated cell death (RCD) that is triggered by changes in the extracellular or intracellular milieu that are picked up by certain death receptors. Thanks to its potent capacity to induce immunological responses and overcome apoptotic resistance, it has garnered significant attention as a potential cancer treatment. Basic information for the creation of nano-biomedical treatments is provided by studies on the mechanisms underlying tumor necroptosis. Receptor-interacting protein kinase 1 (RIPK1)-RIPK3-mediated necroptosis, Toll-like receptor domain-containing adapter-inducing interferon (IFN)-β (TRIF)-RIPK3-mediated necroptosis, Z-DNA-binding protein 1 (ZBP1)-RIPK3-mediated necroptosis, and IFNR-mediated necroptosis are the four signaling pathways that collectively account for triggered necroptosis in this review. Necroptosis has garnered significant interest as a possible cancer treatment strategy because, in contrast to apoptosis, it elicits immunological responses that are relevant to therapy. Thus, a thorough discussion is held on the connections between tumor cell necroptosis and the immune environment, cancer immunosurveillance, and cells such as dendritic cells (DCs), cytotoxic T cells, natural killer (NK) cells, natural killer T (NKT) cells, and their respective cytokines. Lastly, a summary of the most recent nanomedicines that cause necroptosis in order to cause immunogenic cell death is provided in order to emphasize their promise for cancer immunotherapy.
Collapse
Affiliation(s)
- Ziyao Zhang
- The Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; (Z.Z.); (F.Z.); (Y.N.); (H.W.); (G.L.)
| | - Fangming Zhang
- The Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; (Z.Z.); (F.Z.); (Y.N.); (H.W.); (G.L.)
| | - Wenjing Xie
- School of Chemistry and Chemical Engineering, Guizhou University, Guiyang 550025, China;
| | - Yubo Niu
- The Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; (Z.Z.); (F.Z.); (Y.N.); (H.W.); (G.L.)
| | - Haonan Wang
- The Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; (Z.Z.); (F.Z.); (Y.N.); (H.W.); (G.L.)
| | - Guofeng Li
- The Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; (Z.Z.); (F.Z.); (Y.N.); (H.W.); (G.L.)
| | - Lingyun Zhao
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China;
| | - Xing Wang
- The Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; (Z.Z.); (F.Z.); (Y.N.); (H.W.); (G.L.)
| | - Wensheng Xie
- The Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; (Z.Z.); (F.Z.); (Y.N.); (H.W.); (G.L.)
| |
Collapse
|
4
|
Zhu T, Wu BW. Recognition of necroptosis: From molecular mechanisms to detection methods. Biomed Pharmacother 2024; 178:117196. [PMID: 39053418 DOI: 10.1016/j.biopha.2024.117196] [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: 05/11/2024] [Revised: 07/05/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024] Open
Abstract
Necroptosis is a crucial modality of programmed cell death characterized by distinct morphological and biochemical hallmarks, including cell membrane rupture, organelle swelling, cytoplasmic and nuclear disintegration, cellular contents leakage, and release of damage-associated molecular patterns (DAMPs), accompanied by the inflammatory responses. Studies have shown that necroptosis is involved in the etiology and evolution of a variety of pathologies including organ damage, inflammation disorders, and cancer. Despite its significance, the field of necroptosis research grapples with the challenge of non-standardized detection methodologies. In this review, we introduce the fundamental concepts and molecular mechanisms of necroptosis and critically appraise the principles, merits, and inherent limitations of current detection technologies. This endeavor seeks to establish a methodological framework for necroptosis detection, thereby propelling deeper insights into the research of cell necroptosis.
Collapse
Affiliation(s)
- Ting Zhu
- Department of pharmacy, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441000, China
| | - Bo-Wen Wu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
5
|
Cao X, Tan J, Zheng R, Wang F, Zhou L, Yi J, Yuan R, Dai Q, Song L, Dai A. Targeting necroptosis: a promising avenue for respiratory disease treatment. Cell Commun Signal 2024; 22:418. [PMID: 39192326 DOI: 10.1186/s12964-024-01804-6] [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: 06/20/2024] [Accepted: 08/22/2024] [Indexed: 08/29/2024] Open
Abstract
Respiratory diseases are a growing concern in public health because of their potential to endanger the global community. Cell death contributes critically to the pathophysiology of respiratory diseases. Recent evidence indicates that necroptosis, a unique form of programmed cell death (PCD), plays a vital role in the molecular mechanisms underlying respiratory diseases, distinguishing it from apoptosis and conventional necrosis. Necroptosis is a type of inflammatory cell death governed by receptor-interacting serine/threonine protein kinase 1 (RIPK1), RIPK3, and mixed-lineage kinase domain-like protein (MLKL), resulting in the release of intracellular contents and inflammatory factors capable of initiating an inflammatory response in adjacent tissues. These necroinflammatory conditions can result in significant organ dysfunction and long-lasting tissue damage within the lungs. Despite evidence linking necroptosis to various respiratory diseases, there are currently no specific alternative treatments that target this mechanism. This review provides a comprehensive overview of the most recent advancements in understanding the significance and mechanisms of necroptosis. Specifically, this review emphasizes the intricate association between necroptosis and respiratory diseases, highlighting the potential use of necroptosis as an innovative therapeutic approach for treating these conditions.
Collapse
Affiliation(s)
- Xianya Cao
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People's Republic of China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
| | - Junlan Tan
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410021, People's Republic of China
| | - Runxiu Zheng
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People's Republic of China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
| | - Feiying Wang
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China
| | - Lingling Zhou
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China
| | - Jian Yi
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410021, People's Republic of China
| | - Rong Yuan
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China
| | - Qin Dai
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China
| | - Lan Song
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China
| | - Aiguo Dai
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China.
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China.
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410021, People's Republic of China.
| |
Collapse
|
6
|
Lecomte K, Toniolo A, Hoste E. Cell death as an architect of adult skin stem cell niches. Cell Death Differ 2024; 31:957-969. [PMID: 38649745 PMCID: PMC11303411 DOI: 10.1038/s41418-024-01297-3] [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: 12/16/2023] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 04/25/2024] Open
Abstract
Our skin provides a physical and immunological barrier against dehydration and environmental insults ranging from microbial attacks, toxins and UV irradiation to wounding. Proper functioning of the skin barrier largely depends on the interplay between keratinocytes- the epithelial cells of the skin- and immune cells. Two spatially distinct populations of keratinocyte stem cells (SCs) maintain the epidermal barrier function and the hair follicle. These SCs are inherently long-lived, but cell death can occur within their niches and impacts their functionality. The default cell death programme in skin is apoptosis, an orderly and non-inflammatory suicide programme. However, recent findings are shedding light on the significance of various modes of regulated necrotic cell death, which are lytic and can provoke inflammation within the local skin environment. While the presence of dying cells was generally regarded as a mere consequence of inflammation, findings in various human dermatological conditions and experimental mouse models of aberrant cell death control demonstrated that cell death programmes in keratinocytes (KCs) can drive skin inflammation and even tumour initiation. When cells die, they need to be removed by phagocytosis and KCs can function as non-professional phagocytes of apoptotic cells with important implications for their SC capacities. It is becoming apparent that in conditions of heightened SC activity, distinct cell death modalities differentially impact the different skin SC populations in their local niches. Here, we describe how regulated cell death modalities functionally affect epidermal SC niches along with their relevance to injury repair, inflammatory skin disorders and cancer.
Collapse
Affiliation(s)
- Kim Lecomte
- VIB Center for Inflammation Research, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
| | - Annagiada Toniolo
- VIB Center for Inflammation Research, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
| | - Esther Hoste
- VIB Center for Inflammation Research, 9052, Ghent, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium.
| |
Collapse
|
7
|
Asai K, Lee HK, Sato S, Shimizu E, Jung J, Okazaki T, Ogawa M, Shimmura S, Tsubota K, Ogawa Y, Negishi K, Hirayama M. The Necroptosis Pathway Is Upregulated in the Cornea in Mice With Ocular Graft-Versus-Host Disease. Invest Ophthalmol Vis Sci 2024; 65:38. [PMID: 39189995 PMCID: PMC11361379 DOI: 10.1167/iovs.65.10.38] [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: 04/29/2024] [Accepted: 07/26/2024] [Indexed: 08/28/2024] Open
Abstract
Purpose To identify molecular signatures specific for ocular graft-versus-host disease (GVHD) by proteomic analysis of corneas from mice with GVHD. Methods We identified differentially expressed proteins (DEPs) in corneal samples from GVHD model mice and syngeneic control mice 4 weeks after bone marrow transplantation. Data-independent acquisition analysis was performed on individual samples, and the roles of DEPs in biological pathways related to GVHD were evaluated via bioinformatics and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. Results Three important signaling pathways were upregulated in the cornea in mice with GVHD: (1) the necroptosis pathway, (2) the mitogen-activated protein kinase (MAPK) pathway, and (3) as previously reported, the neutrophil extracellular trap (NET) pathway. In those signaling pathways, we identified new upregulated molecules, including (1) receptor-interacting protein kinase 1 (RIPK1), RIPK3, interferon regulatory factor 9, the interferon-induced double-stranded RNA-activated protein kinase lipoxygenase, and high mobility group box1 (HMGB1) which are damage-associated molecular patterns (DAMPs) in the necroptosis pathway; (2) the sequentially upregulated interleukin 1 (IL-1) receptor-associated kinase (IRAK), an evolutionarily conserved signaling intermediate in the Toll pathway (ECSIT), and p38, which is downstream of the IL-1 receptor and increased CDC42/Rac (Rac2), a Rho family GTPase in the MAPK pathway; and (3) the integrin components CR3 and macrophage-1 antigen (MAC-1), which are DAMPs, and the pyroptosis-related protein gasdermin D (GSDMD) in the NET pathway. Conclusions These novel molecules may help researchers elucidate the pathogenesis of GVHD and identify new therapeutic targets for corneal changes in patients with ocular GVHD.
Collapse
Affiliation(s)
- Kazuki Asai
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Hyung Keun Lee
- Ophthalmology, Yonsei University College of Medicine, Seoul, South Korea
| | - Shinri Sato
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Eisuke Shimizu
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Jaehun Jung
- Ophthalmology, Yonsei University College of Medicine, Seoul, South Korea
| | - Takahiro Okazaki
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Mamoru Ogawa
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Shigeto Shimmura
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Department of Clinical Regenerative Medicine, Fujita Health University, Tokyo, Japan
| | - Kazuo Tsubota
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Tsubota Laboratory, Inc., Tokyo, Japan
| | - Yoko Ogawa
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Masatoshi Hirayama
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
8
|
Yu X, Yuan J, Shi L, Dai S, Yue L, Yan M. Necroptosis in bacterial infections. Front Immunol 2024; 15:1394857. [PMID: 38933265 PMCID: PMC11199740 DOI: 10.3389/fimmu.2024.1394857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
Necroptosis, a recently discovered form of cell-programmed death that is distinct from apoptosis, has been confirmed to play a significant role in the pathogenesis of bacterial infections in various animal models. Necroptosis is advantageous to the host, but in some cases, it can be detrimental. To understand the impact of necroptosis on the pathogenesis of bacterial infections, we described the roles and molecular mechanisms of necroptosis caused by different bacterial infections in this review.
Collapse
Affiliation(s)
- Xing Yu
- Department of Pathogen Biology and Immunology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Jin Yuan
- Clinical Laboratory, Puer Hospital of Traditional Chinese Medicine, Puer, China
| | - Linxi Shi
- Department of Pathogen Biology and Immunology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Shuying Dai
- Department of Pathogen Biology and Immunology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Lei Yue
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Min Yan
- Department of Pathogen Biology and Immunology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, China
| |
Collapse
|
9
|
Zhou Y, Huang X, Jin Y, Qiu M, Ambe PC, Basharat Z, Hong W. The role of mitochondrial damage-associated molecular patterns in acute pancreatitis. Biomed Pharmacother 2024; 175:116690. [PMID: 38718519 DOI: 10.1016/j.biopha.2024.116690] [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: 02/08/2024] [Revised: 04/27/2024] [Accepted: 04/29/2024] [Indexed: 06/03/2024] Open
Abstract
Acute pancreatitis (AP) is one of the most common gastrointestinal tract diseases with significant morbidity and mortality. Current treatments remain unspecific and supportive due to the severity and clinical course of AP, which can fluctuate rapidly and unpredictably. Mitochondria, cellular power plant to produce energy, are involved in a variety of physiological or pathological activities in human body. There is a growing evidence indicating that mitochondria damage-associated molecular patterns (mtDAMPs) play an important role in pathogenesis and progression of AP. With the pro-inflammatory properties, released mtDAMPs may damage pancreatic cells by binding with receptors, activating downstream molecules and releasing inflammatory factors. This review focuses on the possible interaction between AP and mtDAMPs, which include cytochrome c (Cyt c), mitochondrial transcription factor A (TFAM), mitochondrial DNA (mtDNA), cardiolipin (CL), adenosine triphosphate (ATP) and succinate, with focus on experimental research and potential therapeutic targets in clinical practice. Preventing or diminishing the release of mtDAMPs or targeting the mtDAMPs receptors might have a role in AP progression.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Gastroenterology and Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China; School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Xiaoyi Huang
- Department of Gastroenterology and Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China; School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Yinglu Jin
- Department of Gastroenterology and Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China; School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Minhao Qiu
- Department of Gastroenterology and Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Peter C Ambe
- Department of General Surgery, Visceral Surgery and Coloproctology, Vinzenz-Pallotti-Hospital Bensberg, Vinzenz-Pallotti-Str. 20-24, Bensberg 51429, Germany
| | | | - Wandong Hong
- Department of Gastroenterology and Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
10
|
Meier P, Legrand AJ, Adam D, Silke J. Immunogenic cell death in cancer: targeting necroptosis to induce antitumour immunity. Nat Rev Cancer 2024; 24:299-315. [PMID: 38454135 DOI: 10.1038/s41568-024-00674-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/26/2024] [Indexed: 03/09/2024]
Abstract
Most metastatic cancers remain incurable due to the emergence of apoptosis-resistant clones, fuelled by intratumour heterogeneity and tumour evolution. To improve treatment, therapies should not only kill cancer cells but also activate the immune system against the tumour to eliminate any residual cancer cells that survive treatment. While current cancer therapies rely heavily on apoptosis - a largely immunologically silent form of cell death - there is growing interest in harnessing immunogenic forms of cell death such as necroptosis. Unlike apoptosis, necroptosis generates second messengers that act on immune cells in the tumour microenvironment, alerting them of danger. This lytic form of cell death optimizes the provision of antigens and adjuvanticity for immune cells, potentially boosting anticancer treatment approaches by combining cellular suicide and immune response approaches. In this Review, we discuss the mechanisms of necroptosis and how it activates antigen-presenting cells, drives cross-priming of CD8+ T cells and induces antitumour immune responses. We also examine the opportunities and potential drawbacks of such strategies for exposing cancer cells to immunological attacks.
Collapse
Affiliation(s)
- Pascal Meier
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, London, UK.
| | - Arnaud J Legrand
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, London, UK
| | - Dieter Adam
- Institut für Immunologie, Christian-Albrechts-Universität zu Kiel, Kiel, Germany.
| | - John Silke
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia.
| |
Collapse
|
11
|
Ciftci YC, Vatansever İE, Akgül B. Unraveling the intriguing interplay: Exploring the role of lncRNAs in caspase-independent cell death. WILEY INTERDISCIPLINARY REVIEWS. RNA 2024; 15:e1862. [PMID: 38837618 DOI: 10.1002/wrna.1862] [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: 11/05/2023] [Revised: 05/12/2024] [Accepted: 05/13/2024] [Indexed: 06/07/2024]
Abstract
Cell death plays a crucial role in various physiological and pathological processes. Until recently, programmed cell death was mainly attributed to caspase-dependent apoptosis. However, emerging evidence suggests that caspase-independent cell death (CICD) mechanisms also contribute significantly to cellular demise. We and others have reported and functionally characterized numerous long noncoding RNAs (lncRNAs) that modulate caspase-dependent apoptotic pathways potentially in a pathway-dependent manner. However, the interplay between lncRNAs and CICD pathways has not been comprehensively documented. One major reason for this is that most CICD pathways have been recently discovered with some being partially characterized at the molecular level. In this review, we discuss the emerging evidence that implicates specific lncRNAs in the regulation and execution of CICD. We summarize the diverse mechanisms through which lncRNAs modulate different forms of CICD, including ferroptosis, necroptosis, cuproptosis, and others. Furthermore, we highlight the intricate regulatory networks involving lncRNAs, protein-coding genes, and signaling pathways that orchestrate CICD in health and disease. Understanding the molecular mechanisms and functional implications of lncRNAs in CICD may unravel novel therapeutic targets and diagnostic tools for various diseases, paving the way for innovative strategies in disease management and personalized medicine. This article is categorized under: RNA in Disease and Development > RNA in Disease.
Collapse
Affiliation(s)
- Yusuf Cem Ciftci
- Noncoding RNA Laboratory, Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Gülbahçeköyü, Urla, Turkey
| | - İpek Erdoğan Vatansever
- Noncoding RNA Laboratory, Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Gülbahçeköyü, Urla, Turkey
| | - Bünyamin Akgül
- Noncoding RNA Laboratory, Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Gülbahçeköyü, Urla, Turkey
| |
Collapse
|
12
|
Tang Y, Wei J, Ge X, Yu C, Lu W, Qian Y, Yang H, Fu D, Fang Y, Zhou X, Wang Z, Xiao Q, Ding K. Intratumoral injection of interferon gamma promotes the efficacy of anti-PD1 treatment in colorectal cancer. Cancer Lett 2024; 588:216798. [PMID: 38467181 DOI: 10.1016/j.canlet.2024.216798] [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: 03/20/2023] [Revised: 02/21/2024] [Accepted: 03/05/2024] [Indexed: 03/13/2024]
Abstract
Immune checkpoint inhibitors (ICIs) offer new options for the treatment of patients with solid cancers worldwide. The majority of colorectal cancers (CRC) are proficient in mismatch-repair (pMMR) genes, harboring fewer tumor antigens and are insensitive to ICIs. These tumors are often found to be immune-deserted. We hypothesized that forcing immune cell infiltration into the tumor microenvironment followed by immune ignition by PD1 blockade may initiate a positive immune cycle that can boost antitumor immunity. Bioinformatics using a public database suggested that IFNγ was a key indicator of immune status and prognosis in CRC. Intratumoral administration of IFNγ increased immune cells infiltration into the tumor, but induced PD-L1 expression. A combined treatment strategy using IFNγ and anti-PD-1 antibody significantly increased T cell killing of tumor cells in vitro and showed synergistic inhibition of tumor growth in a mouse model of CRC. CyTOF found drastic changes in the immune microenvironment upon combined immunotherapy. Treatment with IFNγ and anti-PD1 antibody in CT26 tumors significantly increased infiltration of polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs). IFNγ had a more pronounced effect in decreasing intratumoral M2-like macrophages, while PD1 blockade increased the population of CD8+Ly6C + T cells in the tumor microenvironment, creating a more pro-inflammatory microenvironment. Additionally, PD1 induced increased expression of lymphocyte activating 3 (LAG3) in a significant fraction of CD8+ T cells and Treg cells, indicating potential drug resistance and feedback mechanisms. In conclusion, our work provides preclinical data for the Combined immunotherapy of CRC using intratumoral delivery of IFNγ and systemic anti-PD1 monoclonoal antibody.
Collapse
Affiliation(s)
- Yang Tang
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for CANCER, China; Cancer Center of Zhejiang University, Hangzhou, Zhejiang, 310058, China; Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, China
| | - Jingsun Wei
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for CANCER, China; Cancer Center of Zhejiang University, Hangzhou, Zhejiang, 310058, China; Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, China
| | - Xiaoxu Ge
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for CANCER, China; Cancer Center of Zhejiang University, Hangzhou, Zhejiang, 310058, China; Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, China
| | - Chengxuan Yu
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for CANCER, China; Cancer Center of Zhejiang University, Hangzhou, Zhejiang, 310058, China; Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, China
| | - Wei Lu
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for CANCER, China; Cancer Center of Zhejiang University, Hangzhou, Zhejiang, 310058, China; Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, China
| | - Yucheng Qian
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for CANCER, China; Cancer Center of Zhejiang University, Hangzhou, Zhejiang, 310058, China; Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, China
| | - Hang Yang
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for CANCER, China; Cancer Center of Zhejiang University, Hangzhou, Zhejiang, 310058, China; Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, China
| | - Dongliang Fu
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for CANCER, China; Cancer Center of Zhejiang University, Hangzhou, Zhejiang, 310058, China; Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, China
| | - Yimin Fang
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for CANCER, China; Cancer Center of Zhejiang University, Hangzhou, Zhejiang, 310058, China; Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, China
| | - Xinyi Zhou
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for CANCER, China; Cancer Center of Zhejiang University, Hangzhou, Zhejiang, 310058, China; Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, China
| | - Zhanhuai Wang
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for CANCER, China; Cancer Center of Zhejiang University, Hangzhou, Zhejiang, 310058, China; Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, China
| | - Qian Xiao
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for CANCER, China; Cancer Center of Zhejiang University, Hangzhou, Zhejiang, 310058, China; Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, China
| | - Kefeng Ding
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for CANCER, China; Cancer Center of Zhejiang University, Hangzhou, Zhejiang, 310058, China; Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, China.
| |
Collapse
|
13
|
Zou Y, Kamoi K, Zong Y, Zhang J, Yang M, Ohno-Matsui K. Vaccines and the Eye: Current Understanding of the Molecular and Immunological Effects of Vaccination on the Eye. Int J Mol Sci 2024; 25:4755. [PMID: 38731972 PMCID: PMC11084287 DOI: 10.3390/ijms25094755] [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: 03/23/2024] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Vaccination is a public health cornerstone that protects against numerous infectious diseases. Despite its benefits, immunization implications on ocular health warrant thorough investigation, particularly in the context of vaccine-induced ocular inflammation. This review aimed to elucidate the complex interplay between vaccination and the eye, focusing on the molecular and immunological pathways implicated in vaccine-associated ocular adverse effects. Through an in-depth analysis of recent advancements and the existing literature, we explored various mechanisms of vaccine-induced ocular inflammation, such as direct infection by live attenuated vaccines, immune complex formation, adjuvant-induced autoimmunity, molecular mimicry, hypersensitivity reactions, PEG-induced allergic reactions, Type 1 IFN activation, free extracellular RNA, and specific components. We further examined the specific ocular conditions associated with vaccination, such as uveitis, optic neuritis, and retinitis, and discussed the potential impact of novel vaccines, including those against SARS-CoV-2. This review sheds light on the intricate relationships between vaccination, the immune system, and ocular tissues, offering insights into informed discussions and future research directions aimed at optimizing vaccine safety and ophthalmological care. Our analysis underscores the importance of vigilance and further research to understand and mitigate the ocular side effects of vaccines, thereby ensuring the continued success of vaccination programs, while preserving ocular health.
Collapse
Affiliation(s)
| | - Koju Kamoi
- Department of Ophthalmology and Visual Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan; (Y.Z.); (Y.Z.); (J.Z.); (M.Y.); (K.O.-M.)
| | | | | | | | | |
Collapse
|
14
|
Divandari M, Javadifar A, Moghadam AB, Janatabadi AA. RIPK3 and RIPK1 gene expression in pterygium: unveiling molecular insights into pathogenesis. Mol Biol Rep 2024; 51:524. [PMID: 38630344 DOI: 10.1007/s11033-024-09368-x] [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: 01/05/2024] [Accepted: 02/20/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Pterygium, characterized by the abnormal proliferation of epithelial cells, matrix remodeling, vascularization, and lesion migration, is a prevalent ocular surface disease involving the growth of fibrovascular tissue on the cornea. Despite the unclear underlying causes of pterygium, numerous investigations have indicated the involvement of cell death pathways in the regulation of cell cycle dynamics. Consequently, the objective of this study was to assess the expression levels of necroptosis markers in individuals diagnosed with pterygium, aiming to shed light on the potential role of necroptosis in the pathogenesis of this condition. METHODS This study aimed to investigate the expression patterns of receptor-interacting serine/threonine kinase 3 (RIPK3) and receptor-interacting serine/threonine kinase 1 (RIPK1) genes in pterygium tissues. 41 patients undergoing pterygium excision surgery were recruited. Resected pterygium samples and normal conjunctival tissues were collected, and RIPK3 and RIPK1 mRNA levels were measured using quantitative real-time PCR. RESULTS Our findings reveal that the expression of RIPK3 is significantly increased in samples obtained from individuals with pterygium. However, no significant alterations were observed in the expression of RIPK1 in these samples. Results showed significantly higher RIPK3 expression in pterygium tissues compared to controls. Moreover, increased RIPK3 levels correlated negatively with pterygium recurrence rates. CONCLUSIONS These findings suggest RIPK3 may play a protective role against pterygium recurrence through necroptosis.
Collapse
Affiliation(s)
- Mahnaz Divandari
- Department of Biology, Sabzevar Branch, Islamic Azad University, Sabzevar, Iran
| | - Amin Javadifar
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of medical sciences, Mashhad, Iran
| | | | - Ali Akbar Janatabadi
- Department of Biology, Sabzevar Branch, Islamic Azad University, Sabzevar, Iran.
| |
Collapse
|
15
|
Nurmi K, Silventoinen K, Keskitalo S, Rajamäki K, Kouri VP, Kinnunen M, Jalil S, Maldonado R, Wartiovaara K, Nievas EI, Denita-Juárez SP, Duncan CJA, Kuismin O, Saarela J, Romo I, Martelius T, Parantainen J, Beklen A, Bilicka M, Matikainen S, Nordström DC, Kaustio M, Wartiovaara-Kautto U, Kilpivaara O, Klein C, Hauck F, Jahkola T, Hautala T, Varjosalo M, Barreto G, Seppänen MRJ, Eklund KK. Truncating NFKB1 variants cause combined NLRP3 inflammasome activation and type I interferon signaling and predispose to necrotizing fasciitis. Cell Rep Med 2024; 5:101503. [PMID: 38593810 PMCID: PMC11031424 DOI: 10.1016/j.xcrm.2024.101503] [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: 12/14/2022] [Revised: 01/04/2024] [Accepted: 03/18/2024] [Indexed: 04/11/2024]
Abstract
In monogenic autoinflammatory diseases, mutations in genes regulating innate immune responses often lead to uncontrolled activation of inflammasome pathways or the type I interferon (IFN-I) response. We describe a mechanism of autoinflammation potentially predisposing patients to life-threatening necrotizing soft tissue inflammation. Six unrelated families are identified in which affected members present with necrotizing fasciitis or severe soft tissue inflammations. Exome sequencing reveals truncating monoallelic loss-of-function variants of nuclear factor κ light-chain enhancer of activated B cells (NFKB1) in affected patients. In patients' macrophages and in NFKB1-variant-bearing THP-1 cells, activation increases both interleukin (IL)-1β secretion and IFN-I signaling. Truncation of NF-κB1 impairs autophagy, accompanied by the accumulation of reactive oxygen species and reduced degradation of inflammasome receptor nucleotide-binding oligomerization domain, leucine-rich repeat-containing protein 3 (NLRP3), and Toll/IL-1 receptor domain-containing adaptor protein inducing IFN-β (TRIF), thus leading to combined excessive inflammasome and IFN-I activity. Many of the patients respond to anti-inflammatory treatment, and targeting IL-1β and/or IFN-I signaling could represent a therapeutic approach for these patients.
Collapse
Affiliation(s)
- Katariina Nurmi
- Faculty of Medicine, Clinicum, Translational Immunology Research Program, Research Program Unit (RPU), University of Helsinki (UH), 00014 Helsinki, Finland
| | - Kristiina Silventoinen
- Faculty of Medicine, Clinicum, Translational Immunology Research Program, Research Program Unit (RPU), University of Helsinki (UH), 00014 Helsinki, Finland
| | - Salla Keskitalo
- Systems Biology/Pathology Research Group, iCAN Digital Precision Cancer Medicine Flagship, Institute of Biotechnology, HiLIFE, UH, 00014 Helsinki, Finland
| | - Kristiina Rajamäki
- Faculty of Medicine, Clinicum, Translational Immunology Research Program, Research Program Unit (RPU), University of Helsinki (UH), 00014 Helsinki, Finland; Department of Medical and Clinical Genetics, Applied Tumor Genomics Research Program, RPU, UH, 00014 Helsinki, Finland
| | - Vesa-Petteri Kouri
- Faculty of Medicine, Clinicum, Translational Immunology Research Program, Research Program Unit (RPU), University of Helsinki (UH), 00014 Helsinki, Finland
| | - Matias Kinnunen
- Systems Biology/Pathology Research Group, iCAN Digital Precision Cancer Medicine Flagship, Institute of Biotechnology, HiLIFE, UH, 00014 Helsinki, Finland
| | - Sami Jalil
- Clinical Genetics UH and Helsinki University Hospital (HUH), 00014 Helsinki, Finland
| | - Rocio Maldonado
- Clinical Genetics UH and Helsinki University Hospital (HUH), 00014 Helsinki, Finland
| | - Kirmo Wartiovaara
- Clinical Genetics UH and Helsinki University Hospital (HUH), 00014 Helsinki, Finland
| | | | | | - Christopher J A Duncan
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE1 4HH, UK
| | - Outi Kuismin
- Department of Clinical Genetics, Oulu University Hospital (OUH), 90014 Oulu, Finland; PEDEGO Research Unit and Medical Research Center Oulu, OUH and University of Oulu (OU), 90014 Oulu, Finland
| | - Janna Saarela
- Institute for Molecular Medicine Finland, HiLIFE, UH, 00014 Helsinki, Finland; Centre for Molecular Medicine Norway, University of Oslo, 0313 Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, 0450 Oslo, Norway
| | - Inka Romo
- Inflammation Center, Department of Infectious Disease, HUH, 00029 Helsinki, Finland
| | - Timi Martelius
- Inflammation Center, Department of Infectious Disease, HUH, 00029 Helsinki, Finland
| | - Jukka Parantainen
- Faculty of Medicine, Clinicum, Translational Immunology Research Program, Research Program Unit (RPU), University of Helsinki (UH), 00014 Helsinki, Finland
| | - Arzu Beklen
- Faculty of Medicine, Clinicum, Translational Immunology Research Program, Research Program Unit (RPU), University of Helsinki (UH), 00014 Helsinki, Finland
| | - Marcelina Bilicka
- Faculty of Medicine, Clinicum, Translational Immunology Research Program, Research Program Unit (RPU), University of Helsinki (UH), 00014 Helsinki, Finland
| | - Sampsa Matikainen
- Faculty of Medicine, Clinicum, Translational Immunology Research Program, Research Program Unit (RPU), University of Helsinki (UH), 00014 Helsinki, Finland
| | - Dan C Nordström
- Faculty of Medicine, Clinicum, Translational Immunology Research Program, Research Program Unit (RPU), University of Helsinki (UH), 00014 Helsinki, Finland; Department of Internal Medicine and Rehabilitation, HUH and UH, 00029 Helsinki, Finland
| | - Meri Kaustio
- Institute for Molecular Medicine Finland, HiLIFE, UH, 00014 Helsinki, Finland
| | - Ulla Wartiovaara-Kautto
- Department of Hematology, HUH, Comprehensive Cancer Center, UH, 00029 Helsinki, Finland; Applied Tumor Genomics Research Program, RPU, Faculty of Medicine, UH, 00014 Helsinki, Finland
| | - Outi Kilpivaara
- Applied Tumor Genomics Research Program, RPU, Faculty of Medicine, UH, 00014 Helsinki, Finland; Department of Medical and Clinical Genetics/Medicum, Faculty of Medicine, UH, 00014 Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, UH, 00014 Helsinki, Finland; HUS Diagnostic Center, HUSLAB Laboratory of Genetics, HUH, 00029 Helsinki, Finland
| | - Christoph Klein
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, 80337 Munich, Germany
| | - Fabian Hauck
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, 80337 Munich, Germany
| | - Tiina Jahkola
- Department of Plastic Surgery, HUH, 00029 Helsinki, Finland
| | - Timo Hautala
- Research Unit of Internal Medicine and Biomedicine, OU, and Infectious Diseases Clinic, OUH, 90014 Oulu, Finland
| | - Markku Varjosalo
- Systems Biology/Pathology Research Group, iCAN Digital Precision Cancer Medicine Flagship, Institute of Biotechnology, HiLIFE, UH, 00014 Helsinki, Finland
| | - Goncalo Barreto
- Faculty of Medicine, Clinicum, Translational Immunology Research Program, Research Program Unit (RPU), University of Helsinki (UH), 00014 Helsinki, Finland
| | - Mikko R J Seppänen
- Adult Immunodeficiency Unit, Infectious Diseases, Inflammation Center, HUH and UH, 00029 Helsinki, Finland; Rare Disease Center, Children and Adolescents, HUH and UH, 00029 Helsinki, Finland.
| | - Kari K Eklund
- Faculty of Medicine, Clinicum, Translational Immunology Research Program, Research Program Unit (RPU), University of Helsinki (UH), 00014 Helsinki, Finland; Department of Rheumatology, HUH and UH, 00029 Helsinki, Finland; Orton Orthopaedic Hospital, 00280 Helsinki, Finland.
| |
Collapse
|
16
|
Witwit H, Khafaji R, Salaniwal A, Kim AS, Cubitt B, Jackson N, Ye C, Weiss SR, Martinez-Sobrido L, de la Torre JC. Activation of protein kinase receptor (PKR) plays a pro-viral role in mammarenavirus-infected cells. J Virol 2024; 98:e0188323. [PMID: 38376197 PMCID: PMC10949842 DOI: 10.1128/jvi.01883-23] [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: 12/04/2023] [Accepted: 01/26/2024] [Indexed: 02/21/2024] Open
Abstract
Many viruses, including mammarenaviruses, have evolved mechanisms to counteract different components of the host cell innate immunity, which is required to facilitate robust virus multiplication. The double-stranded RNA (dsRNA) sensor protein kinase receptor (PKR) pathway plays a critical role in the cell anti-viral response. Whether PKR can restrict the multiplication of the Old World mammarenavirus lymphocytic choriomeningitis virus (LCMV) and the mechanisms by which LCMV may counteract the anti-viral functions of PKR have not yet been investigated. Here we present evidence that LCMV infection results in very limited levels of PKR activation, but LCMV multiplication is enhanced in the absence of PKR. In contrast, infection with a recombinant LCMV with a mutation affecting the 3'-5' exonuclease (ExoN) activity of the viral nucleoprotein resulted in robust PKR activation in the absence of detectable levels of dsRNA, which was associated with severely restricted virus multiplication that was alleviated in the absence of PKR. However, pharmacological inhibition of PKR activation resulted in reduced levels of LCMV multiplication. These findings uncovered a complex role of the PKR pathway in LCMV-infected cells involving both pro- and anti-viral activities.IMPORTANCEAs with many other viruses, the prototypic Old World mammarenavirus LCMV can interfere with the host cell innate immune response to infection, which includes the dsRNA sensor PKR pathway. A detailed understanding of LCMV-PKR interactions can provide novel insights about mammarenavirus-host cell interactions and facilitate the development of effective anti-viral strategies against human pathogenic mammarenaviruses. In the present work, we present evidence that LCMV multiplication is enhanced in PKR-deficient cells, but pharmacological inhibition of PKR activation unexpectedly resulted in severely restricted propagation of LCMV. Likewise, we document a robust PKR activation in LCMV-infected cells in the absence of detectable levels of dsRNA. Our findings have revealed a complex role of the PKR pathway during LCMV infection and uncovered the activation of PKR as a druggable target for the development of anti-viral drugs against human pathogenic mammarenaviruses.
Collapse
Affiliation(s)
- Haydar Witwit
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Roaa Khafaji
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Arul Salaniwal
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Arthur S. Kim
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
- Department of Chemistry, The Scripps Research Institute, La Jolla, California, USA
| | - Beatrice Cubitt
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | | | - Chengjin Ye
- Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Susan R. Weiss
- Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Juan Carlos de la Torre
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| |
Collapse
|
17
|
Wu X, Nagy LE, Gautheron J. Mediators of necroptosis: from cell death to metabolic regulation. EMBO Mol Med 2024; 16:219-237. [PMID: 38195700 PMCID: PMC10897313 DOI: 10.1038/s44321-023-00011-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: 09/21/2023] [Revised: 11/08/2023] [Accepted: 11/20/2023] [Indexed: 01/11/2024] Open
Abstract
Necroptosis, a programmed cell death mechanism distinct from apoptosis, has garnered attention for its role in various pathological conditions. While initially recognized for its involvement in cell death, recent research has revealed that key necroptotic mediators, including receptor-interacting protein kinases (RIPKs) and mixed lineage kinase domain-like protein (MLKL), possess additional functions that go beyond inducing cell demise. These functions encompass influencing critical aspects of metabolic regulation, such as energy metabolism, glucose homeostasis, and lipid metabolism. Dysregulated necroptosis has been implicated in metabolic diseases, including obesity, diabetes, metabolic dysfunction-associated steatotic liver disease (MASLD) and alcohol-associated liver disease (ALD), contributing to chronic inflammation and tissue damage. This review provides insight into the multifaceted role of necroptosis, encompassing both cell death and these extra-necroptotic functions, in the context of metabolic diseases. Understanding this intricate interplay is crucial for developing targeted therapeutic strategies in diseases that currently lack effective treatments.
Collapse
Affiliation(s)
- Xiaoqin Wu
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, USA
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Laura E Nagy
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, USA
- Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, OH, USA
- Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Jérémie Gautheron
- Sorbonne Université, Inserm UMRS_938, Centre de Recherche Saint-Antoine (CRSA), Paris, 75012, France.
| |
Collapse
|
18
|
Zhang S, Liu Y, Ma Z, Gao S, Chen L, Zhong H, Zhang C, Li T, Chen W, Zhang Y, Lin N. Osteoking promotes bone formation and bone defect repair through ZBP1-STAT1-PKR-MLKL-mediated necroptosis. Chin Med 2024; 19:13. [PMID: 38238785 PMCID: PMC10797925 DOI: 10.1186/s13020-024-00883-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 12/29/2023] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND Osteoking has been used for fracture therapy with a satisfying clinical efficacy. However, its therapeutic properties and the underlying mechanisms remain elusive. METHOD A bone defect rat model was established to evaluate the pharmacological effects of Osteoking by the dynamic observation of X-ray, micro-CT and histopathologic examination. Transcriptome profiling was performed to identify bone defect-related genes and Osteoking effective targets. Then, a "disease-related gene-drug target" interaction network was constructed and a list of key network targets were screened, which were experimentally verified. RESULTS Osteoking effectively promoted bone defect repair in rats by accelerating the repair of cortical bone and the growth of trabeculae. Histopathologically, the bone defect rats displayed lower histopathologic scores in cortical bone, cancellous bone and bone connection than normal controls. In contrast, Osteoking exerted a favorable effect with a dose-dependent manner. The abnormal serum levels of bone turnover markers, bone growth factors and bone metabolism-related biochemical indexes in bone defect rats were also reversed by Osteoking treatment. Following the transcriptome-based network investigation, we hypothesized that osteoking might attenuate the levels of ZBP1-STAT1-PKR-MLKL-mediated necroptosis involved into bone defect. Experimentally, the expression levels of ZBP1, STAT1, PKR and the hallmark inflammatory cytokines for the end of necroptosis were distinctly elevated in bone defect rats, but were all effectively reversed by Osteoking treatment, which were also suppressed the activities of RIPK1, RIPK3 and MLKL in bone tissue supernatants. CONCLUSIONS Osteoking may promote bone formation and bone defect repair by regulating ZBP1-STAT1-PKR axis, leading to inhibit RIPK1/RIPK3/MLKL activation-mediated necroptosis.
Collapse
Affiliation(s)
- Suya Zhang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou, 510405, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China
| | - Yudong Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China
| | - Zhaochen Ma
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China
| | - Shuangrong Gao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China
| | - Lin Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China
| | - Honggang Zhong
- BioMechanics Lab, Wang Jing Hospital, China Academy of Chinese Medical Sciences, Beijing, 100010, China
| | - Chu Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China
| | - Tao Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China
| | - Weiheng Chen
- Third Affiliated Hospital of Beijing University of Chinese Medicine, No. 51 Anwai Xiaoguanjie, Chaoyang District, Beijing, 100029, China
| | - Yanqiong Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China.
| | - Na Lin
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou, 510405, China.
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China.
| |
Collapse
|
19
|
Mohseni-Moghaddam P, Khaleghzadeh-Ahangar H, Atabaki R. Role of Necroptosis, a Regulated Cell Death, in Seizure and Epilepsy. Neurochem Res 2024; 49:1-13. [PMID: 37646959 DOI: 10.1007/s11064-023-04010-x] [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: 05/10/2023] [Revised: 07/19/2023] [Accepted: 08/04/2023] [Indexed: 09/01/2023]
Abstract
Epilepsy is a chronic neurological disease that is characterized by spontaneous and recurrent seizures. Regulated cell death is a controlled process and has been shown to be involved in neurodegenerative diseases. Necroptosis is a type of regulated cell death, and its association with epilepsy has been documented. Necroptosis signaling can be divided into two pathways: canonical and non-canonical pathways. Inhibition of caspase-8, dimerization of receptor-interacting protein kinase 1 (RIP1) and RIP3, activation of mixed-lineage kinase domain-like protein (MLKL), movement of MLKL to the plasma membrane, and cell rupture occurred in these pathways. Through literature review, it has been revealed that there is a relationship between seizure, neuroinflammation, and oxidative stress. The seizure activity triggers the activation of various pathways within the central nervous system, including TNF-α/matrix metalloproteases, Neogenin and Calpain/ Jun N-terminal Kinase 1, which result in distinct responses in the brain. These responses involve the activation of neurons and astrocytes, consequently leading to an increase in the expression levels of proteins and genes such as RIP1, RIP3, and MLKL in a time-dependent manner in regions such as the hippocampus (CA1, CA3, dentate gyrus, and hilus), piriform cortex, and amygdala. Furthermore, the imbalance in calcium ions, depletion of adenosine triphosphate, and elevation of extracellular glutamate and potassium within these pathways lead to the progression of necroptosis, a reduction in seizure threshold, and increased susceptibility to epilepsy. Therefore, it is plausible that therapeutic targeting of these pathways could potentially provide a promising approach for managing seizures and epilepsy.
Collapse
Affiliation(s)
- Parvaneh Mohseni-Moghaddam
- Department of Physiology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hossein Khaleghzadeh-Ahangar
- Department of Physiology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
- Immunoregulation Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Rabi Atabaki
- Shahid Fakouri High School, Department of Biology Education, Department of Education, Jouybar, Iran.
| |
Collapse
|
20
|
Williams J, Bonner J, Kibler K, Jacobs BL. Type I Interferon: Monkeypox/Mpox Viruses Achilles Heel? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1451:125-137. [PMID: 38801575 DOI: 10.1007/978-3-031-57165-7_8] [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] [Indexed: 05/29/2024]
Abstract
Poxviruses are notorious for having acquired/evolved numerous genes to counteract host innate immunity. Chordopoxviruses have acquired/evolved at least three different inhibitors of host necroptotic death: E3, which blocks ZBP1-dependent necroptotic cell death, and vIRD and vMLKL that inhibit necroptosis downstream of initial cell death signaling. While this suggests the importance of the necroptotic cell death pathway in inhibiting chordopoxvirus replication, several chordopoxviruses have lost one or more of these inhibitory functions. Monkeypox/mpox virus (MPXV) has lost a portion of the N-terminus of its E3 homologue. The N-terminus of the vaccinia virus E3 homologue serves to inhibit activation of the interferon-inducible antiviral protein, ZBP1. This likely makes MPXV unique among the orthopoxviruses in being sensitive to interferon (IFN) treatment in many mammals, including humans, which encode a complete necroptotic cell death pathway. Thus, IFN sensitivity may be the Achille's Heel for viruses like MPXV that cannot fully inhibit IFN-inducible, ZBP1-dependent antiviral pathways.
Collapse
Affiliation(s)
- Jacqueline Williams
- Biodesign Center for Immunotherapy, Vaccines and Virotherapy, Arizona State University, Tempe, USA
- ASU-Banner Center for Neurodegenerative Diseases, Arizona State University, Tempe, USA
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, USA
| | - James Bonner
- Biodesign Center for Immunotherapy, Vaccines and Virotherapy, Arizona State University, Tempe, USA
- ASU-Banner Center for Neurodegenerative Diseases, Arizona State University, Tempe, USA
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, USA
- School of Life Sciences, Arizona State University, Tempe, USA
| | - Karen Kibler
- Biodesign Center for Immunotherapy, Vaccines and Virotherapy, Arizona State University, Tempe, USA
- ASU-Banner Center for Neurodegenerative Diseases, Arizona State University, Tempe, USA
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, USA
| | - Bertram L Jacobs
- Biodesign Center for Immunotherapy, Vaccines and Virotherapy, Arizona State University, Tempe, USA.
- ASU-Banner Center for Neurodegenerative Diseases, Arizona State University, Tempe, USA.
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, USA.
- School of Life Sciences, Arizona State University, Tempe, USA.
| |
Collapse
|
21
|
Robinson GI, Li D, Wang B, Rahman T, Gerasymchuk M, Hudson D, Kovalchuk O, Kovalchuk I. Psilocybin and Eugenol Reduce Inflammation in Human 3D EpiIntestinal Tissue. Life (Basel) 2023; 13:2345. [PMID: 38137946 PMCID: PMC10744792 DOI: 10.3390/life13122345] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/09/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Inflammation plays a pivotal role in the development and progression of inflammatory bowel disease (IBD), by contributing to tissue damage and exacerbating the immune response. The investigation of serotonin receptor 2A (5-HT2A) ligands and transient receptor potential (TRP) channel ligands is of significant interest due to their potential to modulate key inflammatory pathways, mitigate the pathological effects of inflammation, and offer new avenues for therapeutic interventions in IBD. This study investigates the anti-inflammatory effects of 5-HT2A ligands, including psilocybin, 4-AcO-DMT, and ketanserin, in combination with TRP channel ligands, including capsaicin, curcumin, and eugenol, on the inflammatory response induced by tumor necrosis factor (TNF)-α and interferon (IFN)-γ in human 3D EpiIntestinal tissue. Enzyme-linked immunosorbent assay was used to assess the expression of pro-inflammatory markers TNF-α, IFN-γ, IL-6, IL-8, MCP-1, and GM-CSF. Our results show that psilocybin, 4-AcO-DMT, and eugenol significantly reduce TNF-α and IFN-γ levels, while capsaicin and curcumin decrease these markers to a lesser extent. Psilocybin effectively lowers IL-6 and IL-8 levels, but curcumin, capsaicin, and 4-AcO-DMT have limited effects on these markers. In addition, psilocybin can significantly decrease MCP-1 and GM-CSF levels. While ketanserin lowers IL-6 and GM-CSF levels, there are no effects seen on TNF-α, IFN-γ, IL-8, or MCP-1. Although synergistic effects between 5-HT2A and TRP channel ligands are minimal in this study, the results provide further evidence of the anti-inflammatory effects of psilocybin and eugenol. Further research is needed to understand the mechanisms of action and the feasibility of using these compounds as anti-inflammatory therapies for conditions like IBD.
Collapse
Affiliation(s)
- Gregory Ian Robinson
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Dongping Li
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Bo Wang
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Tahiat Rahman
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Marta Gerasymchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Darryl Hudson
- GoodCap Pharmaceuticals, 520 3rd Avenue SW, Suite 1900, Calgary, AB T2P 0R3, Canada
| | - Olga Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Igor Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| |
Collapse
|
22
|
Witwit H, Khafaji R, Salaniwal A, Kim AS, Cubitt B, Jackson N, Ye C, Weiss SR, Martinez-Sobrido L, de la Torre JC. Activation of Protein Kinase R (PKR) Plays a Pro-Viral Role in Mammarenavirus Infected Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.05.570143. [PMID: 38106082 PMCID: PMC10723269 DOI: 10.1101/2023.12.05.570143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Many viruses, including mammarenaviruses, have evolved mechanisms to counteract different components of the host cell innate immunity, which is required to facilitate robust virus multiplication. The double strand (ds)RNA sensor protein kinase receptor (PKR) pathway plays a critical role in the cell antiviral response. Whether PKR can restrict the multiplication of the Old World mammarenavirus lymphocytic choriomeningitis virus (LCMV) and the mechanisms by which LCMV may counteract the antiviral functions of PKR have not yet been investigated. Here we present evidence that LCMV infection results in very limited levels of PKR activation, but LCMV multiplication is enhanced in the absence of PKR. In contrast, infection with a recombinant LCMV with a mutation affecting the 3'-5' exonuclease (ExoN) activity of the viral nucleoprotein (NP) resulted in robust PKR activation in the absence of detectable levels of dsRNA, which was associated with severely restricted virus multiplication that was alleviated in the absence of PKR. However, pharmacological inhibition of PKR activation resulted in reduced levels of LCMV multiplication. These findings uncovered a complex role of the PKR pathway in LCMV-infected cells involving both pro-and antiviral activities.
Collapse
Affiliation(s)
- Haydar Witwit
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037
| | - Roaa Khafaji
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037
| | - Arul Salaniwal
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037
| | - Arthur S. Kim
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037
| | - Beatrice Cubitt
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037
| | | | - Chengjin Ye
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Susan R Weiss
- Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
| | | | - Juan Carlos de la Torre
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037
| |
Collapse
|
23
|
Hassanein EHM, Ibrahim IM, Abd El-Maksoud MS, Abd El-Aziz MK, Abd-Alhameed EK, Althagafy HS. Targeting necroptosis in fibrosis. Mol Biol Rep 2023; 50:10471-10484. [PMID: 37910384 PMCID: PMC10676318 DOI: 10.1007/s11033-023-08857-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 09/27/2023] [Indexed: 11/03/2023]
Abstract
Necroptosis, a type of programmed cell death that resembles necrosis, is now known to depend on a different molecular mechanism from apoptosis, according to several recent studies. Many efforts have reported the possible influence of necroptosis in human disorders and concluded the crucial role in the pathophysiology of various diseases, including liver diseases, renal injuries, cancers, and others. Fibrosis is the most common end-stage pathological cascade of several chronic inflammatory disorders. In this review, we explain the impact of necroptosis and fibrosis, for which necroptosis has been demonstrated to be a contributing factor. We also go over the inhibitors of necroptosis and how they have been applied to fibrosis models. This review helps to clarify the role of necroptosis in fibrosis and will encourage clinical efforts to target this pathway of programmed cell death.
Collapse
Affiliation(s)
- Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt.
| | - Islam M Ibrahim
- Graduated Student, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Mostafa S Abd El-Maksoud
- Graduated Student, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Mostafa K Abd El-Aziz
- Graduated Student, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Esraa K Abd-Alhameed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Hanan S Althagafy
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| |
Collapse
|
24
|
Zhang W, Jiang H, Wu G, Huang P, Wang H, An H, Liu S, Zhang W. The pathogenesis and potential therapeutic targets in sepsis. MedComm (Beijing) 2023; 4:e418. [PMID: 38020710 PMCID: PMC10661353 DOI: 10.1002/mco2.418] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 10/01/2023] [Accepted: 10/12/2023] [Indexed: 12/01/2023] Open
Abstract
Sepsis is defined as "a life-threatening organ dysfunction caused by dysregulated host systemic inflammatory and immune response to infection." At present, sepsis continues to pose a grave healthcare concern worldwide. Despite the use of supportive measures in treating traditional sepsis, such as intravenous fluids, vasoactive substances, and oxygen plus antibiotics to eradicate harmful pathogens, there is an ongoing increase in both the morbidity and mortality associated with sepsis during clinical interventions. Therefore, it is urgent to design specific pharmacologic agents for the treatment of sepsis and convert them into a novel targeted treatment strategy. Herein, we provide an overview of the molecular mechanisms that may be involved in sepsis, such as the inflammatory response, immune dysfunction, complement deactivation, mitochondrial damage, and endoplasmic reticulum stress. Additionally, we highlight important targets involved in sepsis-related regulatory mechanisms, including GSDMD, HMGB1, STING, and SQSTM1, among others. We summarize the latest advancements in potential therapeutic drugs that specifically target these signaling pathways and paramount targets, covering both preclinical studies and clinical trials. In addition, this review provides a detailed description of the crosstalk and function between signaling pathways and vital targets, which provides more opportunities for the clinical development of new treatments for sepsis.
Collapse
Affiliation(s)
- Wendan Zhang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
- Faculty of PediatricsNational Engineering Laboratory for Birth defects prevention and control of key technologyBeijing Key Laboratory of Pediatric Organ Failurethe Chinese PLA General HospitalBeijingChina
| | - Honghong Jiang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
- Faculty of PediatricsNational Engineering Laboratory for Birth defects prevention and control of key technologyBeijing Key Laboratory of Pediatric Organ Failurethe Chinese PLA General HospitalBeijingChina
| | - Gaosong Wu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Pengli Huang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Haonan Wang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Huazhasng An
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational MedicineThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanShandongChina
| | - Sanhong Liu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Weidong Zhang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
- Department of PhytochemistrySchool of PharmacySecond Military Medical UniversityShanghaiChina
- The Research Center for Traditional Chinese MedicineShanghai Institute of Infectious Diseases and BiosecurityShanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of Medicinal Plant DevelopmentChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
25
|
Hao M, Han X, Yao Z, Zhang H, Zhao M, Peng M, Wang K, Shan Q, Sang X, Wu X, Wang L, Lv Q, Yang Q, Bao Y, Kuang H, Zhang H, Cao G. The pathogenesis of organ fibrosis: Focus on necroptosis. Br J Pharmacol 2023; 180:2862-2879. [PMID: 36111431 DOI: 10.1111/bph.15952] [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: 04/14/2022] [Revised: 07/20/2022] [Accepted: 07/28/2022] [Indexed: 11/29/2022] Open
Abstract
Fibrosis is a common process of tissue repair response to multiple injuries in all chronic progressive diseases, which features with excessive deposition of extracellular matrix. Fibrosis can occur in all organs and tends to be nonreversible with the progress of the disease. Different cells types in different organs are involved in the occurrence and development of fibrosis, that is, hepatic stellate cells, pancreatic stellate cells, fibroblasts and myofibroblasts. Various types of programmed cell death, including apoptosis, autophagy, ferroptosis and necroptosis, are closely related to organ fibrosis. Among these programmed cell death types, necroptosis, an emerging regulated cell death type, is regarded as a huge potential target to ameliorate organ fibrosis. In this review, we summarize the role of necroptosis signalling in organ fibrosis and collate the small molecule compounds targeting necroptosis. In addition, we discuss the potential challenges, opportunities and open questions in using necroptosis signalling as a potential target for antifibrotic therapies. LINKED ARTICLES: This article is part of a themed issue on Translational Advances in Fibrosis as a Therapeutic Target. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v180.22/issuetoc.
Collapse
Affiliation(s)
- Min Hao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xin Han
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhouhui Yao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Han Zhang
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Mengting Zhao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Mengyun Peng
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Kuilong Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiyuan Shan
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xianan Sang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xin Wu
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lu Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiang Lv
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiao Yang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yini Bao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Haodan Kuang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Hongyan Zhang
- Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
26
|
Barar E, Shi J. Genome, Metabolism, or Immunity: Which Is the Primary Decider of Pancreatic Cancer Fate through Non-Apoptotic Cell Death? Biomedicines 2023; 11:2792. [PMID: 37893166 PMCID: PMC10603981 DOI: 10.3390/biomedicines11102792] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a solid tumor characterized by poor prognosis and resistance to treatment. Resistance to apoptosis, a cell death process, and anti-apoptotic mechanisms, are some of the hallmarks of cancer. Exploring non-apoptotic cell death mechanisms provides an opportunity to overcome apoptosis resistance in PDAC. Several recent studies evaluated ferroptosis, necroptosis, and pyroptosis as the non-apoptotic cell death processes in PDAC that play a crucial role in the prognosis and treatment of this disease. Ferroptosis, necroptosis, and pyroptosis play a crucial role in PDAC development via several signaling pathways, gene expression, and immunity regulation. This review summarizes the current understanding of how ferroptosis, necroptosis, and pyroptosis interact with signaling pathways, the genome, the immune system, the metabolism, and other factors in the prognosis and treatment of PDAC.
Collapse
Affiliation(s)
- Erfaneh Barar
- Liver and Pancreatobiliary Diseases Research Center, Digestive Disease Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran 1416753955, Iran
| | - Jiaqi Shi
- Department of Pathology & Clinical Labs, Rogel Cancer Center, Center for RNA Biomedicine, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
27
|
Du L, Wu Y, Pan Y, Wang L, Zhang H, Li J, Liu Y, Zhang H, He P. Lipopolysaccharide and Glycolipoprotein Coordinately Triggered Necroptosis Contributes to the Pathogenesis of Leptospira Infection in Mice. J Infect Dis 2023; 228:944-956. [PMID: 37166078 DOI: 10.1093/infdis/jiad151] [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: 09/21/2022] [Revised: 01/08/2023] [Accepted: 05/09/2023] [Indexed: 05/12/2023] Open
Abstract
Leptospirosis is a recurring but neglected zoonotic disease caused by pathogenic Leptospira. The explicit underlying mechanism of necroptosis and its role in Leptospira infection have not yet been elucidated. Here we reported that leptospiral pathogen-associated molecular patterns, lipopolysaccharide, and glycolipoprotein activate the necroptotic RIPK1-RIPK3-MLKL cascade through the TLR4 signaling pathway in mouse macrophages. Using the murine acute leptospirosis model, we reveal that abolition of necroptosis exhibited significantly improved outcomes in acute phases, with enhanced eradication of Leptospira from liver, mild clinical symptoms, and decreased cytokine production. RIPK3 was also found to exert a necroptosis-independent function in CXCL1 production and neutrophil recruitment, with the consequence of improved Leptospira control. These findings improve our understanding of the mechanism of Leptospira-macrophage interactions, indicating potential therapeutic values by targeting necroptosis signaling pathways.
Collapse
Affiliation(s)
- Lin Du
- Department of Immunology and Microbiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yunqiang Wu
- Department of Immunology and Microbiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuqing Pan
- Department of Immunology and Microbiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lingxia Wang
- Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Haiwei Zhang
- Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jiayin Li
- Department of Immunology and Microbiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ya'nan Liu
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haibing Zhang
- Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ping He
- Department of Immunology and Microbiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
28
|
Xiao H, Zhao Q, Yuan J, Liang W, Wu R, Wen Y, Du S, Wang Y, Zhao S, Lang Y, Yan Q, Huang X, Cao S. IFN-γ promotes PANoptosis in Pasteurella multocida toxin-induced pneumonia in mice. Vet Microbiol 2023; 285:109848. [PMID: 37722207 DOI: 10.1016/j.vetmic.2023.109848] [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: 03/06/2023] [Revised: 08/07/2023] [Accepted: 08/11/2023] [Indexed: 09/20/2023]
Abstract
Interferon-γ (IFN-γ) is a pleiotropic cytokine that regulates diverse biological functions, including modulation of inflammatory response and innate and adaptive immunity. In our study, we found that IFN-γ plays an important role in the regulation of Pasteurella multocida toxin-associated pneumonia. In work described here, we demonstrated that rPMT induced a lethal pneumonia in WT mice and the severity of the pneumonia was substantially alleviated in IFN-γ-deficient mice, IFN-γ deficiency significantly elevated the survival rate and reduced the pathological lesions of the lungs after rPMT challenged. Notably, IFN-γ deficiency significantly decreased myeloperoxidase (MPO) expression abundance in the lung tissue, and the MPO was mainly expressed in the lung tissue injury region of WT mice. More importantly, IFN-γ deficiency impaired the activation of PANoptosis specific markers, including the caspase 3, GSDMD, and MLKL, and reduced the expression of IL-1β. Cumulatively, this study demonstrates that IFN-γ promotes PANoptosis in PMT induced pneumonia in mice, providing a basis for studying the pathogenic mechanism of PMT.
Collapse
Affiliation(s)
- Hang Xiao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Qin Zhao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China; National Demonstration Center for Experimental Animal Education, Chengdu 611130, China
| | - Jianlin Yuan
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Wei Liang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Rui Wu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China; National Demonstration Center for Experimental Animal Education, Chengdu 611130, China
| | - Yiping Wen
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China; National Demonstration Center for Experimental Animal Education, Chengdu 611130, China
| | - Senyan Du
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China; National Demonstration Center for Experimental Animal Education, Chengdu 611130, China
| | - Yiping Wang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China; National Demonstration Center for Experimental Animal Education, Chengdu 611130, China
| | - Shan Zhao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China; National Demonstration Center for Experimental Animal Education, Chengdu 611130, China
| | - Yifei Lang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China; National Demonstration Center for Experimental Animal Education, Chengdu 611130, China
| | - Qigui Yan
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China; National Demonstration Center for Experimental Animal Education, Chengdu 611130, China
| | - Xiaobo Huang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China; National Demonstration Center for Experimental Animal Education, Chengdu 611130, China
| | - Sanjie Cao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China; National Demonstration Center for Experimental Animal Education, Chengdu 611130, China.
| |
Collapse
|
29
|
He XY, Wang F, Suo XG, Gu MZ, Wang JN, Xu CH, Dong YH, He Y, Zhang Y, Ji ML, Chen Y, Zhang MM, Fan YG, Wen JG, Jin J, Wang J, Li J, Zhuang CL, Liu MM, Meng XM. Compound-42 alleviates acute kidney injury by targeting RIPK3-mediated necroptosis. Br J Pharmacol 2023; 180:2641-2660. [PMID: 37248964 DOI: 10.1111/bph.16152] [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: 07/20/2022] [Revised: 05/02/2023] [Accepted: 05/15/2023] [Indexed: 05/31/2023] Open
Abstract
BACKGROUND AND PURPOSE Necroptosis plays an essential role in acute kidney injury and is mediated by receptor-interacting protein kinase 1 (RIPK1), receptor-interacting protein kinase 3 (RIPK3), and mixed lineage kinase domain-like pseudokinase (MLKL). A novel RIPK3 inhibitor, compound 42 (Cpd-42) alleviates the systemic inflammatory response. The current study was designed to investigate whether Cpd-42 exhibits protective effects on acute kidney injury and reveal the underlying mechanisms. EXPERIMENTAL APPROACH The effects of Cpd-42 were determined in vivo through cisplatin- and ischaemia/reperfusion (I/R)-induced acute kidney injury and in vitro through cisplatin- and hypoxia/re-oxygenation (H/R)-induced cell damage. Transmission electron microscopy and periodic acid-Schiff staining were used to identify renal pathology. Cellular thermal shift assay and RIPK3-knockout mouse renal tubule epithelial cells were used to explore the relationship between Cpd-42 and RIPK3. Molecular docking and site-directed mutagenesis were used to determine the binding site of RIPK3 with Cpd-42. KEY RESULTS Cpd-42 reduced human proximal tubule epithelial cell line (HK-2) cell damage, necroptosis and inflammatory responses in vitro. Furthermore, in vivo, cisplatin- and I/R-induced acute kidney injury was alleviated by Cpd-42 treatment. Cpd-42 inhibited necroptosis by interacting with two key hydrogen bonds of RIPK3 at Thr94 and Ser146, which further blocked the phosphorylation of RIPK3 and mitigated acute kidney injury. CONCLUSION AND IMPLICATIONS Acting as a novel RIPK3 inhibitor, Cpd-42 reduced kidney damage, inflammatory response and necroptosis in acute kidney injury by binding to sites Thr94 and Ser146 on RIPK3. Cpd-42 could be a promising treatment for acute kidney injury.
Collapse
Affiliation(s)
- Xiao-Yan He
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China
| | - Fang Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China
- Department of Pharmacy, Lu'an Hospital of Anhui Medical University, Lu'an People's Hospital of Anhui Province, Lu'an, China
| | - Xiao-Guo Suo
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China
| | - Ming-Zhen Gu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China
| | - Jia-Nan Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China
| | - Chuan-Hui Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China
| | - Yu-Hang Dong
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China
| | - Yuan He
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China
| | - Yao Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China
| | - Ming-Lu Ji
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China
| | - Ying Chen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China
| | - Meng-Meng Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China
| | - Yin-Guang Fan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Jia-Gen Wen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China
| | - Juan Jin
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Jie Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China
| | - Jun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China
| | - Chun-Lin Zhuang
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Ming-Ming Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China
| | - Xiao-Ming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China
| |
Collapse
|
30
|
Garnish SE, Martin KR, Kauppi M, Jackson VE, Ambrose R, Eng VV, Chiou S, Meng Y, Frank D, Tovey Crutchfield EC, Patel KM, Jacobsen AV, Atkin-Smith GK, Di Rago L, Doerflinger M, Horne CR, Hall C, Young SN, Cook M, Athanasopoulos V, Vinuesa CG, Lawlor KE, Wicks IP, Ebert G, Ng AP, Slade CA, Pearson JS, Samson AL, Silke J, Murphy JM, Hildebrand JM. A common human MLKL polymorphism confers resistance to negative regulation by phosphorylation. Nat Commun 2023; 14:6046. [PMID: 37770424 PMCID: PMC10539340 DOI: 10.1038/s41467-023-41724-6] [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: 09/27/2022] [Accepted: 09/13/2023] [Indexed: 09/30/2023] Open
Abstract
Across the globe, 2-3% of humans carry the p.Ser132Pro single nucleotide polymorphism in MLKL, the terminal effector protein of the inflammatory form of programmed cell death, necroptosis. Here we show that this substitution confers a gain in necroptotic function in human cells, with more rapid accumulation of activated MLKLS132P in biological membranes and MLKLS132P overriding pharmacological and endogenous inhibition of MLKL. In mouse cells, the equivalent Mlkl S131P mutation confers a gene dosage dependent reduction in sensitivity to TNF-induced necroptosis in both hematopoietic and non-hematopoietic cells, but enhanced sensitivity to IFN-β induced death in non-hematopoietic cells. In vivo, MlklS131P homozygosity reduces the capacity to clear Salmonella from major organs and retards recovery of hematopoietic stem cells. Thus, by dysregulating necroptosis, the S131P substitution impairs the return to homeostasis after systemic challenge. Present day carriers of the MLKL S132P polymorphism may be the key to understanding how MLKL and necroptosis modulate the progression of complex polygenic human disease.
Collapse
Affiliation(s)
- Sarah E Garnish
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Katherine R Martin
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Maria Kauppi
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Victoria E Jackson
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Rebecca Ambrose
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Vik Ven Eng
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - Shene Chiou
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Yanxiang Meng
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Daniel Frank
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
| | - Emma C Tovey Crutchfield
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Faculty of Medicine, Dentistry and Health Sciences, Parkville, VIC, Australia
| | - Komal M Patel
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
| | - Annette V Jacobsen
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Georgia K Atkin-Smith
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Ladina Di Rago
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Marcel Doerflinger
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Christopher R Horne
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Cathrine Hall
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
| | - Samuel N Young
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
| | - Matthew Cook
- Centre for Personalised Immunology and Canberra Clinical Genomics, Australian National University, Canberra, ACT, Australia
- Cambridge Institute for Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK
| | - Vicki Athanasopoulos
- Department of Immunology and Infection, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Carola G Vinuesa
- Centre for Personalised Immunology and Canberra Clinical Genomics, Australian National University, Canberra, ACT, Australia
- Department of Immunology and Infection, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- The Francis Crick Institute, London, UK
- University College London, London, UK
- China Australia Centre for Personalized Immunology (CACPI), Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Kate E Lawlor
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Ian P Wicks
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Gregor Ebert
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Ashley P Ng
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
- Clinical Haematology Department, The Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Parkville, VIC, Australia
| | - Charlotte A Slade
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
- Department of Clinical Immunology & Allergy, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Jaclyn S Pearson
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
- Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - André L Samson
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - John Silke
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - James M Murphy
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Joanne M Hildebrand
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia.
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia.
| |
Collapse
|
31
|
Wang Y, Hong Y, Mao S, Pan J, Cui Y, Lu J, Wen T, Wang X, Luo Y. Downregulation of miR-124-3p suppresses the development of the deep retinal blood vessels by enhancing the Stat1/Ripk1 pathway in mouse retinal microglia. Exp Eye Res 2023; 233:109551. [PMID: 37356537 DOI: 10.1016/j.exer.2023.109551] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 06/18/2023] [Accepted: 06/22/2023] [Indexed: 06/27/2023]
Abstract
The study aimed to investigate the role of microRNA (miR)-124-3p in retinal angiogenesis in a mouse model. An intravitreal injection of miR-124-3p antagomir was used to knockdown the expression of miR-124-3p in the mouse retina at postnatal day (P)3. Immunofluorescent staining of both retinal frozen sections and whole retina were used to observe retinal vascular development in the P6, P9 and P12 mice, as well as the changes in retinal ganglion cells, astrocytes, Müller cells and microglia. Whole retinal RNA extracted from P9 mice was used for transcriptome sequencing. Following gene set enrichment analysis, the enriched genes caused by miR-124-3p inhibition were analyzed by immunofluorescent staining and western blot. Results indicated that deep vascular development was significantly inhibited by the activation of M1 phenotype microglia. Moreover, there were no notable effects on superficial retinal vascular development, the retinal ganglion cells, astrocytes, and Müller cells. The expression of the Stat1/Irf9/Eif2ak2/Ripk1 axis in the miR-124-3p knockdown group was significantly increased. The microglia penetrated deep into the retina and the activation of Ripk1(+) microglia significantly increased, which was accompanied by an increased level of apoptosis to inhibit the deep vascular sprout. Downregulation of miR-124-3p during the early retinal development can suppress the development of the deep retinal blood vessels by enhancing the expression level of the Stat1/Irf9/Eif2ak2/Ripk1 axis and inducing the cell apoptosis of the activation of Ripk1(+) microglia.
Collapse
Affiliation(s)
- Yishen Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Yiwen Hong
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Shudi Mao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Jianying Pan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Yamei Cui
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Jing Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Tao Wen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Xiao Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Yan Luo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
32
|
Hong L, Williams NL, Jaffe M, Shields CE, Haynes KA. Synthetic Reader-Actuators Targeted to Polycomb-Silenced Genes Block Triple-Negative Breast Cancer Proliferation and Invasion. GEN BIOTECHNOLOGY 2023; 2:301-316. [PMID: 37928406 PMCID: PMC10623628 DOI: 10.1089/genbio.2023.0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 06/06/2023] [Indexed: 11/07/2023]
Abstract
Scientists have used pharmacological inhibitors of polycomb proteins to restore the expression of tumor suppressor genes and stop cancer proliferation and invasion. A major limitation of this approach is that key transcriptional activators, such as TP53 and BAF SWI/SNF, are often mutated in cancer. Poor clinical results for polycomb-targeting therapies in solid cancers, including triple-negative breast cancer (TNBC), could discourage the further development of epigenetic monotherapies. Here, we performed epigenome actuation with a synthetic reader-actuator (SRA) that binds trimethylated histone H3 lysine 27 in polycomb chromatin and modulates core transcriptional activators. In SRA-expressing TNBC BT-549 cells, 122 genes become upregulated ≥2-fold, including the genes involved in cell death, cell cycle arrest, and migration inhibition. The SRA-expressing spheroids showed reduced size in Matrigel and loss of invasion. Therefore, targeting Mediator-recruiting regulators to silenced chromatin can activate tumor suppressors and stimulate anti-cancer phenotypes, and further development of robust gene regulators might benefit TNBC patients.
Collapse
Affiliation(s)
- Lauren Hong
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA; and Emory University, Atlanta, Georgia, USA
| | - Natecia L. Williams
- Wallace H. Coulter Department of Biomedical Engineering, Emory University, Atlanta, Georgia, USA
| | - Maya Jaffe
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA; and Emory University, Atlanta, Georgia, USA
| | - Cara E. Shields
- Wallace H. Coulter Department of Biomedical Engineering, Emory University, Atlanta, Georgia, USA
| | - Karmella A. Haynes
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA; and Emory University, Atlanta, Georgia, USA
- Wallace H. Coulter Department of Biomedical Engineering, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
33
|
Martinez-Osorio V, Abdelwahab Y, Ros U. The Many Faces of MLKL, the Executor of Necroptosis. Int J Mol Sci 2023; 24:10108. [PMID: 37373257 DOI: 10.3390/ijms241210108] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 05/31/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Necroptosis is a recently discovered form of regulated cell death characterized by the disruption of plasma membrane integrity and the release of intracellular content. Mixed lineage kinase domain-like (MLKL) protein is the main player of this cell death pathway as it mediates the final step of plasma membrane permeabilization. Despite the significant progress in our knowledge of the necroptotic pathway and MLKL biology, the precise mechanism of how MLKL functions remain unclear. To understand in what way MLKL executes necroptosis, it is crucial to decipher how the molecular machinery of regulated cell death is activated in response to different stimuli or stressors. It is also indispensable to unveiling the structural elements of MLKL and the cellular players that are required for its regulation. In this review, we discuss the key steps that lead to MLKL activation, possible models that explain how it becomes the death executor in necroptosis, and its emerging alternative functions. We also summarize the current knowledge about the role of MLKL in human disease and provide an overview of existing strategies aimed at developing new inhibitors that target MLKL for necroptosis intervention.
Collapse
Affiliation(s)
- Veronica Martinez-Osorio
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Yasmin Abdelwahab
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Uris Ros
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
34
|
Guo XX, Pu Q, Hu JJ, Chang XJ, Li AL, Li XY. The role of regulated necrosis in inflammation and ocular surface diseases. Exp Eye Res 2023:109537. [PMID: 37302745 DOI: 10.1016/j.exer.2023.109537] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/28/2023] [Accepted: 06/08/2023] [Indexed: 06/13/2023]
Abstract
In recent decades, numerous types of regulated cell death have been identified, including pyroptosis, ferroptosis and necroptosis. Regulated necrosis is characterized by a series of amplified inflammatory responses that result in cell death. Therefore, it has been suggested to play an essential role in the pathogenesis of ocular surface diseases. The cell morphological features and molecular mechanisms of regulated necrosis are discussed in this review. Furthermore, it summarizes the role of ocular surface diseases, such as dry eye, keratitis, and cornea alkali burn, as potential disease prevention and treatment targets.
Collapse
Affiliation(s)
- Xiao-Xiao Guo
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Qi Pu
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Jing-Jie Hu
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Xue-Jiao Chang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Ao-Ling Li
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Xin-Yu Li
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
35
|
Ma J, Xu C, Jiang N, Meng Y, Zhou Y, Xue M, Liu W, Li Y, Fan Y. Transcriptomics in Rare Minnow ( Gobiocypris rarus) towards Attenuated and Virulent Grass Carp Reovirus Genotype II Infection. Animals (Basel) 2023; 13:1870. [PMID: 37889762 PMCID: PMC10251909 DOI: 10.3390/ani13111870] [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: 04/21/2023] [Revised: 05/31/2023] [Accepted: 06/02/2023] [Indexed: 10/29/2023] Open
Abstract
Grass carp reovirus genotype Ⅱ (GCRV Ⅱ) causes a variety of fish hemorrhagic disease, which seriously affects the sustainable development of grass carp aquaculture in China. Rare minnow (Gobiocypris rarus) is an ideal model fish to study the pathogenesis of GCRV Ⅱ. To investigate the involved molecular responses against the GCRV Ⅱ infection, we performed comparative transcriptomic analysis in the spleen and liver of rare minnow injected with virulent strain DY197 and attenuated strain QJ205. Results showed that the virulent DY197 strain induced more differently expressed genes (DEGs) than the attenuated QJ205 strain, and tissue-specific responses were induced. In the spleen, the attenuated and virulent strains induced different DEGs; the attenuated QJ205 infection activated steroid synthesis pathway that involved in membrane formation; however, virulent DY197 infection activated innate immunity and apoptosis related pathways while suppressing cell proliferation and migration related pathways that are important for damage tissue repair, as well as hemorrhage related pathways. In the liver, the attenuated and virulent strains infection induced similar DEGs; both strains infection activated immunity and apoptosis related pathways but suppressed metabolism-related pathways; virulent DY197 infection especially activated protein digestion and absorption-related pathways and suppressed steroid synthesis pathway. To conclude, virulent strain infection especially induced tissue-specific alterations and caused severe suppression of hemorrhage-related pathways in spleen. Our findings will contribute to better understanding of the interactions between host and GCRV II.
Collapse
Affiliation(s)
- Jie Ma
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (J.M.); (C.X.); (N.J.); (Y.M.); (Y.Z.); (M.X.); (W.L.); (Y.L.)
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China
| | - Chen Xu
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (J.M.); (C.X.); (N.J.); (Y.M.); (Y.Z.); (M.X.); (W.L.); (Y.L.)
| | - Nan Jiang
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (J.M.); (C.X.); (N.J.); (Y.M.); (Y.Z.); (M.X.); (W.L.); (Y.L.)
| | - Yan Meng
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (J.M.); (C.X.); (N.J.); (Y.M.); (Y.Z.); (M.X.); (W.L.); (Y.L.)
| | - Yong Zhou
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (J.M.); (C.X.); (N.J.); (Y.M.); (Y.Z.); (M.X.); (W.L.); (Y.L.)
| | - Mingyang Xue
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (J.M.); (C.X.); (N.J.); (Y.M.); (Y.Z.); (M.X.); (W.L.); (Y.L.)
| | - Wenzhi Liu
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (J.M.); (C.X.); (N.J.); (Y.M.); (Y.Z.); (M.X.); (W.L.); (Y.L.)
| | - Yiqun Li
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (J.M.); (C.X.); (N.J.); (Y.M.); (Y.Z.); (M.X.); (W.L.); (Y.L.)
| | - Yuding Fan
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (J.M.); (C.X.); (N.J.); (Y.M.); (Y.Z.); (M.X.); (W.L.); (Y.L.)
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China
| |
Collapse
|
36
|
Kongsomboonvech AK, García-López L, Njume F, Rodriguez F, Souza SP, Rosenberg A, Jensen KDC. Variation in CD8 T cell IFNγ differentiation to strains of Toxoplasma gondii is characterized by small effect QTLs with contribution from ROP16. Front Cell Infect Microbiol 2023; 13:1130965. [PMID: 37287466 PMCID: PMC10242045 DOI: 10.3389/fcimb.2023.1130965] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 04/17/2023] [Indexed: 06/09/2023] Open
Abstract
Introduction Toxoplasma gondii induces a strong CD8 T cell response characterized by the secretion of IFNγ that promotes host survival during infection. The initiation of CD8 T cell IFNγ responses in vitro differs widely between clonal lineage strains of T. gondii, in which type I strains are low inducers, while types II and III strains are high inducers. We hypothesized this phenotype is due to a polymorphic "Regulator Of CD8 T cell Response" (ROCTR). Methods Therefore, we screened F1 progeny from genetic crosses between the clonal lineage strains to identify ROCTR. Naïve antigen-specific CD8 T cells (T57) isolated from transnuclear mice, which are specific for the endogenous and vacuolar TGD057 antigen, were measured for their ability to become activated, transcribe Ifng and produce IFNγ in response to T. gondii infected macrophages. Results Genetic mapping returned four non-interacting quantitative trait loci (QTL) with small effect on T. gondii chromosomes (chr) VIIb-VIII, X and XII. These loci encompass multiple gene candidates highlighted by ROP16 (chrVIIb-VIII), GRA35 (chrX), TgNSM (chrX), and a pair of uncharacterized NTPases (chrXII), whose locus we report to be significantly truncated in the type I RH background. Although none of the chromosome X and XII candidates bore evidence for regulating CD8 T cell IFNγ responses, type I variants of ROP16 lowered Ifng transcription early after T cell activation. During our search for ROCTR, we also noted the parasitophorous vacuole membrane (PVM) targeting factor for dense granules (GRAs), GRA43, repressed the response suggesting PVM-associated GRAs are important for CD8 T cell activation. Furthermore, RIPK3 expression in macrophages was an absolute requirement for CD8 T cell IFNγ differentiation implicating the necroptosis pathway in T cell immunity to T. gondii. Discussion Collectively, our data suggest that while CD8 T cell IFNγ production to T. gondii strains vary dramatically, it is not controlled by a single polymorphism with strong effect. However, early in the differentiation process, polymorphisms in ROP16 can regulate commitment of responding CD8 T cells to IFNγ production which may have bearing on immunity to T. gondii.
Collapse
Affiliation(s)
- Angel K. Kongsomboonvech
- Department of Molecular and Cell Biology, University of California, Merced, Merced, CA, United States
- Quantitative Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States
| | - Laura García-López
- Department of Molecular and Cell Biology, University of California, Merced, Merced, CA, United States
- Quantitative Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States
| | - Ferdinand Njume
- Department of Molecular and Cell Biology, University of California, Merced, Merced, CA, United States
| | - Felipe Rodriguez
- Department of Molecular and Cell Biology, University of California, Merced, Merced, CA, United States
| | - Scott P. Souza
- Department of Molecular and Cell Biology, University of California, Merced, Merced, CA, United States
- Quantitative Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States
| | - Alex Rosenberg
- The Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States
| | - Kirk D. C. Jensen
- Department of Molecular and Cell Biology, University of California, Merced, Merced, CA, United States
- Health Sciences Research Institute, University of California, Merced, Merced, CA, United States
| |
Collapse
|
37
|
Bozgeyik E, Bagis H, Bozgeyik I, Kocahan S. The roles of long non-coding RNAs in the necroptotic signaling of colon cancer cells. Mol Biol Rep 2023; 50:5021-5028. [PMID: 37097538 DOI: 10.1007/s11033-023-08441-1] [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: 02/23/2023] [Accepted: 04/06/2023] [Indexed: 04/26/2023]
Abstract
BACKGROUND Necroptosis is a controlled form of necrosis which can be stimulated in cases where the apoptosis signal is absent. Necroptosis can be induced by DR family ligands and by various intracellular and extracellular stimuli that triggers the activation of DR family ligands. Necrostatins, which are specific RIP1 antagonists, prevent necroptosis by inhibiting RIP1 kinase, allowing survival and propagation of cells in the presence of DR ligands. Furthermore, there is a mounting evidence that long non-coding RNA (lncRNA) molecules accomplish vital functions in cell death processes such as apoptosis, autophagy, pyroptosis, and necroptosis. Accordingly, here we aimed to decipher the lncRNAs that are involved in the control and maintenance of necroptosis signaling. METHODS AND RESULTS Colon cancer cell lines, HT-29 and HCT-116 were used for the study. For the chemical modulation of necroptosis signaling, 5-Fluorouracil, TNF-α and/or Necrostatin-1 were used. Gene expression levels were determined by quantitative real-time PCR. Remarkably, lncRNA P50-associated COX-2 extragenic RNA (PACER) was identified to be suppressed in necroptosis-induced colon cancers, whereas the expression of PACER was restored when necroptosis was suppressed. In addition, no detectable change was observed in HCT-116 colon cancer cells, as these cells lack the expression of RIP3 kinase. CONCLUSIONS Collectively, current findings clearly imply that PACER have key regulatory roles in the control of necroptotic cell death signaling circuitry. Notably, the tumor promoter activity of PACER might be responsible for the lack of necroptotic death signal in cancer cells. Also, RIP3 kinase seems to be essential component in PACER-associated necroptosis.
Collapse
Affiliation(s)
- Esra Bozgeyik
- Department of Medical Services and Techniques, Vocational School of Health Services, Adiyaman University, Adiyaman, Turkey.
| | - Haydar Bagis
- Department of Medical Genetics, Faculty of Medicine, Adiyaman University, Adiyaman, Turkey
| | - Ibrahim Bozgeyik
- Department of Medical Biology, Faculty of Medicine, Adiyaman University, Adiyaman, Turkey
| | - Sayad Kocahan
- Department of Physiology, Gulhane Medical Faculty, Health Sciences University, Ankara, Turkey
| |
Collapse
|
38
|
Yang D, Tang M, Zhang M, Ren H, Li X, Zhang Z, He B, Peng S, Wang W, Fang D, Song Y, Xiong Y, Liu ZZ, Liang L, Shi W, Fu C, Hu Y, Jose PA, Zhou L, Han Y, Zeng C. Downregulation of G protein-coupled receptor kinase 4 protects against kidney ischemia-reperfusion injury. Kidney Int 2023; 103:719-734. [PMID: 36669643 DOI: 10.1016/j.kint.2022.12.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 10/27/2022] [Accepted: 12/12/2022] [Indexed: 01/19/2023]
Abstract
Ischemia/reperfusion injury of the kidney is associated with high morbidity and mortality, and treatment of this injury remains a challenge. G protein-coupled receptor kinase 4 (GRK4) plays a vital role in essential hypertension and myocardial infarction, but its function in kidney ischemia/reperfusion injury remains undetermined. Among the GRK subtypes (GRK2-6) expressed in kidneys, the increase in GRK4 expression was much more apparent than that of the other four GRKs 24 hours after injury and was found to accumulate in the nuclei of injured mouse and human renal tubule cells. Gain- and loss-of-function experiments revealed that GRK4 overexpression exacerbated acute kidney ischemia/reperfusion injury, whereas kidney tubule-specific knockout of GRK4 decreased injury-induced kidney dysfunction. Necroptosis was the major type of tubule cell death mediated by GRK4, because GRK4 significantly increased receptor interacting kinase (RIPK)1 expression and phosphorylation, subsequently leading to RIPK3 and mixed lineage kinase domain-like protein (MLKL) phosphorylation after kidney ischemia/reperfusion injury, but was reversed by necrostatin-1 pretreatment (an RIPK1 inhibitor). Using co-immunoprecipitation, mass spectrometry, and siRNA screening studies, we identified signal transducer and activator of transcription (STAT)1 as a GRK4 binding protein, which co-localized with GRK4 in the nuclei of renal tubule cells. Additionally, GRK4 phosphorylated STAT1 at serine 727, whose inactive mutation effectively reversed GRK4-mediated RIPK1 activation and tubule cell death. Kidney-targeted GRK4 silencing with nanoparticle delivery considerably ameliorated kidney ischemia/reperfusion injury. Thus, our findings reveal that GRK4 triggers necroptosis and aggravates kidney ischemia/reperfusion injury, and its downregulation may provide a promising therapeutic strategy for kidney protection.
Collapse
Affiliation(s)
- Donghai Yang
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, People's Republic of China; Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, People's Republic of China
| | - Ming Tang
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, People's Republic of China; Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, People's Republic of China
| | - Mingming Zhang
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, People's Republic of China; Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, People's Republic of China
| | - Hongmei Ren
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, People's Republic of China; Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, People's Republic of China
| | - Xiaoping Li
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, People's Republic of China; Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, People's Republic of China
| | - Ziyue Zhang
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, People's Republic of China; Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, People's Republic of China
| | - Bo He
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, People's Republic of China; Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, People's Republic of China
| | - Song Peng
- Department of Urology, Daping Hospital, The Third Military Medical University, Chongqing, People's Republic of China
| | - Wei Wang
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, People's Republic of China; Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, People's Republic of China
| | - Dandong Fang
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, People's Republic of China; Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, People's Republic of China
| | - Yi Song
- Department of Cardiac Surgery, Daping Hospital, The Third Military Medical University, Chongqing, People's Republic of China
| | - Yao Xiong
- Cardiovascular Research Center of Chongqing College, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Chongqing, People's Republic of China
| | - Zhi Zhao Liu
- Cardiovascular Research Center of Chongqing College, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Chongqing, People's Republic of China
| | - Lijia Liang
- Cardiovascular Research Center of Chongqing College, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Chongqing, People's Republic of China
| | - Weibin Shi
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, People's Republic of China; Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, People's Republic of China
| | - Chunjiang Fu
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, People's Republic of China; Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, People's Republic of China
| | - Yijie Hu
- Department of Cardiac Surgery, Daping Hospital, The Third Military Medical University, Chongqing, People's Republic of China
| | - Pedro A Jose
- Division of Renal Diseases & Hypertension, Department of Medicine and Pharmacology-Physiology, The George Washington University School of Medicine & Health Sciences, Washington D.C., USA
| | - Lin Zhou
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, People's Republic of China; Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, People's Republic of China
| | - Yu Han
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, People's Republic of China; Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, People's Republic of China.
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, People's Republic of China; Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, People's Republic of China; Cardiovascular Research Center of Chongqing College, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Chongqing, People's Republic of China; State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, The Third Military Medical University, Chongqing, People's Republic of China.
| |
Collapse
|
39
|
Zhang J, Tu H, Zheng Z, Zhao X, Lin X. RNF31 promotes tumorigenesis via inhibiting RIPK1 kinase-dependent apoptosis. Oncogene 2023; 42:1585-1596. [PMID: 36997719 DOI: 10.1038/s41388-023-02669-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 03/06/2023] [Accepted: 03/13/2023] [Indexed: 04/01/2023]
Abstract
It is well established that interferon (IFN) and tumor necrosis factor (TNF) could synergistically promote antitumor toxicity and avoid resistance of antigen-negative tumors during cancer immunotherapy. The linear ubiquitin chain assembly complex (LUBAC) has been widely known to regulate receptor-interacting protein kinase-1(RIPK1) kinase activity and TNF-mediated cell death during inflammation and embryogenesis. However, whether LUBAC and RIPK1 kinase activity in tumor microenvironment could regulate antitumor immunity are still not very clear. Here, we demonstrated a cancer cell-intrinsic role of LUBAC complex in tumor microenvironment to promote tumorigenesis. Lacking LUBAC component RNF31 in B16 melanoma cells but not immune cells including macrophages or dendritic cells greatly impaired tumor growth by increasing intratumoral CD8+ T cells infiltration. Mechanistically, we found that tumor cells without RNF31 shown severe apoptosis-mediated cell death caused by TNFα/IFNγ in the tumor microenvironment. Most importantly, we found that RNF31 could limit RIPK1 kinase activity and further prevent tumor cell death in a transcription-independent manner, suggesting a crucial role of RIPK1 kinase activity in tumorigenesis. Together, our results demonstrate an essential role of RNF31 and RIPK1 kinase activity in tumorigenesis and imply that RNF31 inhibition could be harnessed to enhance antitumor toxicity during tumor immunotherapy.
Collapse
Affiliation(s)
- Jie Zhang
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, Tsinghua University School of Medicine, Beijing, 100084, China
| | - Hailin Tu
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, Tsinghua University School of Medicine, Beijing, 100084, China
| | - Zheyu Zheng
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, Tsinghua University School of Medicine, Beijing, 100084, China
| | - Xueqiang Zhao
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, Tsinghua University School of Medicine, Beijing, 100084, China
| | - Xin Lin
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China.
- Department of Basic Medical Sciences, Tsinghua University School of Medicine, Beijing, 100084, China.
| |
Collapse
|
40
|
Susceptibility and Permissivity of Zebrafish (Danio rerio) Larvae to Cypriniviruses. Viruses 2023; 15:v15030768. [PMID: 36992477 PMCID: PMC10051318 DOI: 10.3390/v15030768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/08/2023] [Accepted: 03/10/2023] [Indexed: 03/19/2023] Open
Abstract
The zebrafish (Danio rerio) represents an increasingly important model organism in virology. We evaluated its utility in the study of economically important viruses from the genus Cyprinivirus (anguillid herpesvirus 1, cyprinid herpesvirus 2 and cyprinid herpesvirus 3 (CyHV-3)). This revealed that zebrafish larvae were not susceptible to these viruses after immersion in contaminated water, but that infections could be established using artificial infection models in vitro (zebrafish cell lines) and in vivo (microinjection of larvae). However, infections were transient, with rapid viral clearance associated with apoptosis-like death of infected cells. Transcriptomic analysis of CyHV-3-infected larvae revealed upregulation of interferon-stimulated genes, in particular those encoding nucleic acid sensors, mediators of programmed cell death and related genes. It was notable that uncharacterized non-coding RNA genes and retrotransposons were also among those most upregulated. CRISPR/Cas9 knockout of the zebrafish gene encoding protein kinase R (PKR) and a related gene encoding a protein kinase containing Z-DNA binding domains (PKZ) had no impact on CyHV-3 clearance in larvae. Our study strongly supports the importance of innate immunity-virus interactions in the adaptation of cypriniviruses to their natural hosts. It also highlights the potential of the CyHV-3-zebrafish model, versus the CyHV-3-carp model, for study of these interactions.
Collapse
|
41
|
Kandhari K, Mishra JPN, Agarwal R, Singh RP. Acacetin induces sustained ERK1/2 activation and RIP1-dependent necroptotic death in breast cancer cells. Toxicol Appl Pharmacol 2023; 462:116409. [PMID: 36740148 DOI: 10.1016/j.taap.2023.116409] [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] [Received: 07/23/2022] [Revised: 01/25/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
Acacetin (AC), a naturally occurring flavonoid has shown anticancer potential. Herein, we studied the mechanisms of cell death and growth inhibition by AC in breast carcinoma T-47D and MDA-MB-231 cells. AC (10-40 μM) significantly decreased the levels of G2/M phase cyclins and CDKs, simultaneously increasing the expression of CDK inhibitors including Cip1/p21. A concentration-dependent increase in cell death was noted in both breast cancer cell lines with no such considerable effects on MCF-10A non-tumorigenic breast cells. The cell death-inducing potential of AC was further confirmed using confocal microscopy and flow cytometry analysis. AC resulted in mitochondrial superoxide generation, DNA damage, and ROS generation. N-acetyl cysteine (NAC) pre-treatment inhibited ROS generation and partially reversed ERK1/2 activation as well as cell death by AC. Further, AC enhanced the expression of RIP1 and RIP3, which mediate necroptosis. RIP1-specific inhibitor Necrostatin-1 (NS-1) reversed the AC-induced DNA damage and cell death. Collectively, these findings, for the first time, suggested that AC exerts its antitumor potential through ROS induction and RIP1-dependent necroptosis in breast carcinoma cells.
Collapse
Affiliation(s)
- Kushal Kandhari
- School of Life Sciences, Central University of Gujarat, Gandhinagar, Gujarat, India; Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jai P N Mishra
- School of Life Sciences, Central University of Gujarat, Gandhinagar, Gujarat, India
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Rana P Singh
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India.
| |
Collapse
|
42
|
Quan D, Hou R, Shao H, Zhang X, Yu J, Zhang W, Yuan H, Zhuang C. Structure-Based Design of Novel Alkynyl Thio-Benzoxazepinone Receptor-Interacting Protein Kinase-1 Inhibitors: Extending the Chemical Space from the Allosteric to ATP Binding Pockets. J Med Chem 2023; 66:3073-3087. [PMID: 36724216 DOI: 10.1021/acs.jmedchem.2c02067] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Systemic inflammatory response syndrome (SIRS), characterized by severe systemic inflammation, represents a major cause of health loss, potentially leading to multiple organ failure, shock, and death. Exploring potent RIPK1 inhibitors is an effective therapeutic strategy for SIRS. Recently, we described thio-benzoxazepinones as novel RIPK1 inhibitors and confirmed their anti-inflammatory activity. Herein, we further synthesized novel thio-benzoxazepinones by introducing substitutions on the benzene ring by an alkynyl bridge in order to extend the chemical space from the RIPK1 allosteric to ATP binding pockets. The in vitro cell and kinase assays found that compounds 2 and 29 showed highly potent activity against necroptosis (EC50 = 3.7 and 3.2 nM) and high RIPK1 inhibitory activity (Kd = 9.7 and 70 nM). Prominently, these two analogues possessed better in vivo anti-inflammatory effects than the clinical candidate GSK'772 and effectively blocked hypothermia and deaths in a TNFα-induced SIRS model.
Collapse
Affiliation(s)
- Danni Quan
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Ruilin Hou
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Hongming Shao
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Xinqi Zhang
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Jianqiang Yu
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Wannian Zhang
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China.,School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Hongbin Yuan
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Chunlin Zhuang
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China.,School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
43
|
Lomphithak T, Fadeel B. Die hard: cell death mechanisms and their implications in nanotoxicology. Toxicol Sci 2023; 192:kfad008. [PMID: 36752525 PMCID: PMC10109533 DOI: 10.1093/toxsci/kfad008] [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] [Indexed: 02/09/2023] Open
Abstract
Cell death is a fundamental biological process, and its fine-tuned regulation is required for life. However, the complexity of regulated cell death is often reduced to a matter of live-dead discrimination. Here, we provide a perspective on programmed or regulated cell death, focusing on apoptosis, pyroptosis, necroptosis, and ferroptosis (the latter three cell death modalities are examples of regulated necrosis). We also touch on other, recently described manifestations of (pathological) cell death including cuproptosis. Furthermore, we address how engineered nanomaterials impact on regulated cell death. We posit that an improved understanding of nanomaterial-induced perturbations of cell death may allow for a better prediction of the consequences of human exposure and could also yield novel approaches by which to mitigate these effects. Finally, we provide examples of the harnessing of nanomaterials to achieve cancer cell killing through the induction of regulated cell death.
Collapse
Affiliation(s)
- Thanpisit Lomphithak
- Division of Molecular Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Bengt Fadeel
- Division of Molecular Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| |
Collapse
|
44
|
Williams NL, Hong L, Jaffe M, Shields CE, Haynes KA. PIC recruitment by synthetic reader-actuators to polycomb-silenced genes blocks triple-negative breast cancer invasion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.23.525196. [PMID: 36747762 PMCID: PMC9900809 DOI: 10.1101/2023.01.23.525196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Scientists have used small molecule inhibitors and genetic knockdown of gene-silencing polycomb repressive complexes (PRC1/2) to determine if restoring the expression of tumor suppressor genes can block proliferation and invasion of cancer cells. A major limitation of this approach is that inhibitors can not restore key transcriptional activators that are mutated in many cancers, such as p53 and members of the BRAF SWI/SNF complex. Furthermore, small molecule inhibitors can alter the activity of, rather than inhibit, the polycomb enzyme EZH2. While chromatin has been shown to play a major role in gene regulation in cancer, poor clinical results for polycomb chromatin-targeting therapies for diseases like triple-negative breast cancer (TNBC) could discourage further development of this emerging avenue for treatment. To overcome the limitations of inhibiting polycomb to study epigenetic regulation, we developed an engineered chromatin protein to manipulate transcription. The synthetic reader-actuator (SRA) is a fusion protein that directly binds a target chromatin modification and regulates gene expression. Here, we report the activity of an SRA built from polycomb chromodomain and VP64 modules that bind H3K27me3 and subunits of the Mediator complex, respectively. In SRA-expressing BT-549 cells, we identified 122 upregulated differentially expressed genes (UpDEGs, ≥ 2-fold activation, adjusted p < 0.05). On-target epigenetic regulation was determined by identifying UpDEGs at H3K27me3-enriched, closed chromatin. SRA activity induced activation of genes involved in cell death, cell cycle arrest, and the inhibition of migration and invasion. SRA-expressing BT-549 cells showed reduced spheroid size in Matrigel over time, loss of invasion, and activation of apoptosis. These results show that Mediator-recruiting regulators broadly targeted to silenced chromatin activate silenced tumor suppressor genes and stimulate anti-cancer phenotypes. Therefore further development of gene-activating epigenetic therapies might benefit TNBC patients.
Collapse
Affiliation(s)
- Natecia L Williams
- Wallace H. Coulter Department of Biomedical Engineering, Emory University, Atlanta, GA 30312 USA
| | - Lauren Hong
- Georgia Institute of Technology, Atlanta, GA 30332
| | - Maya Jaffe
- Georgia Institute of Technology, Atlanta, GA 30332
| | - Cara E Shields
- Wallace H. Coulter Department of Biomedical Engineering, Emory University, Atlanta, GA 30312 USA
| | - Karmella A Haynes
- Wallace H. Coulter Department of Biomedical Engineering, Emory University, Atlanta, GA 30312 USA
| |
Collapse
|
45
|
Abstract
Herpes simplex virus (HSV)-1 and HSV-2 are ubiquitous human pathogens that infect keratinized epithelial surfaces and establish lifelong latent infection in sensory neurons of the peripheral nervous system. HSV-1 causes oral cold sores, and HSV-2 causes genital lesions characterized by recurrence at the site of the initial infection. In multicellular organisms, cell death plays a pivotal role in host defense by eliminating pathogen-infected cells. Apoptosis and necrosis are readily distinguished types of cell death. Apoptosis, the main form of programmed cell death, depends on the activity of certain caspases, a family of cysteine proteases. Necroptosis, a regulated form of necrosis that is unleashed when caspase activity is compromised, requires the activation of receptor-interacting protein (RIP) kinase 3 (RIPK3) through its interaction with other RIP homotypic interaction motif (RHIM)-containing proteins such as RIPK1. To ensure lifelong infection in the host, HSV carries out sophisticated molecular strategies to evade host cell death responses during viral infection. HSV-1 is a well-characterized pathogen that encodes potent viral inhibitors that modulate both caspase activation in the apoptosis pathway and RIPK3 activation in the necroptosis pathway in a dramatic, species-specific fashion. The viral UL39-encoded viral protein ICP6, the large subunit of the virus-encoded ribonucleotide reductase, functions as a suppressor of both caspase-8 and RHIM-dependent RIPK3 activities in the natural human host. In contrast, ICP6 RHIM-mediated recruitment of RIPK3 in the nonnatural mouse host drives the direct activation of necroptosis. This chapter provides an overview of the current state of the knowledge on molecular interactions between HSV-1 viral proteins and host cell death pathways and highlights how HSV-1 manipulates cell death signals for the benefit of viral propagation.
Collapse
Affiliation(s)
- Sudan He
- Center of Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China.
- Suzhou Institute of Systems Medicine, Suzhou, 215123, China.
| | - Jiahuai Han
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China.
| |
Collapse
|
46
|
Zhang R, Song Y, Su X. Necroptosis and Alzheimer's Disease: Pathogenic Mechanisms and Therapeutic Opportunities. J Alzheimers Dis 2023; 94:S367-S386. [PMID: 36463451 PMCID: PMC10473100 DOI: 10.3233/jad-220809] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2022] [Indexed: 11/30/2022]
Abstract
Alzheimer's disease (AD) is considered to be the most common neurodegenerative disease, with clinical symptoms encompassing progressive memory loss and cognitive impairment. Necroptosis is a form of programmed necrosis that promotes cell death and neuroinflammation, which further mediates the pathogenesis of several neurodegenerative diseases, especially AD. Current evidence has strongly suggested that necroptosis is activated in AD brains, resulting in neuronal death and cognitive impairment. We searched the PubMed database, screening all articles published before September 28, 2022 related to necroptosis in the context of AD pathology. The keywords in the search included: "necroptosis", "Alzheimer's disease", "signaling pathways", "Aβ", Aβo", "Tau", "p-Tau", "neuronal death", "BBB damage", "neuroinflammation", "microglia", "mitochondrial dysfunction", "granulovacuolar degeneration", "synaptic loss", "axonal degeneration", "Nec-1", "Nec-1s", "GSK872", "NSA", "OGA", "RIPK1", "RIPK3", and "MLKL". Results show that necroptosis has been involved in multiple pathological processes of AD, including amyloid-β aggregation, Tau accumulation, neuronal death, and blood-brain barrier damage, etc. More importantly, existing research on AD necroptosis interventions, including drug intervention and potential gene targets, as well as its current clinical development status, was discussed. Finally, the issues pertaining to necroptosis in AD were presented. Accordingly, this review may provide further insight into clinical perspectives and challenges for the future treatment of AD by targeting the necroptosis pathway.
Collapse
Affiliation(s)
- Ruxin Zhang
- Linfen People’s Hospital, Linfen, Shanxi, China
| | | | - Xuefeng Su
- Linfen People’s Hospital, Linfen, Shanxi, China
| |
Collapse
|
47
|
Thomas PG, Shubina M, Balachandran S. ZBP1/DAI-Dependent Cell Death Pathways in Influenza A Virus Immunity and Pathogenesis. Curr Top Microbiol Immunol 2023; 442:41-63. [PMID: 31970498 DOI: 10.1007/82_2019_190] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Influenza A viruses (IAV) are members of the Orthomyxoviridae family of negative-sense RNA viruses. The greatest diversity of IAV strains is found in aquatic birds, but a subset of strains infects other avian as well as mammalian species, including humans. In aquatic birds, infection is largely restricted to the gastrointestinal tract and spread is through feces, while in humans and other mammals, respiratory epithelial cells are the primary sites supporting productive replication and transmission. IAV triggers the death of most cell types in which it replicates, both in culture and in vivo. When well controlled, such cell death is considered an effective host defense mechanism that eliminates infected cells and limits virus spread. Unchecked or inopportune cell death also results in immunopathology. In this chapter, we discuss the impact of cell death in restricting virus spread, supporting the adaptive immune response and driving pathogenesis in the mammalian respiratory tract. Recent studies have begun to shed light on the signaling pathways underlying IAV-activated cell death. These pathways, initiated by the pathogen sensor protein ZBP1 (also called DAI and DLM1), cause infected cells to undergo apoptosis, necroptosis, and pyroptosis. We outline mechanisms of ZBP1-mediated cell death signaling following IAV infection.
Collapse
Affiliation(s)
- Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, MS 351, 262 Danny Thomas Place, 38105, Memphis, TN, USA.
| | - Maria Shubina
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Room 224 Reimann Building, 333 Cottman Ave., 19111, Philadelphia, PA, USA
| | - Siddharth Balachandran
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Room 224 Reimann Building, 333 Cottman Ave., 19111, Philadelphia, PA, USA.
| |
Collapse
|
48
|
Lu Y, Wang D, Zhu Y, Du Y, Zhang J, Yang H. A risk model developed based on necroptosis to assess progression for ischemic cardiomyopathy and identify possible therapeutic drugs. Front Pharmacol 2022; 13:1039857. [PMID: 36518671 PMCID: PMC9744324 DOI: 10.3389/fphar.2022.1039857] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 10/28/2022] [Indexed: 09/26/2023] Open
Abstract
Object: Ischemic cardiomyopathy (ICM), with high morbidity and mortality, is the most common cause of heart failure. Cardiovascular remodeling secondary to chronic myocardial ischemia is the main cause of its progression. A recently identified type of programmed cell death called necroptosis is crucial in the development of various cardiovascular diseases. However, the function role of necroptosis in cardiac remodeling of ICM has not been elucidated. Our study aimed to screen for genes associated with necroptosis and construct a risk score to assess the progression and evaluate the prognosis of ICM patients, and further to search for potentially therapeutic drugs. Methods: The gene expression profiling was obtained from the GEO database. LASSO regression analysis was used to construct necroptosis-related gene signatures associated with ICM progression and prognosis. TF-gene and miRNA-gene networks were constructed to identify the regulatory targets of potential necroptosis-related signature genes. Pathway alterations in patients with high necroptosis-related score (NRS) were analyzed by GO, KEGG, GSEA analysis, and immune cell infiltration was estimated by ImmuCellAI analysis. CMap analysis was performed to screen potential small molecule compounds targeting patients with high NRS. Independent risk analyses were performed using nomograms. Results: Six necroptosis-related signature genes (STAT4, TNFSF10, CHMP5, CHMP18, JAK1, and CFLAR) were used to define the NRS, with areas under the ROC curves of 0.833, 0.765, and 0.75 for training test, test set, and validation set, respectively. Transcription factors FOXC1 and hsa-miR-124-3p miRNA may be regulators of signature genes. Patients with higher NRS have pathway enriched in fibrosis and metabolism and elevated nTreg cells. AZD-7762 may be an effective drug to improve the prognosis of patients with high NRS. A feature-based nomogram was constructed from which patients could derive clinical benefit. Conclusion: Our results reveal 6 necroptosis gene signatures that can evaluate the progression and prognosis of ICM with high clinical value, and identify potential targets that could help improve cardiovascular remodeling.
Collapse
Affiliation(s)
- Yang Lu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dashuai Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yaoxi Zhu
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yimei Du
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinying Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Han Yang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
49
|
Tu H, Xiong W, Zhang J, Zhao X, Lin X. Tyrosine phosphorylation regulates RIPK1 activity to limit cell death and inflammation. Nat Commun 2022; 13:6603. [PMID: 36329033 PMCID: PMC9632600 DOI: 10.1038/s41467-022-34080-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 10/13/2022] [Indexed: 11/05/2022] Open
Abstract
Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) is a cytosolic protein kinase that regulates multiple inflammatory and cell death pathways. Serine/Threonine phosphorylation of RIPK1 is known to suppress RIPK1 kinase-mediated cell death in the contexts of inflammation, infection and embryogenesis, however, regulation by tyrosine phosphorylation has not been reported. Here, we show that non-receptor tyrosine kinases Janus kinase 1 (JAK1) and SRC are able to phosphorylate RIPK1 at Y384 (Y383 in murine RIPK1), leading to suppression of TNF-induced cell death. Mice bearing a homozygous Ripk1 mutation that prevents tyrosine phosphorylation of RIPK1 (Ripk1Y383F/Y383F), develop systemic inflammation and emergency haematopoiesis. Mechanistically, Ripk1Y383F/Y383F mutation promotes RIPK1 kinase activation and enhances TNF-induced apoptosis and necroptosis, which is partially due to impaired recruitment and activation of MAP kinase-activated protein kinase 2 (MK2). The systemic inflammation and emergency haematopoiesis in Ripk1Y383F/Y383F mice are largely alleviated by RIPK1 kinase inhibition, and prevented by genomic deletions targeted to the upstream pathway (either to Tumor necrosis factor receptor 1 or RIPK3 and Caspase8 simultaneously). In summary, our results demonstrate that tyrosine phosphorylation of RIPK1 is critical for regulating RIPK1 activity to limit cell death and inflammation.
Collapse
Affiliation(s)
- Hailin Tu
- grid.12527.330000 0001 0662 3178Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
| | - Weihang Xiong
- grid.12527.330000 0001 0662 3178Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China ,grid.452723.50000 0004 7887 9190Tsinghua University–Peking University Center for Life Sciences, Beijing, 100084 China
| | - Jie Zhang
- grid.12527.330000 0001 0662 3178Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
| | - Xueqiang Zhao
- grid.12527.330000 0001 0662 3178Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
| | - Xin Lin
- grid.12527.330000 0001 0662 3178Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China ,grid.452723.50000 0004 7887 9190Tsinghua University–Peking University Center for Life Sciences, Beijing, 100084 China
| |
Collapse
|
50
|
Jayaraman A, Reynolds R. Diverse pathways to neuronal necroptosis in Alzheimer's disease. Eur J Neurosci 2022; 56:5428-5441. [PMID: 35377966 DOI: 10.1111/ejn.15662] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 03/21/2022] [Accepted: 03/29/2022] [Indexed: 12/14/2022]
Abstract
Necroptosis, or programmed necrosis, involves the kinase activity of receptor interacting kinases 1 and 3, the activation of the pseudokinase mixed lineage kinase domain-like and formation of a complex called the necrosome. It is one of the non-apoptotic cell death pathways that has gained interest in the recent years, especially as a neuronal cell death pathway occurring in Alzheimer's disease. In this review, we focus our discussion on the various molecular mechanisms that could trigger neuronal death through necroptosis and have been shown to play a role in Alzheimer's disease pathogenesis and neuroinflammation. We describe how each of these pathways, such as tumour necrosis factor signalling, reactive oxygen species, endosomal sorting complex, post-translational modifications and certain individual molecules, is dysregulated or activated in Alzheimer's disease, and how this dysregulation/activation could trigger necroptosis. At the cellular level, many of these molecular mechanisms and pathways may act in parallel to synergize with each other or inhibit one another, and changes in the balance between them may determine different cellular vulnerabilities at different disease stages. However, from a therapeutic standpoint, it remains unclear how best to target one or more of these pathways, given that such diverse pathways could all contribute to necroptotic cell death in Alzheimer's disease.
Collapse
Affiliation(s)
- Anusha Jayaraman
- Centre for Molecular Neuropathology, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Richard Reynolds
- Centre for Molecular Neuropathology, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.,Division of Neuroscience, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|