1
|
Ladiwala P, Ndahiro N, Hauk P, Wen J, Sargunas J, Chen YJ, Barton E, Betenbaugh MJ. Unraveling Cytotoxicity in HEK293 Cells During Recombinant AAV Production for Gene Therapy Applications. Biotechnol J 2025; 20:e202400501. [PMID: 40079705 DOI: 10.1002/biot.202400501] [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: 08/14/2024] [Revised: 01/05/2025] [Accepted: 01/23/2025] [Indexed: 03/15/2025]
Abstract
Transient transfection of HEK293 cells represents the dominant technique for the production of recombinant adeno-associated virus (AAV) vectors. However, recombinant AAV (rAAV) production is cytotoxic, potentially impacting process performance, product yields, and quality, complicating downstream processing. This study characterizes cell death response for rAAV producing HEK293 cells and explores the potential to control cytotoxicity. Initial analysis of triple transfected cells revealed caspase-mediated apoptosis as a likely mechanism of cellular death. Next, the causes of this cytotoxicity were investigated by dissecting transfection steps. Exposing cells to polyethyleneimine (PEI) alone or complexed with a blank plasmid at typical concentrations had a limited impact on cell growth. However, the inclusion of plasmid constructs containing genes to produce rAAVs triggered significant cell death, with the helper plasmid being the most toxic both independently and in combination with packaging and transgene plasmids. Additionally, apoptosis in transfected cultures could be inhibited using the pan-caspase inhibitor, N-benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone (Z-VAD.fmk), leading to a 65% increase in peak viable cell density (VCD). Although the rAAV genome titer remained relatively unaltered, capsid levels declined upon cell death inhibition. Consequently, the ratio of full to empty capsids, an important product quality attribute (PQA) for rAAVs increased following caspase inhibition. This study provides insights into apoptosis activation in rAAVs and uncovers avenues for its modulation to alter PQAs.
Collapse
Affiliation(s)
- Pranay Ladiwala
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Nelson Ndahiro
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Pricila Hauk
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Junneng Wen
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Justin Sargunas
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Yu-Ju Chen
- Pfizer Inc., Chesterfield, Missouri, USA
| | | | - Michael J Betenbaugh
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
2
|
Lien TS, Sun DS, Chang HH. Targeted Delivery to Dying Cells Through P-Selectin-PSGL-1 Axis: A Promising Strategy for Enhanced Drug Efficacy in Liver Injury Models. Cells 2024; 13:1778. [PMID: 39513885 PMCID: PMC11545035 DOI: 10.3390/cells13211778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/23/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
To minimize off-target adverse effects and improve drug efficacy, various tissue-specific drug delivery systems have been developed. However, even in diseased organs, both normal and stressed, dying cells coexist, and a targeted delivery system specifically for dying cells has yet to be explored to mitigate off-target effects within the same organ. This study aimed to establish such a system. By examining the surfaces of dying cells in vitro, we identified P-selectin glycoprotein ligand-1 (PSGL-1) as a universal marker for dying cells, positioning it as a potential target for selective drug delivery. We demonstrated that liposomes conjugated with the PSGL-1 binding protein P-selectin had significantly greater binding efficiency to dying cells compared to control proteins such as E-selectin, L-selectin, galectin-1, and C-type lectin-like receptor 2. Using thioacetamide (TAA) to induce hepatitis and hepatocyte damage in mice, we assessed the effectiveness of our P-selectin-based delivery system. In vivo, P-selectin-conjugated liposomes effectively delivered fluorescent dye and the apoptosis inhibitor z-DEVD to TAA-damaged livers in wild-type mice, but not in PSGL-1 knockout mice. In TAA-treated wild-type mice, unconjugated liposomes required a 100-fold higher z-DEVD dose compared to P-selectin-conjugated liposomes to achieve a comparable, albeit less effective, therapeutic outcome in lowering plasma alanine transaminase levels and alleviating thrombocytopenia. This emphasizes that P-selectin conjugation enhances drug delivery efficiency by approximately 100-fold in mice. These results suggest that P-selectin-based liposomes could be a promising strategy for targeted drug delivery, enabling both diagnosis and treatment by specifically delivering cell-labeling agents and rescue agents to dying cells via the P-selectin-PSGL-1 axis at the individual cell level.
Collapse
Grants
- 104-2320-B-320 -009 -MY3, 107-2311-B-320-002-MY3, 111-2320-B320-006-MY3, 112-2320-B-320-007 National Science and Technology Council, Taiwan
- TCMMP104-06, TCMMP108-04, TCMMP 111-01, TCAS111-02, TCAS-112-02, TCAS113-04, TCRD112-033, TCRD113-041 Tzu-Chi Medical Foundation
Collapse
Affiliation(s)
| | | | - Hsin-Hou Chang
- Department of Molecular Biology and Human Genetics, Tzu-Chi University, Hualien 970, Taiwan; (T.-S.L.); (D.-S.S.)
| |
Collapse
|
3
|
Bian Y, Shi J, Chen Z, Fang J, Chen W, Zou Y, Yao H, Tu J, Liao Y, Xie X, Shen J. A diagnostic signature developed based on the necroptosis-related genes and its association with immune infiltration in osteosarcoma. Heliyon 2024; 10:e35719. [PMID: 39253245 PMCID: PMC11381599 DOI: 10.1016/j.heliyon.2024.e35719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 09/11/2024] Open
Abstract
Introduction Osteosarcoma is a bone-derived malignancy that often leads to lung metastasis and death. Material and methods The RNA-seq data of TARGET-osteosarcoma were collected from TARGET database. GSE16088 and GSE12865 datasets of osteosarcoma x from Gene Expression Database (GEO) were donwloaded. ConsensusClusterPlus was used for molecular subtype classification. Univariate Cox and Lasso regression was employed to develop a risk model. To analyze the regulatory effects of model feature genes on the malignant phenotype of osteosarcoma cell lines, qRT-PCR, Transwell and wound healing assays were performed. The abundance of immune cell infiltration was assessed using MCP-Counter, Gene Set Enrichment Analysis (GSEA), and ESTIMATE. The Tumor Immune Dysfunction and Exclusion (TIDE) software was employed to evaluate immunotherapy and response to conventional chemotherapy drugs. Results Three clusters (C1, C2 and C3) were classified using 39 necroptosis score-associated genes. In general, C1 and C2 showed better prognosis outcome and lower death rate than C3. Specifically, C2 could benefit more from immunotherapy, while C3 was more sensitive to traditional medicines, and C1 had higher immune cell infiltration. Next, an 8-gene signature and a risk score model were developed, with a low risk score indicating better survival and immune cell infiltration. ROC analysis showed that 1-, 3-, and 5-year overall survival of osteosarcoma could be correctly predicted by the risk score model. Cellular experiments revealed that the model feature gene IFITM3 promoted the osteosarcoma cell migration and invasion. Furthermore, the overall survival of osteosarcoma patients from TARGET and validation datasets can be accurately evaluated using the nomogram model. Conclusions Our prognostic model developed using necroptosis genes could facilitate the prognostic prediction for patients suffering from osteosarcoma, offering potential osteosarcoma targets.
Collapse
Affiliation(s)
- Yiying Bian
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Jixiang Shi
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Ziyun Chen
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Ji Fang
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Weidong Chen
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Yutong Zou
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Hao Yao
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Jian Tu
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Yan Liao
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Xianbiao Xie
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Jingnan Shen
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| |
Collapse
|
4
|
Bae H, Jang Y, Karki R, Han JH. Implications of inflammatory cell death-PANoptosis in health and disease. Arch Pharm Res 2024; 47:617-631. [PMID: 38987410 DOI: 10.1007/s12272-024-01506-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 07/06/2024] [Indexed: 07/12/2024]
Abstract
Regulated cell death (RCD) pathways, such as pyroptosis, apoptosis, and necroptosis, are essential for maintaining the body's balance, defending against pathogens, and eliminating abnormal cells that could lead to diseases like cancer. Although these pathways operate through distinct mechanisms, recent genetic and pharmacological studies have shown that they can interact and influence each other. The concept of "PANoptosis" has emerged, highlighting the interplay between pyroptosis, apoptosis, and necroptosis, especially during cellular responses to infections. This article provides a concise overview of PANoptosis and its molecular mechanisms, exploring its implications in various diseases. The review focuses on the extensive interactions among different RCD pathways, emphasizing the role of PANoptosis in infections, cytokine storms, inflammatory diseases, and cancer. Understanding PANoptosis is crucial for developing novel treatments for conditions involving infections, sterile inflammations, and cancer.
Collapse
Affiliation(s)
- Hyun Bae
- Department of Biological Sciences, College of Natural Science, Seoul National University, Seoul, 08826, South Korea
| | - Yeonseo Jang
- Department of Biological Sciences, College of Natural Science, Seoul National University, Seoul, 08826, South Korea
| | - Rajendra Karki
- Department of Biological Sciences, College of Natural Science, Seoul National University, Seoul, 08826, South Korea.
- Nexus Institute of Research and Innovation (NIRI), Kathmandu, Nepal.
| | - Joo-Hui Han
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Woosuk University, Wanju, 55338, Republic of Korea.
| |
Collapse
|
5
|
He S, Silva LD, Rutter GA, Lim GE. A high-throughput screening approach to discover potential colorectal cancer chemotherapeutics: Repurposing drugs to disrupt 14-3-3 protein-BAD interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.14.571727. [PMID: 38168191 PMCID: PMC10760183 DOI: 10.1101/2023.12.14.571727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Inducing apoptosis in different types of cancer cells is an effective therapeutic strategy. However, the success of existing chemotherapeutics can be compromised by tumor cell resistance and systemic off-target effects. Therefore, the discovery of pro-apoptotic compounds with minimal systemic side-effects is crucial. 14-3-3 proteins are molecular scaffolds that serve as important regulators of cell survival. Our previous study demonstrated that 14-3-3ζ can sequester BAD, a pro-apoptotic member of the BCL-2 protein family, in the cytoplasm and prevent its translocation to mitochondria to inhibit the induction of apoptosis. Despite being a critical mechanism of cell survival, it is unclear whether disrupting 14-3-3 protein:BAD interactions could be harnessed as a chemotherapeutic approach. Herein, we established a BRET-based high-throughput drug screening approach (Z'-score= 0.52) capable of identifying molecules that can disrupt 14-3-3ζ:BAD interactions. An FDA-approved drug library containing 1971 compounds was used for screening, and the capacity of identified hits to induce cell death was examined in NIH3T3-fibroblasts and colorectal cancer cell lines, HT-29 and Caco-2. Our in vitro results suggest that terfenadine, penfluridol, and lomitapide could be potentially repurposed for treating colorectal cancer. Moreover, our screening method demonstrates the feasibility of identifying pro-apoptotic agents that can be applied towards conditions where aberrant cell growth or function are key determinants of disease pathogenesis.
Collapse
Affiliation(s)
- Siyi He
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
- Cardiometabolic axis, Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Luis Delgadillo Silva
- Cardiometabolic axis, Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Guy A. Rutter
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
- Cardiometabolic axis, Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Department of Diabetes, Endocrinology and Medicine, Faculty of Medicine, Imperial College, London, UK
- LKC School of Medicine, Nanyang Technological College, Singapore, Republic of Singapore
| | - Gareth E. Lim
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
- Cardiometabolic axis, Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, Québec, Canada
| |
Collapse
|
6
|
Noguchi S, Yasumura M. Potential therapeutic efficiency of pan-ERBB inhibitors for canine glioma. Vet Res Commun 2023; 47:2207-2213. [PMID: 36991174 DOI: 10.1007/s11259-023-10117-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: 01/07/2023] [Accepted: 03/23/2023] [Indexed: 03/31/2023]
Abstract
Canine glioma is one of the most common brain tumors with poor prognosis, making effective chemotherapy highly desirable. Previous studies have suggested that ERBB4, a signaling molecule involving one of the epidermal growth factor receptors (EGFR), may be a promising therapeutic target. In this study, the anti-tumor effects of pan-ERBB inhibitors, which can inhibit the phosphorylation of ERBB4, were evaluated both in vitro and in vivo using a canine glioblastoma cell line. The results demonstrated that both afatinib and dacomitinib effectively reduced the expression of phosphorylated ERBB4, and significantly decreased the number of viable cells, ultimately prolonging the survival time of orthotopically xenografted mice. Further downstream of ERBB4, afatinib was found to suppress the expression of phosphorylated Akt and phosphorylated Extracellular signal-related kinases1 and 2 (ERK1/2) and induced apoptotic cell death. Thus, pan-ERBB inhibition is a promising therapeutic strategy for the treatment of canine gliomas.
Collapse
Affiliation(s)
- Shunsuke Noguchi
- Laboratory of Veterinary Radiology, Graduate School of Veterinary Science, Osaka Metropolitan University, 1-58 Rinku Ourai Kita, Izumisano-shi, Osaka, 598-8531, Japan.
| | - Moeka Yasumura
- Laboratory of Veterinary Radiology, College of Life, Environment, and Advanced Sciences, Osaka Metropolitan University, 1-58 Rinku Ourai Kita, Izumisano-shi, Osaka, 598-8531, Japan
| |
Collapse
|
7
|
Matsuoka Y, Tsujimoto Y. Housing conditions affect enterocyte death mode and turnover rate in mouse small intestine. Sci Rep 2023; 13:20423. [PMID: 37993588 PMCID: PMC10665386 DOI: 10.1038/s41598-023-47660-1] [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: 03/24/2023] [Accepted: 11/16/2023] [Indexed: 11/24/2023] Open
Abstract
Small intestinal enterocytes are continuously renewed. Shedding/death of enterocytes involves receptor-interacting protein kinase 1 (RIPK1)-dependent (but RIPK3-independent) necrotic death, but the regulatory mechanism of the processes is not fully understood. Here, we show that mouse housing conditions, such as the type of bedding material and the presence or absence of a Shepherd Shack, affect enterocyte turnover rate and determine whether enterocyte shedding/death is RIPK1-independent or -dependent. Mice housed with ALPHA-dri (αDri, hard paper chip) bedding material without a Shepherd Shack had a higher, largely RIPK1-dependent enterocyte turnover rate and higher blood corticosterone levels, suggesting the involvement of minor stress, whereas mice housed with αDri plus a Shepherd Shack or with Soft Chip had a lower, RIPK1-independent turnover rate and lower blood corticosterone levels. Corticosterone administration to a small intestine culture derived from mice housed with αDri plus a Shepherd Shack or with Soft Chip increased enterocyte shedding/death and turnover. By using kinase inhibitors and knockout mice, we showed that the switch from RIPK1-independent to RIPK1-dependent enterocyte shedding/death and turnover involves suppression of TANK-binding kinase 1. Our results demonstrate that housing conditions may cause minor stress, which alters the mode of enterocyte shedding/death and enterocyte turnover rate in mice.
Collapse
Affiliation(s)
- Yosuke Matsuoka
- Department of Oncogenesis and Growth Regulation, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-ku, Osaka, 541-8567, Japan.
- Department of Molecular and Cellular Biology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-ku, Osaka, 541-8567, Japan.
| | - Yoshihide Tsujimoto
- Department of Molecular and Cellular Biology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-ku, Osaka, 541-8567, Japan.
| |
Collapse
|
8
|
Wu E, He W, Wu C, Chen Z, Zhou S, Wu X, Hu Z, Jia K, Pan J, Wang L, Qin J, Liu D, Lu J, Wang H, Li J, Wang S, Sun L. HSPA8 acts as an amyloidase to suppress necroptosis by inhibiting and reversing functional amyloid formation. Cell Res 2023; 33:851-866. [PMID: 37580406 PMCID: PMC10624691 DOI: 10.1038/s41422-023-00859-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 07/13/2023] [Indexed: 08/16/2023] Open
Abstract
Ultra-stable fibrous structure is a hallmark of amyloids. In contrast to canonical disease-related amyloids, emerging research indicates that a significant number of cellular amyloids, termed 'functional amyloids', contribute to signal transduction as temporal signaling hubs in humans. However, it is unclear how these functional amyloids are effectively disassembled to terminate signal transduction. RHIM motif-containing amyloids, the largest functional amyloid family discovered thus far, play an important role in mediating necroptosis signal transduction in mammalian cells. Here, we identify heat shock protein family A member 8 (HSPA8) as a new type of enzyme - which we name as 'amyloidase' - that directly disassembles RHIM-amyloids to inhibit necroptosis signaling in cells and mice. Different from its role in chaperone-mediated autophagy where it selects substrates containing a KFERQ-like motif, HSPA8 specifically recognizes RHIM-containing proteins through a hydrophobic hexapeptide motif N(X1)φ(X3). The SBD domain of HSPA8 interacts with RHIM-containing proteins, preventing proximate RHIM monomers from stacking into functional fibrils; furthermore, with the NBD domain supplying energy via ATP hydrolysis, HSPA8 breaks down pre-formed RHIM-amyloids into non-functional monomers. Notably, HSPA8's amyloidase activity in disassembling functional RHIM-amyloids does not require its co-chaperone system. Using this amyloidase activity, HSPA8 reverses the initiator RHIM-amyloids (formed by RIP1, ZBP1, and TRIF) to prevent necroptosis initiation, and reverses RIP3-amyloid to prevent necroptosis execution, thus eliminating multi-level RHIM-amyloids to effectively prevent spontaneous necroptosis activation. The discovery that HSPA8 acts as an amyloidase dismantling functional amyloids provides a fundamental understanding of the reversibility nature of functional amyloids, a property distinguishing them from disease-related amyloids that are unbreakable in vivo.
Collapse
Affiliation(s)
- Erpeng Wu
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Wenyan He
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Chenlu Wu
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Zhangcheng Chen
- State Key Laboratory of Molecular Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China; Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Shijie Zhou
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Xialian Wu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Zhiheng Hu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Kelong Jia
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Jiasong Pan
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Huashan Hospital, Shanghai Engineering Research Center of Industrial Microorganisms, Fudan University, Shanghai, China
| | - Limin Wang
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Jie Qin
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Dan Liu
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Junxia Lu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Huayi Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Jixi Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Huashan Hospital, Shanghai Engineering Research Center of Industrial Microorganisms, Fudan University, Shanghai, China
| | - Sheng Wang
- State Key Laboratory of Molecular Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China; Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Liming Sun
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
9
|
Petersen M, Chorzalska A, Pardo M, Rodriguez A, Morgan J, Ahsan N, Zhao TC, Liang O, Kotula L, Bertone P, Gruppuso PA, Dubielecka PM. Proximity proteomics reveals role of Abelson interactor 1 in the regulation of TAK1/RIPK1 signaling. Mol Oncol 2023; 17:2356-2379. [PMID: 36635880 PMCID: PMC10620119 DOI: 10.1002/1878-0261.13374] [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: 11/30/2022] [Accepted: 01/05/2023] [Indexed: 01/14/2023] Open
Abstract
Dysregulation of the adaptor protein Abelson interactor 1 (ABI1) is linked to malignant transformation. To interrogate the role of ABI1 in cancer development, we mapped the ABI1 interactome using proximity-dependent labeling (PDL) with biotin followed by mass spectrometry. Using a novel PDL data filtering strategy, considering both peptide spectral matches and peak areas of detected peptides, we identified 212 ABI1 proximal interactors. These included WAVE2 complex components such as CYFIP1, NCKAP1, or WASF1, confirming the known role of ABI1 in the regulation of actin-polymerization-dependent processes. We also identified proteins associated with the TAK1-IKK pathway, including TAK1, TAB2, and RIPK1, denoting a newly identified function of ABI1 in TAK1-NF-κB inflammatory signaling. Functional assays using TNFα-stimulated, ABI1-overexpressing or ABI1-deficient cells showed effects on the TAK1-NF-kB pathway-dependent signaling to RIPK1, with ABI1-knockout cells being less susceptible to TNFα-induced, RIPK1-mediated, TAK1-dependent apoptosis. In sum, our PDL-based strategy enabled mapping of the ABI1 proximal interactome, thus revealing a previously unknown role of this adaptor protein in TAK1/RIPK1-based regulation of cell death and survival.
Collapse
Affiliation(s)
- Max Petersen
- Department of Medicine, Alpert Medical SchoolBrown UniversityProvidenceRIUSA
- Division of Hematology/OncologyRhode Island HospitalProvidenceRIUSA
- Division of Biology and Medicine, Department of Pathology and Laboratory MedicineBrown UniversityProvidenceRIUSA
| | - Anna Chorzalska
- Department of Medicine, Alpert Medical SchoolBrown UniversityProvidenceRIUSA
- Division of Hematology/OncologyRhode Island HospitalProvidenceRIUSA
| | - Makayla Pardo
- Department of Medicine, Alpert Medical SchoolBrown UniversityProvidenceRIUSA
- Division of Hematology/OncologyRhode Island HospitalProvidenceRIUSA
| | - Anaelena Rodriguez
- Department of Medicine, Alpert Medical SchoolBrown UniversityProvidenceRIUSA
- Division of Hematology/OncologyRhode Island HospitalProvidenceRIUSA
| | - John Morgan
- Flow Cytometry and Cell Sorting Core FacilityRoger Williams Medical CenterProvidenceRIUSA
| | - Nagib Ahsan
- COBRE Center for Cancer Research Development, Proteomics Core FacilityRhode Island HospitalProvidenceRIUSA
- Department of Chemistry and BiochemistryThe University of OklahomaNormanOKUSA
- Mass Spectrometry, Proteomics and Metabolomics Core Facility, Stephenson Life Sciences Research CenterThe University of OklahomaNormanOKUSA
| | - Ting C. Zhao
- Department of SurgeryRhode Island Hospital and Warren Alpert Medical School of Brown UniversityProvidenceRIUSA
| | - Olin Liang
- Department of Medicine, Alpert Medical SchoolBrown UniversityProvidenceRIUSA
- Division of Hematology/OncologyRhode Island HospitalProvidenceRIUSA
- Legorreta Cancer Center, Alpert Medical SchoolBrown UniversityProvidenceRIUSA
| | - Leszek Kotula
- Department of UrologySUNY Upstate Medical UniversitySyracuseNYUSA
- Department of Biochemistry and Molecular BiologySUNY Upstate Medical UniversitySyracuseNYUSA
- Upstate Cancer CenterSUNY Upstate Medical UniversitySyracuseNYUSA
| | - Paul Bertone
- Department of Medicine, Alpert Medical SchoolBrown UniversityProvidenceRIUSA
- Division of Hematology/OncologyRhode Island HospitalProvidenceRIUSA
- Legorreta Cancer Center, Alpert Medical SchoolBrown UniversityProvidenceRIUSA
| | - Philip A. Gruppuso
- Division of Pediatric EndocrinologyRhode Island Hospital and Warren Alpert Medical School of Brown UniversityProvidenceRIUSA
| | - Patrycja M. Dubielecka
- Department of Medicine, Alpert Medical SchoolBrown UniversityProvidenceRIUSA
- Division of Hematology/OncologyRhode Island HospitalProvidenceRIUSA
- Division of Biology and Medicine, Department of Pathology and Laboratory MedicineBrown UniversityProvidenceRIUSA
- Legorreta Cancer Center, Alpert Medical SchoolBrown UniversityProvidenceRIUSA
| |
Collapse
|
10
|
Gupta P, Singh A, Verma AK, Kant S, Pandey AK, Mishra A, Khare P, Prakash V. Nanoencapsulation of Docetaxel Induces Concurrent Apoptosis and Necroptosis in Human Oral Cancer Cells (SCC-9) via TNF-α/RIP1/RIP3 Pathway. Indian J Clin Biochem 2023; 38:351-360. [PMID: 37234186 PMCID: PMC10205939 DOI: 10.1007/s12291-022-01055-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/22/2022] [Indexed: 11/29/2022]
Abstract
Human oral squamous cell carcinoma is the sixth most frequent malignant cancer, with an unacceptably high death rate that affects people's health. Albeit, there are several clinical approaches for diagnosing and treating oral cancer they are still far from ideal. We previously synthesised and characterised the docetaxel nanoformulation (PLGA-Dtx) and discovered that docetaxel nanoencapsulation may suppress oral cancer cells. The goal of this study was to figure out the mechanism involved in the suppression of oral cancer cell proliferation. We discovered that PLGA-Dtx inhibited SCC-9 cell growth considerably as compared to free docetaxel (Dtx), and that the viability of SCC-9 cells treated with PLGA-Dtx was decreased dose-dependently. MTT assay showed that PLGA-Dtx selectively inhibited the growth of PBMCs from oral cancer patients while sparing PBMCs from normal healthy controls. Further, flow cytometry analysis showed that PLGA-Dtx induced apoptosis and necroptosis in SCC-9 cells. G2/M cell cycle arrest has been confirmed on exposure of PLGA-Dtx for 24 h in SCC-9 cells. Interestingly, western blot investigation found that PLGA-Dtx increased the amounts of necroptic proteins and apoptosis-related proteins more efficiently than Dtx. Furthermore, PLGA-Dtx was more effective in terms of ROS generation, and mitochondrial membrane potential depletion. Pretreatment with necroptosis inhibitor Nec-1 efficiently reversed the ROS production and further recover MMP caused by PLGA-Dtx. Overall, this study revealed a mechanistic model of therapeutic response for PLGA-Dtx in SCC-9 cells and proposed its potency by inducing cell death via activation of concurrent apoptosis and necroptosis in SCC-9 cells via TNF-α/RIP1/RIP3 and caspase-dependent pathway.
Collapse
Affiliation(s)
- Parul Gupta
- Department of Respiratory Medicine, King George’s Medical University, Lucknow, Uttar Pradesh 226003 India
| | - Arpita Singh
- Department of Pharmacology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh 226010 India
| | - Ajay Kumar Verma
- Department of Respiratory Medicine, King George’s Medical University, Lucknow, Uttar Pradesh 226003 India
| | - Surya Kant
- Department of Respiratory Medicine, King George’s Medical University, Lucknow, Uttar Pradesh 226003 India
| | - Anuj Kumar Pandey
- Department of Respiratory Medicine, King George’s Medical University, Lucknow, Uttar Pradesh 226003 India
| | - Anupam Mishra
- Department of Otorhinolaryngology & Head Neck Surgery, King George’s Medical University, Lucknow, Uttar Pradesh 226003 India
| | - Puneet Khare
- Flow Cytometry Facility, Central Instrumentation Facility, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh 226001 India
| | - Ved Prakash
- Department of Pulmonary & Critical Care Medicine, King George’s Medical University, Lucknow, Uttar Pradesh 226003 India
| |
Collapse
|
11
|
Lee CS, Hwang G, Nam YW, Hwang CH, Song J. IKK-mediated TRAF6 and RIPK1 interaction stifles cell death complex assembly leading to the suppression of TNF-α-induced cell death. Cell Death Differ 2023; 30:1575-1584. [PMID: 37085671 PMCID: PMC10244383 DOI: 10.1038/s41418-023-01161-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/23/2023] Open
Abstract
Tumor necrosis factor α (TNF-α) is a pro-inflammatory cytokine capable of inducing extrinsic apoptosis and necroptosis. Tumor necrosis factor receptor-associated factor 6 (TRAF6), an E3 ligase, is a member of the TRAF family of proteins, which mediates inflammatory signals by activating nuclear factor kappa B (NFкB) and mitogen-activated protein kinase (MAPK). Although the functions of TRAF6 have been identified, its role in TNF-α-induced cell death remains poorly understood. Here, we report that TRAF6 is a negative modulator of TNF-α-induced cell death but does not affect TNF-α-induced NFκB activation. TRAF6 deficiency accelerates both TNF-α-induced apoptosis and necroptosis; however, the acceleration can be reversed by reconstituting TRAF6 or TRAF6C70A, suggesting that E3 ligase activity is not required for this activity. Mechanistically, TRAF6 directly interacts with RIPK1 during TNF-α-induced cell death signaling, which prevents RIPK1 from interacting with components of the cell death complex such as itself, FADD or RIPK3. These processes suppress the assembly of the death complex. Notably, IKK was required for TRAF6 to interact with RIPK1. In vivo, Traf6-/- embryos exhibited higher levels of cell death in the liver but could be rescued by the simultaneous knockout of Tnf. Finally, TRAF6 knockdown xenografts were highly sensitive to necroptotic stimuli. We concluded that TRAF6 suppresses TNF-α-induced cell death in coordination with IKK complexes in vivo and in vitro by suppressing the assembly of cell death complex.
Collapse
Affiliation(s)
- Choong-Sil Lee
- Integrated OMICS for Biomedical Science, Yonsei University, Seoul, 03722, Korea
| | - Gyuho Hwang
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Young Woo Nam
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Chi Hyun Hwang
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Jaewhan Song
- Integrated OMICS for Biomedical Science, Yonsei University, Seoul, 03722, Korea.
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea.
| |
Collapse
|
12
|
Keramidas P, Papachristou E, Papi RM, Mantsou A, Choli-Papadopoulou T. Inhibition of PERK Kinase, an Orchestrator of the Unfolded Protein Response (UPR), Significantly Reduces Apoptosis and Inflammation of Lung Epithelial Cells Triggered by SARS-CoV-2 ORF3a Protein. Biomedicines 2023; 11:1585. [PMID: 37371681 DOI: 10.3390/biomedicines11061585] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/25/2023] [Accepted: 05/27/2023] [Indexed: 06/29/2023] Open
Abstract
SARS-CoV-2 ORF3a accessory protein was found to be involved in virus release, immunomodulation and exhibited a pro-apoptotic character. In order to unravel a potential ORF3a-induced apoptotic and inflammatory death mechanism, lung epithelial cells (A549) were transfected with in vitro synthesized ORF3a mRNA. The protein's dynamic involvement as "stress factor" for the endoplasmic reticulum, causing the activation of PERK kinase and other UPR-involved proteins and therefore the upregulation of their signaling pathway executioners (ATF6, XBP-1s, PERK, phospho eIF2a, ATF4, CHOP, GADD34), has been clearly demonstrated. Furthermore, the overexpression of BAX and BH3-only pro-apoptotic protein PUMA, the upregulation of Bcl-2 family genes (BAX, BAK, BID, BAD), the reduced expression of Bcl-2 in mRNA and protein levels, and lastly, the cleavage of PARP-1 and caspase family members (caspase-3,-8 and -9) indicate that ORF3a displays its apoptotic character through the mitochondrial pathway of apoptosis. Moreover, the upregulation of NFκB, phosphorylation of p65 and IκΒα and the elevated expression of pro-inflammatory cytokines (IL-1b, IL-6, IL-8 and IL-18) in transfected cells with ORF3a mRNA indicate that this protein causes the inflammatory response through NFκB activation and therefore triggers lung injury. An intriguing finding of our study is that upon treatment of the ORF3a-transfected cells with GSK2606414, a selective PERK inhibitor, both complications (apoptosis and inflammatory response) were neutralized, and cell survival was favored, whereas treatment of transfected cells with z-VAD (a pan-caspase inhibitor) despite inhibiting cell death, could not ameliorate the inflammatory response of transfected A549 cells. Given the above, we point out that PERK kinase is a "master tactician" and its activation constitutes the main stimulus for the emergence of ORF3a apoptotic and inflammatory nature and therefore could serve as potential target for developing novel therapeutic approaches against COVID-19.
Collapse
Affiliation(s)
- Panagiotis Keramidas
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Eleni Papachristou
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Rigini M Papi
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Aglaia Mantsou
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Theodora Choli-Papadopoulou
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| |
Collapse
|
13
|
Avlas S, Kassis H, Itan M, Reichman H, Dolitzky A, Hazut I, Grisaru-Tal S, Gordon Y, Tsarfaty I, Karo-Atar D, Rozenberg P, Bitton A, Munitz A. CD300b regulates intestinal inflammation and promotes repair in colitis. Front Immunol 2023; 14:1050245. [PMID: 37033950 PMCID: PMC10073762 DOI: 10.3389/fimmu.2023.1050245] [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] [Accepted: 03/07/2023] [Indexed: 04/11/2023] Open
Abstract
Chronic inflammation is a hallmark charataristic of various inflammatory diseases including inflammatory bowel disease. Subsequently, current therapeutic approaches target immune-mediated pathways as means for therapeutic intervention and promotion of mucosal healing and repair. Emerging data demonstrate important roles for CD300 receptor family members in settings of innate immunity as well as in allergic and autoimmune diseases. One of the main pathways mediating the activities of CD300 family members is via promotion of resolution through interactions with ligands expressed by viruses, bacteria, or dead cells (e.g., phospholipids such as PtdSer and/or ceramide). We have recently shown that the expression of CD300a, CD300b and CD300f were elevated in patients with IBD and that CD300f (but not CD300a) regulates colonic inflammation in response to dextran sodium sulphate (DSS)-induced colitis. Whether CD300b has a role in colitis or mucosal healing is largely unknown. Herein, we demonstrate a central and distinct role for CD300b in colonic inflammation and subsequent repair. We show that Cd300b-/- mice display defects in mucosal healing upon cessation of DSS treatment. Cd300b-/- mice display increased weight loss and disease activity index, which is accompanied by increased colonic histopathology, increased infiltration of inflammatory cells and expression of multiple pro-inflammatory upon cessation of DSS cytokines. Furthermore, we demonstrate that soluble CD300b (sCD300b) is increased in the colons of DSS-treated mice and establish that CD300b can bind mouse and human epithelial cells. Finally, we show that CD300b decreases epithelial EpCAM expression, promotes epithelial cell motility and wound healing. These data highlight a key role for CD300b in colonic inflammation and repair processes and suggest that CD300b may be a future therapeutic target in inflammatory GI diseases.
Collapse
|
14
|
Ferreira AC, Sacramento CQ, Pereira-Dutra FS, Fintelman-Rodrigues N, Silva PP, Mattos M, de Freitas CS, Marttorelli A, de Melo GR, Campos MM, Azevedo-Quintanilha IG, Carlos AS, Emídio JV, Garcia CC, Bozza PT, Bozza FA, Souza TML. Severe influenza infection is associated with inflammatory programmed cell death in infected macrophages. Front Cell Infect Microbiol 2023; 13:1067285. [PMID: 36875528 PMCID: PMC9980436 DOI: 10.3389/fcimb.2023.1067285] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 02/03/2023] [Indexed: 02/18/2023] Open
Abstract
Introduction Influenza A virus (IAV) is one of the leading causes of respiratory tract infections in humans, representing a major public health concern. The various types of cell death have a crucial role in IAV pathogenesis because this virus may trigger both apoptosis and necroptosis in airway epithelial cells in parallel. Macrophages play an important role in the clearance of virus particles, priming the adaptive immune response in influenza. However, the contribution of macrophage death to pathogenesis of IAV infection remains unclear. Methods In this work, we investigated IAV-induced macrophage death, along with potential therapeutic intervention. We conducted in vitro and in vivo experiments to evaluate the mechanism and the contribution of macrophages death to the inflammatory response induced by IAV infection. Results We found that IAV or its surface glycoprotein hemagglutinin (HA) triggers inflammatory programmed cell death in human and murine macrophages in a Toll-like receptor-4 (TLR4)- and TNF-dependent manner. Anti-TNF treatment in vivo with the clinically approved drug etanercept prevented the engagement of the necroptotic loop and mouse mortality. Etanercept impaired the IAV-induced proinflammatory cytokine storm and lung injury. Conclusion In summary, we demonstrated a positive feedback loop of events that led to necroptosis and exacerbated inflammation in IAV-infected macrophages. Our results highlight an additional mechanism involved in severe influenza that could be attenuated with clinically available therapies.
Collapse
Affiliation(s)
- André C. Ferreira
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fundação Oswado Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
- National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Fundação Oswado Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
- Preclinical Research Laboratory, Universidade Iguaçu (UNIG), Nova Iguaçu, RJ, Brazil
| | - Carolina Q. Sacramento
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fundação Oswado Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
- National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Fundação Oswado Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
| | - Filipe S. Pereira-Dutra
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fundação Oswado Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
| | - Natália Fintelman-Rodrigues
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fundação Oswado Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
- National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Fundação Oswado Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
| | - Priscila P. Silva
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fundação Oswado Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
- Preclinical Research Laboratory, Universidade Iguaçu (UNIG), Nova Iguaçu, RJ, Brazil
| | - Mayara Mattos
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fundação Oswado Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
- National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Fundação Oswado Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
| | - Caroline S. de Freitas
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fundação Oswado Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
- National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Fundação Oswado Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
| | - Andressa Marttorelli
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fundação Oswado Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
- National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Fundação Oswado Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
| | - Gabrielle R. de Melo
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fundação Oswado Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
| | - Mariana M. Campos
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fundação Oswado Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
| | | | - Aluana S. Carlos
- Preclinical Research Laboratory, Universidade Iguaçu (UNIG), Nova Iguaçu, RJ, Brazil
| | - João Vítor Emídio
- Preclinical Research Laboratory, Universidade Iguaçu (UNIG), Nova Iguaçu, RJ, Brazil
| | - Cristiana C. Garcia
- Respiratory and Measles Virus Laboratory, Oswaldo Cruz Institute, Fundação Oswado Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
| | - Patrícia T. Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fundação Oswado Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
| | - Fernando A. Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fundação Oswado Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
- National Institute of Infectious Disease Evandro Chagas, Fundação Oswado Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
- Department of Critical Care, Instituto D’Or de Pesquisa e Ensino (IDOR), Rio de Janeiro, RJ, Brazil
| | - Thiago M. L. Souza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fundação Oswado Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
- National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Fundação Oswado Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
- *Correspondence: Thiago M. L. Souza,
| |
Collapse
|
15
|
Li X, Zhang Y, Wang J, Li Y, Wang Y, Shi F, Hong L, Li L, Diao H. zVAD alleviates experimental autoimmune hepatitis in mice by increasing the sensitivity of macrophage to TNFR1-dependent necroptosis. J Autoimmun 2022; 133:102904. [PMID: 36108506 DOI: 10.1016/j.jaut.2022.102904] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 08/23/2022] [Accepted: 08/26/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Autoimmune hepatitis (AIH) is characterized by hepatocyte destruction, leading to lymphocyte and macrophage accumulation in the liver. Macrophages are key drivers of AIH. A membrane-permeable pan-caspase inhibitor, Z-Val-Ala-DL-Asp-fluoromethylketone (zVAD), induces macrophage necroptosis in response to certain stimuli. However, the function of zVAD in the pathogenesis of autoimmune hepatitis remains elusive. In this study, we aimed to evaluate the effect and explore the underlying mechanisms of zVAD against AIH. METHODS Murine acute autoimmune liver injury was established by concanavalin A (ConA) injection. Bone marrow-derived macrophages (BMDMs) were used in adoptive cell transfer experiments. The mechanism of action of zVAD was examined using macrophage cell lines and BMDMs. Phosphorylation of mixed lineage kinase domain-like proteins was used as a marker of necroptosis. RESULTS Treatment with zVAD increased necroptosis, reduced inflammatory cytokine production, and alleviated liver injury in a ConA-induced liver injury mouse model. Regardless of zVAD treatment, macrophage deletion resulted in reduced neutrophil accumulation and relieved ConA-induced liver injury. In vitro studies have shown that zVAD pretreatment promotes lipopolysaccharide-induced macrophage necroptosis and leads to reduced pro-inflammatory cytokine and chemokine secretion. Transferring zVAD-pretreated BMDMs in mice notably reduced ConA-associated liver inflammation and injury, resulting in lower mortality than that observed after transferring normal BMDMs. Mechanistically, zVAD treatment increased the expression of tumour necrosis factor receptor (TNFR)-1 and interleukin (IL)-10 in macrophages. TNFR1 expression decreased upon transfection with IL-10-specific small interfering RNAs and blocking of TNFR1 decreased macrophage necroptosis. CONCLUSIONS We found that zVAD alleviated ConA-induced liver injury by increasing the sensitivity of macrophages to necroptosis via IL-10-induced TNFR1 expression. This study provides new insights into the treatment of autoimmune hepatitis via zVAD-induced macrophage necroptosis.
Collapse
Affiliation(s)
- Xuehui Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongting Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinping Wang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yuyu Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuchong Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fan Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liang Hong
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China.
| | - Hongyan Diao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
16
|
Gupta P, Singh A, Verma AK, Kant S, Pandey AK, Khare P, Prakash V. The Anti-Tumor and Immunomodulatory Effects of PLGA-Based Docetaxel Nanoparticles in Lung Cancer: The Potential Involvement of Necroptotic Cell Death through Reactive Oxygen Species and Calcium Build-Up. Vaccines (Basel) 2022; 10:1801. [PMID: 36366309 PMCID: PMC9694303 DOI: 10.3390/vaccines10111801] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/05/2022] [Accepted: 10/19/2022] [Indexed: 04/23/2025] Open
Abstract
Taxanes, microtubule stabilizing agents, are extensively used in the treatment of non-small cell lung cancer (NSCLC). However, their clinical effectiveness remains restricted owing to significant adverse effects and drug resistance. Nanotechnology may guide chemotherapeutic drugs directly and selectively to malignant cells, improving their therapeutic efficiency. In the present study, we synthesized polylactic-co-glycolic acid (PLGA) based nanoparticles encapsulating docetaxel and evaluated their efficacy in non-small cell lung carcinoma (A549) cells and primary immune cells derived from humans. Docetaxel-PLGA nanoparticles (PLGA-Dtx) were synthesized and characterized using distinct methods. Moreover, the cytotoxicity of free docetaxel (Dtx) and Dtx-conjugated nanoparticles (PLGA-Dtx) was studied in A549 cells and peripheral blood mononuclear cells derived from humans. Furthermore, annexin V-FITC/PI staining was used to assess the mode of cell death. Additionally, human peripheral blood mononuclear cells (PBMCs) were used for assessing the associated immune response and cytokine profile following PLGA-Dtx treatment. Spherical PLGA-Dtx nanoparticles with a 150 ± 10 nm diameter and 70% encapsulation efficiency (EE) were synthesized. The MTT assay showed that the IC50 of PLGA-Dtx nanoparticles was significantly lower than free docetaxel in A549 cells. Cytotoxicity data also revealed the selective nature of PLGA-Dtx with no significant effects in normal human bronchial epithelial cells (BEAS-2B) and PBMCs derived from healthy donors. Interestingly, PLGA-Dtx exerted an improved effect and tempted both apoptosis and necroptosis, as evidenced by annexin V and propidium iodide-positive cells. Further, PLGA-Dtx-exposed A549 cells showed increased Cas-3, Cas-9, RIP-1, and RIP-3, indicating apoptosis and necroptosis. An increased pro-inflammatory response manifested from the enhancement of IFN-γ and TNF-α in PLGA-Dtx-exposed PBMCs, posed by the occurrence of necroptosis and the immune stimulatory effect of PLGA-Dtx. In conclusion, PLGA-Dtx has a selective anticancer potential and better immunostimulatory potential. Therefore, PLGA-Dtx may be useful for the treatment of non-small cell lung carcinoma.
Collapse
Affiliation(s)
- Parul Gupta
- Department of Respiratory Medicine, King George’s Medical University, Lucknow 226003, Uttar Pradesh, India
| | - Arpita Singh
- Department of Pharmacology, Dr. Ram Manohar Lohia, Institute of Medical Sciences, Lucknow 226010, Uttar Pradesh, India
| | - Ajay Kumar Verma
- Department of Respiratory Medicine, King George’s Medical University, Lucknow 226003, Uttar Pradesh, India
| | - Surya Kant
- Department of Respiratory Medicine, King George’s Medical University, Lucknow 226003, Uttar Pradesh, India
| | - Anuj Kumar Pandey
- Department of Respiratory Medicine, King George’s Medical University, Lucknow 226003, Uttar Pradesh, India
| | - Puneet Khare
- Flow Cytometry Facility, Central Instrumentation Facility, CSIR-Indian Institute of Toxicology Research, Lucknow 226001, Uttar Pradesh, India
| | - Ved Prakash
- Department of Pulmonary & Critical Care Medicine, King George’s Medical University, Lucknow 226003, Uttar Pradesh, India
| |
Collapse
|
17
|
Abdelbary M, Mohamed R, Gillis EE, Diaz-Sanders K, Baban B, Brands MW, Sullivan JC. Sex differences in apoptosis do not contribute to sex differences in blood pressure or renal T cells in spontaneously hypertensive rats. Front Physiol 2022; 13:1006951. [PMID: 36304583 PMCID: PMC9592703 DOI: 10.3389/fphys.2022.1006951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/23/2022] [Indexed: 11/29/2022] Open
Abstract
Apoptosis is a physiological and anti-inflammatory form of cell death that is indispensable for normal physiology and homeostasis. Several studies have reported aberrant activation of apoptosis in various tissues at the onset of hypertension. However, the functional significance of apoptosis during essential hypertension remains largely undefined. The current study was designed to test the hypothesis that apoptosis contributes to sex differences in blood pressure and the T cell profile in spontaneously hypertensive rats (SHR). Apoptosis was measured in kidney, aorta and spleen of 13-week-old adult hypertensive male and female SHR. Female SHR had greater renal and aortic apoptosis compared to age-matched males; apoptosis in the spleen was comparable between the sexes. Based on well-established sex differences in hypertension, we tested the hypothesis that greater apoptosis in female SHR contributes to the lower BP and pro-inflammatory profile compared to males. Male and female SHR were randomized to receive vehicle or ZVAD-FMK, a cell permeable pan-caspase inhibitor, in established hypertension from 13 to 15 weeks of age or at the onset of hypertension from 6 to 12 weeks or age. Treatment with ZVAD-FMK lowered renal apoptosis in both studies, yet neither BP nor renal T cells were altered in either male or female SHR. These results suggest that apoptosis does not contribute to the control or maintenance of BP in male or female SHR or sex differences in renal T cells.
Collapse
Affiliation(s)
- Mahmoud Abdelbary
- Department of Physiology, Augusta University, Augusta, GA, United States
| | - Riyaz Mohamed
- Department of Physiology, Augusta University, Augusta, GA, United States
| | - Ellen E. Gillis
- Department of Physiology, Augusta University, Augusta, GA, United States
| | - Karl Diaz-Sanders
- Department of Physiology, Augusta University, Augusta, GA, United States
| | - Babak Baban
- Department of Oral Biology, Augusta University, Augusta, GA, United States
| | - Michael W. Brands
- Department of Physiology, Augusta University, Augusta, GA, United States
| | - Jennifer C. Sullivan
- Department of Physiology, Augusta University, Augusta, GA, United States,*Correspondence: Jennifer C. Sullivan,
| |
Collapse
|
18
|
RIPK3 inhibitor-AZD5423 alleviates acute kidney injury by inhibiting necroptosis and inflammation. Int Immunopharmacol 2022; 112:109262. [PMID: 36166972 DOI: 10.1016/j.intimp.2022.109262] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/07/2022] [Accepted: 09/16/2022] [Indexed: 11/20/2022]
Abstract
Acute kidney injury (AKI) is a clinical syndrome that is defined as a sudden decline in renal function and characterized by inflammation and programmed cell death of renal tubular epithelial cells. Necroptosis is a form of regulated cell death that requires activation of receptor interacting protein kinase 3 (RIPK3) and its phosphorylation of the substrate MLKL. RIPK3 plays an important role in acute kidney injury, and hence developing its inhibitors is considered as one of the promising strategies aimed at prevention and treatment of AKI. Recently, we discovered AZD5423 as a novel potent RIPK3 inhibitor using a computer-aided hybrid virtual screening strategy according to three-dimensional structure of RIPK3. Our findings revealed that AZD5423 strongly inhibits activation of RIPK3, and MLKL phosphorylation upon cisplatin-, hypoxia/reoxygenation (H/R)- and TNF-α stimuli as compared with GSK872, which is a previously identified RIPK3 inhibitor. Importantly, AZD5423 exerts effective protection against cisplatin- and ischemia/reperfusion (I/R)-induced AKI mouse model. The results of cellular thermal shift assay and experiments in RIPK3 knockout cells indicated that AZD5423 could directly target RIPK3 to inhibit RIPK3 kinase activity. Mechanistically, the docking of AZD5423 and RIPK3 suggested that the kinase domain of RIPK3 for Lys50, Arg313, Lys29, Arg37 might form hydrogen bonds with AZD5423. Site-directed mutagenesis further revealed that AZD5423 reduces injury response via interacting with the key RIPK3 amino acid residues of Lys50 and Arg313. In conclusion, our study has demonstrated that AZD5423 may serve as a potent inhibitor of RIPK3 kinase and a promising clinical candidate for AKI treatment.
Collapse
|
19
|
The regulation of necroptosis and perspectives for the development of new drugs preventing ischemic/reperfusion of cardiac injury. Apoptosis 2022; 27:697-719. [DOI: 10.1007/s10495-022-01760-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2022] [Indexed: 12/11/2022]
|
20
|
Han X, Li B, Bao J, Wu Z, Chen C, Ni J, Shen J, Song P, Peng Q, Wan R, Wang X, Wu J, Hu G. Endoplasmic reticulum stress promoted acinar cell necroptosis in acute pancreatitis through cathepsinB-mediated AP-1 activation. Front Immunol 2022; 13:968639. [PMID: 36059491 PMCID: PMC9438943 DOI: 10.3389/fimmu.2022.968639] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/01/2022] [Indexed: 11/25/2022] Open
Abstract
Acinar cell death and inflammatory response are two important events which determine the severity of acute pancreatitis (AP). Endoplasmic reticulum (ER) stress and necroptosis are involved in this process, but the relationships between them remain unknown. Here, we analyzed the interaction between ER stress and necroptosis and the underlying mechanisms during AP. Experimental pancreatitis was induced in Balb/C mice by caerulein (Cae) and lipopolysaccharide (LPS) or L-arginine (L-Arg) in vivo, and pancreatic acinar cells were also used to follow cellular mechanisms during cholecystokinin (CCK) stimulation in vitro. AP severity was assessed by serum amylase, lipase levels and histological examination. Changes in ER stress, trypsinogen activation and necroptosis levels were analyzed by western blotting, enzyme-linked immunosorbent assay (ELISA), adenosine triphosphate (ATP) analysis or lactate dehydrogenase (LDH) assay. The protein kinase C (PKC)α -mitogen-activated protein kinase (MAPK) -cJun pathway and cathepsin B (CTSB) activation were evaluated by western blotting. Activating protein 1 (AP-1) binding activity was detected by electrophoretic mobility shift assay (EMSA). We found that ER stress is initiated before necroptosis in CCK-stimulated acinar cells in vitro. Inhibition of ER stress by 4-phenylbutyrate (4-PBA) can significantly alleviate AP severity both in two AP models in vivo. 4-PBA markedly inhibited ER stress and necroptosis of pancreatic acinar cells both in vitro and in vivo. Mechanistically, we found that 4-PBA significantly reduced CTSB maturation and PKCα-JNK-cJun pathway -mediated AP-1 activation during AP. Besides, CTSB inhibitor CA074Me markedly blocked PKCα-JNK-cJun pathway -mediated AP-1 activation and necroptosis in AP. However, pharmacologic inhibition of trypsin activity with benzamidine hydrochloride had no effect on PKCα-JNK-cJun pathway and necroptosis in CCK-stimulated pancreatic acinar cells. Furthermore, SR11302, the inhibitor of AP-1, significantly lowered tumor necrosis factor (TNF) α levels, and its subsequent receptor interacting protein kinases (RIP)3 and phosphorylated mixed lineagekinase domain-like (pMLKL) levels, ATP depletion and LDH release rate in CCK-stimulated pancreatic acinar cells. To sum up, all the results indicated that during AP, ER stress promoted pancreatic acinar cell necroptosis through CTSB maturation, thus induced AP-1 activation and TNFα secretion via PKCα-JNK-cJun pathway, not related with trypsin activity. These findings provided potential therapeutic target and treatment strategies for AP or other cell death-related diseases.
Collapse
Affiliation(s)
- Xiao Han
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Li
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingpiao Bao
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zengkai Wu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Congying Chen
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianbo Ni
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Shen
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pengli Song
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Peng
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rong Wan
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xingpeng Wang
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianghong Wu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Guoyong Hu, ; Jianghong Wu,
| | - Guoyong Hu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Guoyong Hu, ; Jianghong Wu,
| |
Collapse
|
21
|
Srivastava A, Tomar B, Sharma P, Kumari S, Prakash S, Rath SK, Kulkarni OP, Gupta SK, Mulay SR. RIPK3-MLKL signaling activates mitochondrial CaMKII and drives intrarenal extracellular matrix production during CKD. Matrix Biol 2022; 112:72-89. [PMID: 35964866 DOI: 10.1016/j.matbio.2022.08.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/19/2022] [Accepted: 08/09/2022] [Indexed: 11/19/2022]
Abstract
Intrarenal extracellular matrix production is a prevalent feature of all forms of chronic kidney disease (CKD). The transforming growth factor-beta (TGFβ) is believed to be a major driver of extracellular matrix production. Nevertheless, anti-TGFβ therapies have consistently failed to reduce extracellular matrix production in CKD patients indicating the need for novel therapeutic strategies. We have previously shown that necroinflammation contributes to acute kidney injury. Here, we show that chronic/persistent necroinflammation drives intrarenal extracellular matrix production during CKD. We found that renal expression of receptor-interacting protein kinase-1 (RIPK1), RIPK3, and mixed lineage kinase domain-like (MLKL) increases with the expansion of intrarenal extracellular matrix production and declined kidney function in both humans and mice. Furthermore, we found that TGFβ exposure induces the translocation of RIPK3 and MLKL to mitochondria resulting in mitochondrial dysfunction and ROS production. Mitochondrial ROS activates the serine-threonine kinase calcium/calmodulin-dependent protein kinases-II (CaMKII) that increases phosphorylation of Smad2/3 and subsequent production of alpha-smooth muscle actin (αSMA), collagen (Col) 1α1, etc. in response to TGFβ during the intrarenal extracellular matrix production. Consistent with this, deficiency or knockdown of RIPK3 or MLKL as well as pharmacological inhibition of RIPK1, RIPK3, and CaMKII prevents the intrarenal extracellular matrix production in oxalate-induced CKD and unilateral ureteral obstruction (UUO). Together, RIPK1, RIPK3, MLKL, CaMKII, and Smad2/3 are molecular targets to inhibit intrarenal extracellular matrix production and preserve kidney function during CKD.
Collapse
Affiliation(s)
- Anjali Srivastava
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Bhawna Tomar
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Pravesh Sharma
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad, 500078, India
| | - Sunaina Kumari
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Shakti Prakash
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Srikanta Kumar Rath
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Onkar Prakash Kulkarni
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad, 500078, India
| | - Shashi Kumar Gupta
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shrikant R Mulay
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
22
|
Gritsenko A, Díaz-Pino R, López-Castejón G. NLRP3 inflammasome triggers interleukin-37 release from human monocytes. Eur J Immunol 2022; 52:1141-1157. [PMID: 35429346 PMCID: PMC9540663 DOI: 10.1002/eji.202149724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 04/12/2022] [Accepted: 04/13/2022] [Indexed: 01/18/2023]
Abstract
IL-37 is an anti-inflammatory member of the IL-1 family that dampens inflammation associated with many noncommunicable diseases. However, mechanisms of IL-37 regulation remain understudied. We aimed to investigate the enzymatic cleavage of IL-37 that potentiates extracellular signalling, as well as pathways of IL-37 secretion. In human monocytes, mature IL-37 (mIL-37) was released following canonical NLRP3 inflammasome activation. The release of IL-37 was blocked by inhibiting plasma membrane permeability and in gasdermin-D-deficient THP-1 cells. While the cleavage of IL-37 was found to be constitutive, the release of mIL-37 was blocked in NLRP3-deficient THP-1 cells and by NLRP3 inhibitor MCC950 in THP-1s and primary human monocytes. IL-37 secretion also occurred after 18-h exposure to LPS, independently of the alternative NLRP3 inflammasome. This LPS-dependent IL-37 secretion required plasma membrane permeability, but not conventional protein secretion apparatus. Mutagenesis of the suggested caspase-1 cleavage site (D20) or the proposed alternative cleavage site (V46) did not completely block IL-37 processing. Therefore, we propose a novel pathway in which IL-37 is cleaved by caspase-1-independent mechanisms and released following canonical and alternative NLRP3 inflammasome triggers by differential pathways.
Collapse
Affiliation(s)
- Anna Gritsenko
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,School of Biological Sciences, Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Rodrigo Díaz-Pino
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,School of Biological Sciences, Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Gloria López-Castejón
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,School of Biological Sciences, Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
23
|
Jung SY, Park JI, Jeong JH, Song KH, Ahn J, Hwang SG, Kim J, Park JK, Lim DS, Song JY. Receptor interacting protein 1 knockdown induces cell death in liver cancer by suppressing STAT3/ATR activation in a p53-dependent manner. Am J Cancer Res 2022; 12:2594-2611. [PMID: 35812053 PMCID: PMC9251686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/02/2022] [Indexed: 06/15/2023] Open
Abstract
The survival and death of eukaryotic cells are tightly controlled by a variety of proteins in response to the cellular environment. Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) is a receptor-interacting Ser/Thr kinase that has recently been reported as an important regulator of cell survival, apoptosis, and necroptosis; however, its role in liver cancer remains unclear. In this study, we examined the effect of siRNA-mediated RIPK1 knockdown on the survival and death of liver cancer cells. Treatment with siRIPK1 decreased the growth rate of liver cancer cells and increased apoptotic, but not necrotic cell death, which was higher in wild-type p53 (wt-p53) cells than in mutant-type p53 (mt-p53) cells. In addition, RIPK1 knockdown increased p53 expression and G1 phase arrest in wt-p53 cells. Although suppressing p53 did not alter RIPK1 expression, it did attenuate siRIPK1-induced cell death. Interestingly, RIPK1 knockdown also increased the generation of reactive oxygen species and DNA damage by inhibiting signal transduced and activator of transcription 3 (STAT3) and ATM and RAD3-related (ATR) in wt-p53 cells but not in mt-p53 cells. Moreover, STAT3 or ATR inhibition in p53 mutant cells restored siRIPK1-mediated cell death. Together, the results of this study suggest that RIPK1 suppression induces apoptotic cell death by inhibiting the STAT3/ATR axis in a p53-dependent manner. Furthermore, these findings suggest that RIPK1, alone or in combination, may be a promising target for treating liver cancer.
Collapse
Affiliation(s)
- Seung-Youn Jung
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical SciencesSeoul 01812, Republic of Korea
| | - Jeong-In Park
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical SciencesSeoul 01812, Republic of Korea
| | - Jae-Hoon Jeong
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical SciencesSeoul 01812, Republic of Korea
| | - Kyung-Hee Song
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical SciencesSeoul 01812, Republic of Korea
| | - Jiyeon Ahn
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical SciencesSeoul 01812, Republic of Korea
| | - Sang-Gu Hwang
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical SciencesSeoul 01812, Republic of Korea
| | - Jaesung Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical SciencesSeoul 01812, Republic of Korea
| | - Jong-Kuk Park
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical SciencesSeoul 01812, Republic of Korea
| | - Dae-Seog Lim
- Department of Biotechnology, CHA UniversityGyeonggi-do 13488, Republic of Korea
| | - Jie-Young Song
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical SciencesSeoul 01812, Republic of Korea
| |
Collapse
|
24
|
Mark C, Callander NS, Chng K, Miyamoto S, Warrick J. Timelapse viability assay to detect division and death of primary multiple myeloma cells in response to drug treatments with single cell resolution. Integr Biol (Camb) 2022; 14:49-61. [PMID: 35653717 PMCID: PMC9175638 DOI: 10.1093/intbio/zyac006] [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: 11/16/2021] [Revised: 03/15/2022] [Accepted: 04/12/2022] [Indexed: 11/12/2022]
Abstract
Heterogeneity among cancer cells and in the tumor microenvironment (TME) is thought to be a significant contributor to the heterogeneity of clinical therapy response observed between patients and can evolve over time. A primary example of this is multiple myeloma (MM), a generally incurable cancer where such heterogeneity contributes to the persistent evolution of drug resistance. However, there is a paucity of functional assays for studying this heterogeneity in patient samples or for assessing the influence of the patient TME on therapy response. Indeed, the population-averaged data provided by traditional drug response assays and the large number of cells required for screening remain significant hurdles to advancement. To address these hurdles, we developed a suite of accessible technologies for quantifying functional drug response to a panel of therapies in ex vivo three-dimensional culture using small quantities of a patient's own cancer and TME components. This suite includes tools for label-free single-cell identification and quantification of both cell division and death events with a standard brightfield microscope, an open-source software package for objective image analysis and feasible data management of multi-day timelapse experiments, and a new approach to fluorescent detection of cell death that is compatible with long-term imaging of primary cells. These new tools and capabilities are used to enable sensitive, objective, functional characterization of primary MM cell therapy response in the presence of TME components, laying the foundation for future studies and efforts to enable predictive assessment drug efficacy for individual patients.
Collapse
Affiliation(s)
- Christina Mark
- Cancer Biology Graduate Program, University of Wisconsin, Madison, WI 53705, USA
| | - Natalie S Callander
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin, Madison, WI 53705, USA
| | - Kenny Chng
- McArdle Laboratory of Cancer Research, University of Wisconsin, Madison, WI 53705, USA
| | - Shigeki Miyamoto
- Cancer Biology Graduate Program, University of Wisconsin, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI 53705, USA
- McArdle Laboratory of Cancer Research, University of Wisconsin, Madison, WI 53705, USA
- Department of Oncology, University of Wisconsin, Madison, WI 53705, USA
| | - Jay Warrick
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI 53705, USA
- Salus Discovery, LLC, 110 E. Main St. Suite 815, Madison, WI 53703, USA
| |
Collapse
|
25
|
Zhou J, Xu J, Li P, Sun S, Kadier Y, Zhou S, Cheng A. Necroptosis and Viral Myocarditis: Tumor Necrosis Factor α as a Novel Biomarker for the Diagnosis of Viral Myocarditis. Front Cell Dev Biol 2022; 10:826904. [PMID: 35602592 PMCID: PMC9114881 DOI: 10.3389/fcell.2022.826904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Programmed cell death (PCD), including necroptosis, has emerged as a significant pathway in cardiovascular diseases. The infection of viral myocarditis (VMC) could cause cardiomyocytes degeneration, necrosis, and immune-inflammatory myocardial response. In this review, we summarized and evaluated the available evidence on the pathogenesis, molecule mechanism, diagnosis, and potential treatment strategies of viral myocarditis, with a special focus on the novel mechanism of necroptosis for cardiomyocytes death. Studies have shown that tumor necrosis factor-alpha (TNF-α) is an important cytokine involved in the activation of necroptosis; an elevated level of TNF-α is continually reported in patients suffering from VMC, implicating its involvement in the pathogenesis of VMC. It is of great interest to explore the clinical implication of TNF-α. We subsequently conducted a meta-analysis on the efficacy of serum TNF-α expression level and its diagnostic accuracy on acute viral myocarditis detection. Taken together, the review demonstrates a compelling role of necroptosis involved in the pathogenesis of VMC. Further, applying TNF-α as a serological marker for the diagnosis of VMC may be a useful strategy.
Collapse
Affiliation(s)
- Jin Zhou
- Tianjin Chest Hospital, Tianjin, China
| | - Jing Xu
- Tianjin Chest Hospital, Tianjin, China
| | - Peng Li
- Tianjin Chest Hospital, Tianjin, China
| | - Shan Sun
- Tianjin Chest Hospital, Tianjin, China
| | | | - Shiying Zhou
- Hotan District People’s Hospital, Tianjin, China
| | - Aijuan Cheng
- Tianjin Chest Hospital, Tianjin, China
- *Correspondence: Aijuan Cheng,
| |
Collapse
|
26
|
Chen YS, Chuang WC, Kung HN, Cheng CY, Huang DY, Sekar P, Lin WW. Pan-Caspase Inhibitor zVAD Induces Necroptotic and Autophagic Cell Death in TLR3/4-Stimulated Macrophages. Mol Cells 2022; 45:257-272. [PMID: 34949739 PMCID: PMC9001149 DOI: 10.14348/molcells.2021.0193] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/24/2021] [Accepted: 10/15/2021] [Indexed: 11/27/2022] Open
Abstract
In addition to inducing apoptosis, caspase inhibition contributes to necroptosis and/or autophagy depending on the cell type and cellular context. In macrophages, necroptosis can be induced by co-treatment with Toll-like receptor (TLR) ligands (lipopolysaccharide [LPS] for TLR4 and polyinosinic-polycytidylic acid [poly I:C] for TLR3) and a cell-permeable pan-caspase inhibitor zVAD. Here, we elucidated the signaling pathways and molecular mechanisms of cell death. We showed that LPS/zVAD- and poly I:C/zVAD-induced cell death in bone marrow-derived macrophages (BMDMs) was inhibited by receptor-interacting protein kinase 1 (RIP1) inhibitor necrostatin-1 and autophagy inhibitor 3-methyladenine. Electron microscopic images displayed autophagosome/autolysosomes, and immunoblotting data revealed increased LC3II expression. Although zVAD did not affect LPS- or poly I:C-induced activation of IKK, JNK, and p38, it enhanced IRF3 and STAT1 activation as well as type I interferon (IFN) expression. In addition, zVAD inhibited ERK and Akt phosphorylation induced by LPS and poly I:C. Of note, zVAD-induced enhancement of the IRF3/IFN/STAT1 axis was abolished by necrostatin-1, while zVAD-induced inhibition of ERK and Akt was not. Our data further support the involvement of autocrine IFNs action in reactive oxygen species (ROS)-dependent necroptosis, LPS/zVAD-elicited ROS production was inhibited by necrostatin-1, neutralizing antibody of IFN receptor (IFNR) and JAK inhibitor AZD1480. Accordingly, both cell death and ROS production induced by TLR ligands plus zVAD were abrogated in STAT1 knockout macrophages. We conclude that enhanced TRIF-RIP1-dependent autocrine action of IFNβ, rather than inhibition of ERK or Akt, is involved in TLRs/zVAD-induced autophagic and necroptotic cell death via the JAK/STAT1/ROS pathway.
Collapse
Affiliation(s)
- Yuan-Shen Chen
- Department of Neurosurgery, National Taiwan University Hospital Yunlin Branch, Douliu 64041, Taiwan
| | - Wei-Chu Chuang
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei 10617, Taiwan
| | - Hsiu-Ni Kung
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University, Taipei 10617, Taiwan
| | - Ching-Yuan Cheng
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei 10617, Taiwan
| | - Duen-Yi Huang
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei 10617, Taiwan
| | - Ponarulselvam Sekar
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei 11031, Taiwan
| | - Wan-Wan Lin
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei 10617, Taiwan
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei 11031, Taiwan
- Department of Pharmacology, National Defense Medical Center, Taipei 11490, Taiwan
| |
Collapse
|
27
|
Zhao W, Song Y, Wang QQ, Han S, Li XX, Cui Y, Gao H, Yuan R, Yang S. Cryptotanshinone Induces Necroptosis through Ca2+ Release and ROS Production in vitro and in vivo. Curr Mol Pharmacol 2022; 15:1009-1023. [PMID: 35086466 DOI: 10.2174/1874467215666220127112201] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 10/15/2021] [Accepted: 11/01/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Necroptosis is a type of programmed necrosis mediated by receptor-interacting protein kinases 1 and 3 (RIP1 and RIP3), which is morphologically characterized by enlarged organelles, ruptured plasma membrane, and subsequent loss of intracellular contents. Cryptotanshinone (CPT), a diterpene quinone compound extracted from the root of Salvia miltiorrhiza Bunge, has been reported to have significant anticancer activities. However, the detailed mechanism of CPT has not been clearly illustrated. OBJECTIVE The present study aimed to explore the cell death type and mechanisms of CPT-induced in non-small cell lung cancer (NSCLC) cells. METHODS The cytotoxicity of CPT on A549 cells was assessed by MTS assay. Ca2+ release and reactive oxygen species (ROS) generation were detected by flow cytometry. The changes in mitochondrial membrane potential (MMP) were observed through JC-1 staining. The expressions of p-RIP1, p-RIP3, p-MLKL, and MAPKs pathway proteins were analyzed by western blotting analysis. The efficacy of CPT in vivo was evaluated by the Lewis lung carcinoma (LLC) xenograft mice model. Blood samples were collected for hematology analysis. ELISA investigated the effects of CPT on tumor necrosis factor α (TNF-α). Hematoxylin and eosin staining (HE) was used to determine the tumor tissues. Proteins' expression of tumor tissues was quantified by western blotting. RESULTS CPT inhibited the cell viability of A549 cells in a time- and concentration-dependent manner, which was reversed by Necrostatin-1 (Nec-1). In addition, CPT treatment increased the expression of p-RIP1, p-RIP3, p-MLKL, the release of Ca2+, ROS generation, and the MAPKs pathway activated in A549 cells. Moreover, animal experiment results showed that intraperitoneal injection of CPT (15 mg/kg and 30 mg/kg) significantly inhibited tumor growth in C57BL/6 mice without affecting the bodyweight and injuring the organs. CONCLUSION Our findings suggested that CPT-induced necroptosis via RIP1/RIP3/MLKL signaling pathway both in vitro and in vivo, indicating that CPT may be a promising agent in the treatment of NSCLC.
Collapse
Affiliation(s)
- Wentong Zhao
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530000, China
- Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Yuanbo Song
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine
| | - Qin-Qin Wang
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning 530200, China
| | - Shan Han
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530000, China
- Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Xin-Xing Li
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530000, China
- Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Yushun Cui
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning 530200, China
| | - Hongwei Gao
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530000, China
- Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Renyikun Yuan
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530000, China
- Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Shilin Yang
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530000, China
- Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| |
Collapse
|
28
|
Sensing plasma membrane pore formation induces chemokine production in survivors of regulated necrosis. Dev Cell 2022; 57:228-245.e6. [PMID: 35016014 PMCID: PMC8792343 DOI: 10.1016/j.devcel.2021.12.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 10/04/2021] [Accepted: 12/14/2021] [Indexed: 01/26/2023]
Abstract
Although overwhelming plasma membrane integrity loss leads to cell lysis and necrosis, cells can tolerate a limited level of plasma membrane damage, undergo ESCRT-III-mediated repair, and survive. Here, we find that cells which undergo limited plasma membrane damage from the pore-forming actions of MLKL, GSDMD, perforin, or detergents experience local activation of PKCs through Ca2+ influx at the damage sites. S660-phosphorylated PKCs subsequently activate the TAK1/IKKs axis and RelA/Cux1 complex to trigger chemokine expressions. We observe that in late-stage cancers, cells with active MLKL show expression of CXCL8. Similar expression induction is also found in ischemia-injured kidneys. Chemokines generated in this manner are also indispensable for recruiting immune cells to the dead and dying cells. This plasma membrane integrity-sensing pathway is similar to the well-established yeast cell wall integrity signaling pathway at molecular level, and this suggests an evolutionary conserved mechanism to respond to the cellular barrier damage.
Collapse
|
29
|
Needs SH, Bootman MD, Grotzke JE, Kramer HB, Allman SA. Off‐target inhibition of NGLY1 by the polycaspase inhibitor Z‐VAD‐fmk induces cellular autophagy. FEBS J 2022; 289:3115-3131. [PMID: 34995415 PMCID: PMC9304259 DOI: 10.1111/febs.16345] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 09/14/2021] [Accepted: 01/05/2022] [Indexed: 12/28/2022]
Affiliation(s)
- Sarah H. Needs
- School of Life, Health and Chemical Sciences The Open University Milton Keynes UK
- Reading School of Pharmacy University of Reading UK
| | - Martin D. Bootman
- School of Life, Health and Chemical Sciences The Open University Milton Keynes UK
| | | | - Holger B. Kramer
- Department of Physiology, Anatomy and Genetics University of Oxford UK
- MRC London Institute of Medical Sciences UK
| | - Sarah A. Allman
- School of Life, Health and Chemical Sciences The Open University Milton Keynes UK
- Reading School of Pharmacy University of Reading UK
- Leicester School of Pharmacy De Montfort University Leicester UK
| |
Collapse
|
30
|
Hu X, Xu Y, Zhang H, Li Y, Wang X, Xu C, Ni W, Zhou K. Role of necroptosis in traumatic brain and spinal cord injuries. J Adv Res 2021; 40:125-134. [PMID: 36100321 PMCID: PMC9481937 DOI: 10.1016/j.jare.2021.12.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 09/04/2021] [Accepted: 12/08/2021] [Indexed: 11/30/2022] Open
Affiliation(s)
- Xinli Hu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China; The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
| | - Yu Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China; The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
| | - Haojie Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China; The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
| | - Yao Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China; The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
| | - Xiangyang Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China; The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China.
| | - Cong Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China; The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China.
| | - Wenfei Ni
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China; The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China.
| | - Kailiang Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China; The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China.
| |
Collapse
|
31
|
Baty RS. Protective effect of Bosutinib with caspase inhibitors on human K562 cells. Saudi J Biol Sci 2021; 29:2323-2328. [PMID: 35531147 PMCID: PMC9072916 DOI: 10.1016/j.sjbs.2021.11.068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 12/05/2022] Open
Abstract
Introduction Cancer therapy has become increasingly focused on molecularly targeted medications. Despite the fact that multi-cytotoxic medication regimens have proven to be highly effective, many investigations in targeted treatments have focused on a single agent. The precise molecular mechanism of action of second-generation BCR–ABL tyrosine kinase inhibitors, which includes different targets and pathways, can help rationalize therapy in chronic myelogenous leukemia (CML) and other diseases affected by BCR–ABL tyrosine kinase inhibitors (TKIs). Aim The purpose of this study was to analyze if bosutinib (BOS) combined with Boc-D-FMK effectively suppressed proliferation and induced apoptosis in K562 cells to a lesser extent, implying that bosutinib is an effective leukemia treatment and that its combination with Boc-D-FMK is a mild chemotherapeutic agent against leukemia. Methods In this study, bosutinib was obtained together with other materials to perform a cell culture experiment with human cell lines, as well as additional drug treatment. Furthermore, cell viability (MTT assay) and flow cryometry such as viability and cell cycle assays are performed. The target profile of the dual SRC/ABL inhibitor bosutinib was studied in this study as a first kinase inhibitor to target K562 cells, which has recently been linked to the proliferation of myelogenous leukaemia cells, these results suggest the effectiveness of inhibitory activity on cell viability/proliferation, alone generated a potent value of 250 nM (39.27 ± 1.17) for 48 h as optimal dose. Results The cytotoxic effect of bosutinib on the K562 cell line was assessed in vitro using the MTT assay, and the cytotoxicity was further clarified using cell viability and cell cycle assays. Guava Cell Assay software validated the activation of apoptosis. Sub-G1, G0/G1, S, and G2/M phases are depicted. Cell cycle research revealed that K562 cells treated with bosutinib accumulated much more in the sub-G1 phase, which was later validated by a drop peak at the G2/M phase. Conclusion In conclusion, the nature of bosutinib's reduction of cancer cell growth may open the door to future research into the development of green synthesis medicines, particularly for cancer treatment.
Collapse
|
32
|
Verduijn J, Van der Meeren L, Krysko DV, Skirtach AG. Deep learning with digital holographic microscopy discriminates apoptosis and necroptosis. Cell Death Dis 2021; 7:229. [PMID: 34475384 PMCID: PMC8413278 DOI: 10.1038/s41420-021-00616-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/13/2021] [Accepted: 08/19/2021] [Indexed: 02/07/2023]
Abstract
Regulated cell death modalities such as apoptosis and necroptosis play an important role in regulating different cellular processes. Currently, regulated cell death is identified using the golden standard techniques such as fluorescence microscopy and flow cytometry. However, they require fluorescent labels, which are potentially phototoxic. Therefore, there is a need for the development of new label-free methods. In this work, we apply Digital Holographic Microscopy (DHM) coupled with a deep learning algorithm to distinguish between alive, apoptotic and necroptotic cells in murine cancer cells. This method is solely based on label-free quantitative phase images, where the phase delay of light by cells is quantified and is used to calculate their topography. We show that a combination of label-free DHM in a high-throughput set-up (~10,000 cells per condition) can discriminate between apoptosis, necroptosis and alive cells in the L929sAhFas cell line with a precision of over 85%. To the best of our knowledge, this is the first time deep learning in the form of convolutional neural networks is applied to distinguish-with a high accuracy-apoptosis and necroptosis and alive cancer cells from each other in a label-free manner. It is expected that the approach described here will have a profound impact on research in regulated cell death, biomedicine and the field of (cancer) cell biology in general.
Collapse
Affiliation(s)
- Joost Verduijn
- grid.5342.00000 0001 2069 7798Nano-Biotechnology Laboratory, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium ,grid.510942.bCancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Louis Van der Meeren
- grid.5342.00000 0001 2069 7798Nano-Biotechnology Laboratory, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium ,grid.510942.bCancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Dmitri V. Krysko
- grid.510942.bCancer Research Institute Ghent, 9000 Ghent, Belgium ,grid.5342.00000 0001 2069 7798Cell Death Investigation and Therapy (CDIT) Laboratory, Anatomy an Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium ,grid.448878.f0000 0001 2288 8774Department of Pathophysiology, Sechenov First Moscow State Medical University (Sechenov University), 119146 Moscow, Russian Federation
| | - André G. Skirtach
- grid.5342.00000 0001 2069 7798Nano-Biotechnology Laboratory, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium ,grid.510942.bCancer Research Institute Ghent, 9000 Ghent, Belgium
| |
Collapse
|
33
|
van Schaik TA, Chen KS, Shah K. Therapy-Induced Tumor Cell Death: Friend or Foe of Immunotherapy? Front Oncol 2021; 11:678562. [PMID: 34141622 PMCID: PMC8204251 DOI: 10.3389/fonc.2021.678562] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 05/03/2021] [Indexed: 12/13/2022] Open
Abstract
Combinatory treatments using surgery, radiotherapy and/or chemotherapy together with immunotherapy have shown encouraging results for specific subsets of tumors, but a significant proportion of tumors remains unsusceptible. Some of these inconsistencies are thought to be the consequence of an immunosuppressive tumor microenvironment (TME) caused by therapy-induced tumor cell death (TCD). An increased understanding of the molecular mechanisms governing TCD has provided valuable insights in specific signaling cascades activated by treatment and the subsequent effects on the TME. Depending on the treatment variables of conventional chemo-, radio- and immunotherapy and the genetic composition of the tumor cells, particular cell death pathways are activated. Consequently, TCD can either have tolerogenic or immunogenic effects on the local environment and thereby affect the post-treatment anti-tumor response of immune cells. Thus, identification of these events can provide new rationales to increase the efficacy of conventional therapies combined with immunotherapies. In this review, we sought to provide an overview of the molecular mechanisms initiated by conventional therapies and the impact of treatment-induced TCD on the TME. We also provide some perspectives on how we can circumvent tolerogenic effects by adequate treatment selection and manipulation of key signaling cascades.
Collapse
Affiliation(s)
- Thijs A van Schaik
- Center for Stem Cell Therapeutics and Imaging (CSTI), Harvard Medical School, Boston, MA, United States.,Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Kok-Siong Chen
- Center for Stem Cell Therapeutics and Imaging (CSTI), Harvard Medical School, Boston, MA, United States.,Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Khalid Shah
- Center for Stem Cell Therapeutics and Imaging (CSTI), Harvard Medical School, Boston, MA, United States.,Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Harvard Stem Cell Institute, Harvard University, Cambridge, MA, United States
| |
Collapse
|
34
|
Oliveira SR, Dionísio PA, Gaspar MM, Ferreira MBT, Rodrigues CAB, Pereira RG, Estevão MS, Perry MJ, Moreira R, Afonso CAM, Amaral JD, Rodrigues CMP. Discovery of a Necroptosis Inhibitor Improving Dopaminergic Neuronal Loss after MPTP Exposure in Mice. Int J Mol Sci 2021; 22:5289. [PMID: 34069782 PMCID: PMC8157267 DOI: 10.3390/ijms22105289] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/10/2021] [Accepted: 05/14/2021] [Indexed: 11/29/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder, mainly characterized by motor deficits correlated with progressive dopaminergic neuronal loss in the substantia nigra pars compacta (SN). Necroptosis is a caspase-independent form of regulated cell death mediated by the concerted action of receptor-interacting protein 3 (RIP3) and the pseudokinase mixed lineage domain-like protein (MLKL). It is also usually dependent on RIP1 kinase activity, influenced by further cellular clues. Importantly, necroptosis appears to be strongly linked to several neurodegenerative diseases, including PD. Here, we aimed at identifying novel chemical inhibitors of necroptosis in a PD-mimicking model, by conducting a two-step screening. Firstly, we phenotypically screened a library of 31 small molecules using a cellular model of necroptosis and, thereafter, the hit compound effect was validated in vivo in a sub-acute 1-methyl-1-4-phenyl-1,2,3,6-tetrahydropyridine hydrochloride (MPTP) PD-related mouse model. From the initial compounds, we identified one hit-Oxa12-that strongly inhibited necroptosis induced by the pan-caspase inhibitor zVAD-fmk in the BV2 murine microglia cell line. More importantly, mice exposed to MPTP and further treated with Oxa12 showed protection against MPTP-induced dopaminergic neuronal loss in the SN and striatum. In conclusion, we identified Oxa12 as a hit compound that represents a new chemotype to tackle necroptosis. Oxa12 displays in vivo effects, making this compound a drug candidate for further optimization to attenuate PD pathogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Cecília M. P. Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (S.R.O.); (P.A.D.); (M.M.G.); (M.B.T.F.); (C.A.B.R.); (R.G.P.); (M.S.E.); (M.J.P.); (R.M.); (C.A.M.A.); (J.D.A.)
| |
Collapse
|
35
|
Shao CS, Feng N, Zhou S, Zheng XX, Wang P, Zhang JS, Huang Q. Ganoderic acid T improves the radiosensitivity of HeLa cells via converting apoptosis to necroptosis. Toxicol Res (Camb) 2021; 10:531-541. [PMID: 34141167 DOI: 10.1093/toxres/tfab030] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/01/2021] [Accepted: 03/05/2021] [Indexed: 11/13/2022] Open
Abstract
The use of natural substances derived from traditional Chinese medicine and natural plants as safe radiosensitizing adjuvants is a new trend for cancer radiotherapy. Ganoderma lucidum has been used as a traditional Chinese medicine with a history of more than 2000 years. Ganoderic acid T (GAT) is a typical triterpene of G. lucidum, which has strong cytotoxicity to cancer cells, but whether it has radiation sensitization effect has not been explored. In this work, we treated the HeLa cells with different concentrations of GAT before exposure to gamma-ray radiation and investigated its influence on the radiosensitivity. The cell viability, apoptosis rate, necoptosis rate, intracellular ATP level, cell cycle, the amount of H2AX and 53BP1, reactive oxygen species, and mitochondrial membrane potential were examined. Apoptotic, necroptotic, and autophagic biomarker proteins, including caspase 8, cytochrome c, caspase 3, RIPK, MLKL, P62, and LC3, were analyzed. As a result, we confirmed that with treatment of GAT, the gamma-ray radiation induced both apoptosis and necroptosis in HeLa cells, and with increase of GAT, the percentage ratio of necroptosis was increased. The involved pathways and mechanisms were also explored and discussed.
Collapse
Affiliation(s)
- Chang-Sheng Shao
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei 230031, China
| | - Na Feng
- Key Laboratory of Edible Fungi Resources and Utilization (South), Ministry of Agriculture, P. R., China
| | - Shuai Zhou
- Key Laboratory of Edible Fungi Resources and Utilization (South), Ministry of Agriculture, P. R., China
| | - Xin-Xin Zheng
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei 230031, China
| | - Peng Wang
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei 230031, China
| | - Jing-Song Zhang
- Key Laboratory of Edible Fungi Resources and Utilization (South), Ministry of Agriculture, P. R., China
| | - Qing Huang
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei 230031, China
| |
Collapse
|
36
|
Khan I, Yousif A, Chesnokov M, Hong L, Chefetz II. A decade of cell death studies: Breathing new life into necroptosis. Pharmacol Ther 2021; 220:107717. [DOI: 10.1016/j.pharmthera.2020.107717] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/19/2020] [Indexed: 12/31/2022]
|
37
|
Adjemian S, Oltean T, Martens S, Wiernicki B, Goossens V, Vanden Berghe T, Cappe B, Ladik M, Riquet FB, Heyndrickx L, Bridelance J, Vuylsteke M, Vandecasteele K, Vandenabeele P. Ionizing radiation results in a mixture of cellular outcomes including mitotic catastrophe, senescence, methuosis, and iron-dependent cell death. Cell Death Dis 2020; 11:1003. [PMID: 33230108 PMCID: PMC7684309 DOI: 10.1038/s41419-020-03209-y] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 12/21/2022]
Abstract
Radiotherapy is commonly used as a cytotoxic treatment of a wide variety of tumors. Interestingly, few case reports underlined its potential to induce immune-mediated abscopal effects, resulting in regression of metastases, distant from the irradiated site. These observations are rare, and apparently depend on the dose used, suggesting that dose-related cellular responses may be involved in the distant immunogenic responses. Ionizing radiation (IR) has been reported to elicit immunogenic apoptosis, necroptosis, mitotic catastrophe, and senescence. In order to link a cellular outcome with a particular dose of irradiation, we performed a systematic study in a panel of cell lines on the cellular responses at different doses of X-rays. Remarkably, we observed that all cell lines tested responded in a similar fashion to IR with characteristics of mitotic catastrophe, senescence, lipid peroxidation, and caspase activity. Iron chelators (but not Ferrostatin-1 or vitamin E) could prevent the formation of lipid peroxides and cell death induced by IR, suggesting a crucial role of iron-dependent cell death during high-dose irradiation. We also show that in K-Ras-mutated cells, IR can induce morphological features reminiscent of methuosis, a cell death modality that has been recently described following H-Ras or K-Ras mutation overexpression.
Collapse
Affiliation(s)
- Sandy Adjemian
- Unit of Molecular Signaling and Cell Death, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Teodora Oltean
- Unit of Molecular Signaling and Cell Death, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Sofie Martens
- Unit of Molecular Signaling and Cell Death, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Bartosz Wiernicki
- Unit of Molecular Signaling and Cell Death, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Vera Goossens
- VIB Screening Core & UGhent Expertise Centre for Bioassay Development and Screening (C-BIOS), VIB, UGhent, Ghent, Belgium
| | - Tom Vanden Berghe
- Unit of Molecular Signaling and Cell Death, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium.,Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Benjamin Cappe
- Unit of Molecular Signaling and Cell Death, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Maria Ladik
- Unit of Molecular Signaling and Cell Death, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Franck B Riquet
- Unit of Molecular Signaling and Cell Death, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium.,Université de Lille, Lille, France
| | - Liesbeth Heyndrickx
- Unit of Molecular Signaling and Cell Death, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Jolien Bridelance
- Unit of Molecular Signaling and Cell Death, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | | | - Katrien Vandecasteele
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.,Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Ghent, Belgium.,Radiation Oncology, Ghent University Hospital, Ghent, Belgium
| | - Peter Vandenabeele
- Unit of Molecular Signaling and Cell Death, VIB Center for Inflammation Research, Ghent, Belgium. .,Department of Biomedical Molecular Biology, Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium. .,Cancer Research Institute Ghent (CRIG), Ghent, Belgium. .,Methusalem program, Ghent University, Ghent, Belgium.
| |
Collapse
|
38
|
Bedient L, Pokharel SM, Chiok KR, Mohanty I, Beach SS, Miura TA, Bose S. Lytic Cell Death Mechanisms in Human Respiratory Syncytial Virus-Infected Macrophages: Roles of Pyroptosis and Necroptosis. Viruses 2020; 12:v12090932. [PMID: 32854254 PMCID: PMC7552060 DOI: 10.3390/v12090932] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/13/2020] [Accepted: 08/20/2020] [Indexed: 12/21/2022] Open
Abstract
Human respiratory syncytial virus (RSV) is the most common cause of viral bronchiolitis and pneumonia in infants and children worldwide. Inflammation induced by RSV infection is responsible for its hallmark manifestation of bronchiolitis and pneumonia. The cellular debris created through lytic cell death of infected cells is a potent initiator of this inflammation. Macrophages are known to play a pivotal role in the early innate immune and inflammatory response to viral pathogens. However, the lytic cell death mechanisms associated with RSV infection in macrophages remains unknown. Two distinct mechanisms involved in lytic cell death are pyroptosis and necroptosis. Our studies revealed that RSV induces lytic cell death in macrophages via both of these mechanisms, specifically through the ASC (Apoptosis-associated speck like protein containing a caspase recruitment domain)-NLRP3 (nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3) inflammasome activation of both caspase-1 dependent pyroptosis and receptor-interacting serine/threonine-protein kinase 3 (RIPK3), as well as a mixed lineage kinase domain like pseudokinase (MLKL)-dependent necroptosis. In addition, we demonstrated an important role of reactive oxygen species (ROS) during lytic cell death of RSV-infected macrophages.
Collapse
Affiliation(s)
- Lori Bedient
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA; (L.B.); (S.M.P.); (K.R.C.); (I.M.)
| | - Swechha Mainali Pokharel
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA; (L.B.); (S.M.P.); (K.R.C.); (I.M.)
| | - Kim R. Chiok
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA; (L.B.); (S.M.P.); (K.R.C.); (I.M.)
| | - Indira Mohanty
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA; (L.B.); (S.M.P.); (K.R.C.); (I.M.)
| | - Sierra S. Beach
- Department of Biological Sciences, University of Idaho, Moscow, ID 83844, USA; (S.S.B.); (T.A.M.)
| | - Tanya A. Miura
- Department of Biological Sciences, University of Idaho, Moscow, ID 83844, USA; (S.S.B.); (T.A.M.)
| | - Santanu Bose
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA; (L.B.); (S.M.P.); (K.R.C.); (I.M.)
- Correspondence:
| |
Collapse
|
39
|
Hsu SK, Chang WT, Lin IL, Chen YF, Padalwar NB, Cheng KC, Teng YN, Wang CH, Chiu CC. The Role of Necroptosis in ROS-Mediated Cancer Therapies and Its Promising Applications. Cancers (Basel) 2020; 12:E2185. [PMID: 32764483 PMCID: PMC7465132 DOI: 10.3390/cancers12082185] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 07/30/2020] [Indexed: 02/07/2023] Open
Abstract
Over the past decades, promising therapies targeting different signaling pathways have emerged. Among these pathways, apoptosis has been well investigated and targeted to design diverse chemotherapies. However, some patients are chemoresistant to these therapies due to compromised apoptotic cell death. Hence, exploring alternative treatments aimed at different mechanisms of cell death seems to be a potential strategy for bypassing impaired apoptotic cell death. Emerging evidence has shown that necroptosis, a caspase-independent form of cell death with features between apoptosis and necrosis, can overcome the predicament of drug resistance. Furthermore, previous studies have also indicated that there is a close correlation between necroptosis and reactive oxygen species (ROS); both necroptosis and ROS play significant roles both under human physiological conditions such as the regulation of inflammation and in cancer biology. Several small molecules used in experiments and clinical practice eliminate cancer cells via the modulation of ROS and necroptosis. The molecular mechanisms of these promising therapies are discussed in detail in this review.
Collapse
Affiliation(s)
- Sheng-Kai Hsu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Wen-Tsan Chang
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - I-Ling Lin
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Laboratory Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Yih-Fung Chen
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Nitin Balkrushna Padalwar
- Department of Chemistry, National Institute of Technology Tiruchirappalli, Tiruchirappalli 620015, Tamilnadu, India;
| | - Kai-Chun Cheng
- Department of Ophthalmology, Kaohsiung Municipal Hsiaokang Hospital, Kaohsiung 812, Taiwan;
- Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Yen-Ni Teng
- Department of Biological Sciences and Technology, National University of Tainan, Tainan 700, Taiwan;
| | - Chi-Huei Wang
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- The Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
40
|
Fang Y, He Y, Zhai B, Hou C, Xu R, Xing C, Wang X, Ma N, Han G, Wang R. The E3 ubiquitin ligase Itch deficiency promotes antigen-driven B-cell responses in mice. Eur J Immunol 2020; 51:103-114. [PMID: 32652569 DOI: 10.1002/eji.202048640] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/20/2020] [Indexed: 11/10/2022]
Abstract
Deficiency of Itch, an E3 ubiquitin ligase, usually induced severe systemic and progressive autoimmune disease. The Itch function is well studied in T cells but not in B cells. We hypothesize that B-cell-specific Itch deficiency promoted antigen-induced B-cell activation and antibody-expressing plasma cell (PC) production. We found that unlike Itch KO, Itch cKO (CD19cre Itchf/f ) mice did not demonstrated a significant increase in the sizes of spleens and LNs, antibody level, and base mutation of antibody gene. However, in line with the fact that Itch expression decreased in GC B cells, PCs, and plasmablast (PB)-like SP 2/0 cells, Itch deficiency promoted B-cell activation and antibody production induced by antigens including lipopolysaccharide (LPS) and sheep red blood cells (SRBCs). Mechanistically, we found that Itch deficiency promotes antigen-induced cytokine production because Itch controls the proteins (e.g., eIF3a, eIF3c, eIF3h) with translation initiation factor activity. Altogether, our data suggest that Itch deficiency promotes antigen-driven B-cell response. This may provide hints for Itch-targeted treatment of patients with autoimmune disease.
Collapse
Affiliation(s)
- Ying Fang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China.,Department of Rheumatology, First Hospital of Jilin University, Changchun, China
| | - Youdi He
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China.,Department of Neurology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Bing Zhai
- Institute of Military Cognition and Brain Sciences, Beijing, China.,Department of Geriatric Hematology, Chinese PLA General Hospital, Beijing, China
| | - Chunmei Hou
- Institute of Military Cognition and Brain Sciences, Beijing, China
| | - Ruonan Xu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Chen Xing
- Institute of Military Cognition and Brain Sciences, Beijing, China
| | - Xiaoqian Wang
- Staidson (Beijing) Biopharmaceuticals Co., Ltd, Beijing, China
| | - Ning Ma
- Department of Rheumatology, First Hospital of Jilin University, Changchun, China
| | - Gencheng Han
- Institute of Military Cognition and Brain Sciences, Beijing, China
| | - Renxi Wang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
41
|
Huang W, Xie W, Gong J, Wang W, Cai S, Huang Q, Chen Z, Liu Y. Heat stress induces RIP1/RIP3-dependent necroptosis through the MAPK, NF-κB, and c-Jun signaling pathways in pulmonary vascular endothelial cells. Biochem Biophys Res Commun 2020; 528:206-212. [PMID: 32471717 DOI: 10.1016/j.bbrc.2020.04.150] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 04/29/2020] [Indexed: 01/30/2023]
Abstract
Necroptosis represents a newly defined form of regulated necrosis and participates in various human inflammatory diseases. It remains unclear whether necroptosis is presented in heatstroke-induced lung injury. We show that heat stress(HS) triggered an significant upregulation of receptor-interacting protein 1 (RIP1) and mixed lineage kinase domain-like protein (MLKL) expression in a time-dependent manner, without a significant change of receptor-interacting protein 3 (RIP3). Furthermore, co-immunoprecipitation assays showed that RIP1 binds to RIP3 to form the necrosome in heat stress-induced PMVECs. In vitro, necrostatin-1 (Nec-1) pre-treatment reduced heat stress-induced PMVECs necroptosis, which also inhibited HMGB1 translocation from the nucleus into the cytoplasm. Similarly, inhibition for ERK (PD98059), NF-κB (BAY11-7082) and c-Jun (c-Jun peptide), respectively, also suppressed the HMGB1 cytoplasm translocation. Furthermore, siRNA-mediated RIP1/RIP3 knockdown negatively regulated the release of HMGB1 in HS-induced necroptosis through the ERK, NF-κB, and c-Jun signaling pathways. Our study reveals that HS induces RIP1/RIP3-dependent necroptosis through the MAPK, NF-κB, and c-Jun signaling pathways in PMVECs.
Collapse
Affiliation(s)
- Wei Huang
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Weidang Xie
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jian Gong
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China; Department of Intensive Care Medicine, The Third People's Hospital of Longgang District, Shenzhen, 518115, China
| | - Wenyan Wang
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Sumin Cai
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qiaobing Huang
- Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, Southern Medical University, Guangzhou, 510515, China
| | - Zhongqing Chen
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yanan Liu
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
42
|
Fang Y, Gao S, Wang X, Cao Y, Lu J, Chen S, Lenahan C, Zhang JH, Shao A, Zhang J. Programmed Cell Deaths and Potential Crosstalk With Blood-Brain Barrier Dysfunction After Hemorrhagic Stroke. Front Cell Neurosci 2020; 14:68. [PMID: 32317935 PMCID: PMC7146617 DOI: 10.3389/fncel.2020.00068] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/06/2020] [Indexed: 12/13/2022] Open
Abstract
Hemorrhagic stroke is a life-threatening neurological disease characterized by high mortality and morbidity. Various pathophysiological responses are initiated after blood enters the interstitial space of the brain, compressing the brain tissue and thus causing cell death. Recently, three new programmed cell deaths (PCDs), necroptosis, pyroptosis, and ferroptosis, were also found to be important contributors in the pathophysiology of hemorrhagic stroke. Additionally, blood-brain barrier (BBB) dysfunction plays a crucial role in the pathophysiology of hemorrhagic stroke. The primary insult following BBB dysfunction may disrupt the tight junctions (TJs), transporters, transcytosis, and leukocyte adhesion molecule expression, which may lead to brain edema, ionic homeostasis disruption, altered signaling, and immune infiltration, consequently causing neuronal cell death. This review article summarizes recent advances in our knowledge of the mechanisms regarding these new PCDs and reviews their contributions in hemorrhagic stroke and potential crosstalk in BBB dysfunction. Numerous studies revealed that necroptosis, pyroptosis, and ferroptosis participate in cell death after subarachnoid hemorrhage (SAH) and intracerebral hemorrhage (ICH). Endothelial dysfunction caused by these three PCDs may be the critical factor during BBB damage. Also, several signaling pathways were involved in PCDs and BBB dysfunction. These new PCDs (necroptosis, pyroptosis, ferroptosis), as well as BBB dysfunction, each play a critical role after hemorrhagic stroke. A better understanding of the interrelationship among them might provide us with better therapeutic targets for the treatment of hemorrhagic stroke.
Collapse
Affiliation(s)
- Yuanjian Fang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shiqi Gao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoyu Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yang Cao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianan Lu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Sheng Chen
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Cameron Lenahan
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, United States.,Burrell College of Osteopathic Medicine, Las Cruces, NM, United States.,Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, United States.,Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA, United States.,Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda, CA, United States.,Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Brain Research Institute, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, China
| |
Collapse
|
43
|
A 3D Cell Death Assay to Quantitatively Determine Ferroptosis in Spheroids. Cells 2020; 9:cells9030703. [PMID: 32183000 PMCID: PMC7140689 DOI: 10.3390/cells9030703] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/06/2020] [Accepted: 03/10/2020] [Indexed: 12/13/2022] Open
Abstract
The failure of drug efficacy in clinical trials remains a big issue in cancer research. This is largely due to the limitations of two-dimensional (2D) cell cultures, the most used tool in drug screening. Nowadays, three-dimensional (3D) cultures, including spheroids, are acknowledged to be a better model of the in vivo environment, but detailed cell death assays for 3D cultures (including those for ferroptosis) are scarce. In this work, we show that a new cell death analysis method, named 3D Cell Death Assay (3DELTA), can efficiently determine different cell death types including ferroptosis and quantitatively assess cell death in tumour spheroids. Our method uses Sytox dyes as a cell death marker and Triton X-100, which efficiently permeabilizes all cells in spheroids, was used to establish 100% cell death. After optimization of Sytox concentration, Triton X-100 concentration and timing, we showed that the 3DELTA method was able to detect signals from all cells without the need to disaggregate spheroids. Moreover, in this work we demonstrated that 2D experiments cannot be extrapolated to 3D cultures as 3D cultures are less sensitive to cell death induction. In conclusion, 3DELTA is a more cost-effective way to identify and measure cell death type in 3D cultures, including spheroids.
Collapse
|
44
|
Lou X, Zhu H, Ning L, Li C, Li S, Du H, Zhou X, Xu G. EZH2 Regulates Intestinal Inflammation and Necroptosis Through the JNK Signaling Pathway in Intestinal Epithelial Cells. Dig Dis Sci 2019; 64:3518-3527. [PMID: 31273598 DOI: 10.1007/s10620-019-05705-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 06/10/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is a common disorder of chronic intestinal inflammation that can be caused by the disruption of intestinal immune homeostasis. AIM We aimed to evaluate the role of enhancer of zeste homolog 2 (EZH2) in the inflammatory response and explore the association between EZH2 and necroptosis in human epithelial colorectal adenocarcinoma cell lines. METHODS In both in vitro and in vivo models, expression of EZH2 in intestinal tissues was verified by histology. The expression of inflammatory cytokines in cell lines treated with EZH2 siRNA with or without stimulus was analyzed by quantitative real-time polymerase chain reaction. An intestinal necroptosis cell model was established to elucidate whether EZH2 is involved in necroptosis. RESULTS Our present data indicated that EZH2 expression was decreased in in vitro and in vivo models and in patients with inflammatory bowel disease. EZH2 downregulation increased the expression of inflammatory factors, including TNF-α, IL-8, IL-17, CCL5, and CCL20 in a Caco-2 cell model. The JNK pathway was activated with the reduction of EZH2. In the necroptosis model, downregulation of EZH2 was detected with the upregulation of necroptotic markers RIP1 and RIP3. In addition, EZH2 knockdown with siRNA increased p-JNK and p-c-Jun. CONCLUSION Our data suggest that EZH2 plays an important role in the development of intestinal inflammation and necroptosis. Hence, EZH2 could be a potential therapeutic target for IBD.
Collapse
Affiliation(s)
- Xinhe Lou
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Huatuo Zhu
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Longgui Ning
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Chunxiao Li
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Sha Li
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Haojie Du
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Xinxin Zhou
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Guoqiang Xu
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China.
| |
Collapse
|
45
|
Membrane-bound TNF mediates microtubule-targeting chemotherapeutics-induced cancer cytolysis via juxtacrine inter-cancer-cell death signaling. Cell Death Differ 2019; 27:1569-1587. [PMID: 31645676 PMCID: PMC7206059 DOI: 10.1038/s41418-019-0441-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 10/09/2019] [Accepted: 10/09/2019] [Indexed: 12/17/2022] Open
Abstract
Microtubule-targeting agents (MTAs) are a class of most widely used chemotherapeutics and their mechanism of action has long been assumed to be mitotic arrest of rapidly dividing tumor cells. In contrast to such notion, here we show—in many cancer cell types—MTAs function by triggering membrane TNF (memTNF)-mediated cancer-cell-to-cancer-cell killing, which differs greatly from other non-MTA cell-cycle-arresting agents. The killing is through programmed cell death (PCD), either in way of necroptosis when RIP3 kinase is expressed, or of apoptosis in its absence. Mechanistically, MTAs induce memTNF transcription via the JNK-cJun signaling pathway. With respect to chemotherapy regimens, our results establish that memTNF-mediated killing is significantly augmented by IAP antagonists (Smac mimetics) in a broad spectrum of cancer types, and with their effects most prominently manifested in patient-derived xenograft (PDX) models in which cell–cell contacts are highly reminiscent of human tumors. Therefore, our finding indicates that memTNF can serve as a marker for patient responsiveness, and Smac mimetics will be effective adjuvants for MTA chemotherapeutics. The present study reframes our fundamental biochemical understanding of how MTAs take advantage of the natural tight contact of tumor cells and utilize memTNF-mediated death signaling to induce the entire tumor regression.
Collapse
|
46
|
Simader E, Beer L, Laggner M, Vorstandlechner V, Gugerell A, Erb M, Kalinina P, Copic D, Moser D, Spittler A, Tschachler E, Jan Ankersmit H, Mildner M. Tissue-regenerative potential of the secretome of γ-irradiated peripheral blood mononuclear cells is mediated via TNFRSF1B-induced necroptosis. Cell Death Dis 2019; 10:729. [PMID: 31570701 PMCID: PMC6768878 DOI: 10.1038/s41419-019-1974-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 09/05/2019] [Accepted: 09/09/2019] [Indexed: 12/14/2022]
Abstract
Peripheral blood mononuclear cells (PBMCs) have been shown to produce and release a plethora of pro-angiogenetic factors in response to γ-irradiation, partially accounting for their tissue-regenerative capacity. Here, we investigated whether a certain cell subtype of PBMCs is responsible for this effect, and whether the type of cell death affects the pro-angiogenic potential of bioactive molecules released by γ-irradiated PBMCs. PBMCs and PBMC subpopulations, including CD4+ and CD8+ T cells, B cells, monocytes, and natural killer cells, were isolated and subjected to high-dose γ-irradiation. Transcriptome analysis revealed subpopulation-specific responses to γ-irradiation with distinct activation of pro-angiogenic pathways, cytokine production, and death receptor signalling. Analysis of the proteins released showed that interactions of the subsets are important for the generation of a pro-angiogenic secretome. This result was confirmed at the functional level by the finding that the secretome of γ-irradiated PBMCs displayed higher pro-angiogenic activity in an aortic ring assay. Scanning electron microscopy and image stream analysis of γ-irradiated PBMCs revealed distinct morphological changes, indicative for apoptotic and necroptotic cell death. While inhibition of apoptosis had no effect on the pro-angiogenic activity of the secretome, inhibiting necroptosis in stressed PBMCs abolished blood vessel sprouting. Mechanistically, we identified tumor necrosis factor (TNF) receptor superfamily member 1B as the main driver of necroptosis in response to γ-irradiation in PBMCs, which was most likely mediated via membrane-bound TNF-α. In conclusion, our study demonstrates that the pro-angiogenic activity of the secretome of γ-irradiated PBMCs requires interplay of different PBMC subpopulations. Furthermore, we show that TNF-dependent necroptosis is an indispensable molecular process for conferring tissue-regenerative activity and for the pro-angiogenic potential of the PBMC secretome. These findings contribute to a better understanding of secretome-based therapies in regenerative medicine.
Collapse
Affiliation(s)
- Elisabeth Simader
- Department of Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria.,Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria.,FFG Project 852748 "APOSEC", Medical University of Vienna, Vienna, Austria
| | - Lucian Beer
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria.,Department of Radiology and Cancer Research UK Cambridge Center, Cambridge, CB2 0QQ, UK
| | - Maria Laggner
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria.,FFG Project 852748 "APOSEC", Medical University of Vienna, Vienna, Austria.,Vienna Business Agency Project 2343727 "APOSEC to clinic", Medical University Vienna, Vienna, Austria
| | - Vera Vorstandlechner
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria.,FFG Project 852748 "APOSEC", Medical University of Vienna, Vienna, Austria.,Vienna Business Agency Project 2343727 "APOSEC to clinic", Medical University Vienna, Vienna, Austria
| | - Alfred Gugerell
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria.,FFG Project 852748 "APOSEC", Medical University of Vienna, Vienna, Austria.,Vienna Business Agency Project 2343727 "APOSEC to clinic", Medical University Vienna, Vienna, Austria
| | - Michael Erb
- Synlab Analytics and Services Switzerland AG, Birsfelden, Switzerland
| | - Polina Kalinina
- Research Division of Biology and Pathobiology of the SkinDepartment of Dermatology, Research Division of Biology and Pathobiology of the Skin, Medical University of Vienna, Vienna, Austria
| | - Dragan Copic
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria.,FFG Project 852748 "APOSEC", Medical University of Vienna, Vienna, Austria.,Vienna Business Agency Project 2343727 "APOSEC to clinic", Medical University Vienna, Vienna, Austria
| | - Doris Moser
- Division of Oral and Maxillofacial Surgery, Medical University of Vienna, Vienna, Austria
| | - Andreas Spittler
- Research Laboratories, Core Facility Flow Cytometry, Medical University of Vienna, Vienna, Austria
| | - Erwin Tschachler
- Research Division of Biology and Pathobiology of the SkinDepartment of Dermatology, Research Division of Biology and Pathobiology of the Skin, Medical University of Vienna, Vienna, Austria
| | - Hendrik Jan Ankersmit
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria. .,FFG Project 852748 "APOSEC", Medical University of Vienna, Vienna, Austria. .,Vienna Business Agency Project 2343727 "APOSEC to clinic", Medical University Vienna, Vienna, Austria.
| | - Michael Mildner
- Research Division of Biology and Pathobiology of the SkinDepartment of Dermatology, Research Division of Biology and Pathobiology of the Skin, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
47
|
Drp1 and RB interaction to mediate mitochondria-dependent necroptosis induced by cadmium in hepatocytes. Cell Death Dis 2019; 10:523. [PMID: 31285421 PMCID: PMC6614419 DOI: 10.1038/s41419-019-1730-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 06/04/2019] [Accepted: 06/07/2019] [Indexed: 12/22/2022]
Abstract
Mitochondrial quality control (MQC) is implicated in cell death induced by heavy metal pollutants. Dynamin-related protein 1 (Drp1) regulates mitochondrial fission, which is an important part of MQC. Retinoblastoma (RB) protein can regulate MQC in a transcription-independent manner. Necroptosis plays a critical role in hepatic pathologies such as inflammatory, infectious, and xenobiotics-induced injury and diseases. We aimed to explore the role and mechanism of Drp1 interaction with RB in hepatocyte's necroptosis caused by cadmium (Cd). CdCl2 was employed to expose to Institute of Cancer Research (ICR) mice and human hepatic L02 cells. CdCl2 exposure induced necroptosis and hepatic injury both in vivo and in vitro. Moreover, Drp1 and RB protein were up-regulated and translocated to mitochondria in CdCl2-exposed hepatocytes. Inhibition of Drp1 with siRNA (siDNM1L) or inhibitors not only suppressed the RB expression and its mitochondrial translocation, but also alleviated MQC disorder, necroptosis, and hepatotoxicity caused by CdCl2. Moreover, blocking Drp1 with metformin rescued necroptosis and hepatic injury triggered by CdCl2. RB was proved to directly interact with Drp1 at mitochondria to form a complex which then bound to receptor interaction protein kinase (RIPK3) and enhanced the formation of necrosome after CdCl2 exposure. In summary, we found a new molecular mechanism of regulated cell death that Drp1 interacted with RB and promoted them mitochondrial translocation to mediate necroptosis and hepatic injury in hepatocytes induced by Cd-exposure. The mitochondrial Drp1-RB axis would be a novel target for the protection cells from xenobiotics triggering hepatic injury and diseases involved in necroptosis.
Collapse
|
48
|
Khandia R, Dadar M, Munjal A, Dhama K, Karthik K, Tiwari R, Yatoo MI, Iqbal HMN, Singh KP, Joshi SK, Chaicumpa W. A Comprehensive Review of Autophagy and Its Various Roles in Infectious, Non-Infectious, and Lifestyle Diseases: Current Knowledge and Prospects for Disease Prevention, Novel Drug Design, and Therapy. Cells 2019; 8:674. [PMID: 31277291 PMCID: PMC6678135 DOI: 10.3390/cells8070674] [Citation(s) in RCA: 164] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/04/2019] [Accepted: 06/04/2019] [Indexed: 02/05/2023] Open
Abstract
Autophagy (self-eating) is a conserved cellular degradation process that plays important roles in maintaining homeostasis and preventing nutritional, metabolic, and infection-mediated stresses. Autophagy dysfunction can have various pathological consequences, including tumor progression, pathogen hyper-virulence, and neurodegeneration. This review describes the mechanisms of autophagy and its associations with other cell death mechanisms, including apoptosis, necrosis, necroptosis, and autosis. Autophagy has both positive and negative roles in infection, cancer, neural development, metabolism, cardiovascular health, immunity, and iron homeostasis. Genetic defects in autophagy can have pathological consequences, such as static childhood encephalopathy with neurodegeneration in adulthood, Crohn's disease, hereditary spastic paraparesis, Danon disease, X-linked myopathy with excessive autophagy, and sporadic inclusion body myositis. Further studies on the process of autophagy in different microbial infections could help to design and develop novel therapeutic strategies against important pathogenic microbes. This review on the progress and prospects of autophagy research describes various activators and suppressors, which could be used to design novel intervention strategies against numerous diseases and develop therapeutic drugs to protect human and animal health.
Collapse
Affiliation(s)
- Rekha Khandia
- Department of Genetics, Barkatullah University, Bhopal 462 026, Madhya Pradesh, India
| | - Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj 31975/148, Iran
| | - Ashok Munjal
- Department of Genetics, Barkatullah University, Bhopal 462 026, Madhya Pradesh, India.
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243 122, Uttar Pradesh, India.
| | - Kumaragurubaran Karthik
- Central University Laboratory, Tamil Nadu Veterinary and Animal Sciences University, Madhavaram Milk Colony, Chennai, Tamil Nadu 600051, India
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and Immunology, College of Veterinary Sciences, UP Pandit Deen Dayal Upadhayay Pashu Chikitsa Vigyan Vishwavidyalay Evum Go-Anusandhan Sansthan (DUVASU), Mathura, Uttar Pradesh 281 001, India
| | - Mohd Iqbal Yatoo
- Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Shalimar, Srinagar 190025, Jammu and Kashmir, India
| | - Hafiz M N Iqbal
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Monterrey, Ave. Eugenio Garza Sada 2501, Monterrey, N. L., CP 64849, Mexico
| | - Karam Pal Singh
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243 122, Uttar Pradesh, India
| | - Sunil K Joshi
- Department of Pediatrics, Division of Hematology, Oncology and Bone Marrow Transplantation, University of Miami School of Medicine, Miami, FL 33136, USA.
| | - Wanpen Chaicumpa
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
49
|
Pan-caspase inhibitors induce necroptosis via ROS-mediated activation of mixed lineage kinase domain-like protein and p38 in classically activated macrophages. Exp Cell Res 2019; 380:171-179. [DOI: 10.1016/j.yexcr.2019.04.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 04/18/2019] [Accepted: 04/23/2019] [Indexed: 11/22/2022]
|
50
|
Shashaty MGS, Reilly JP, Faust HE, Forker CM, Ittner CAG, Zhang PX, Hotz MJ, Fitzgerald D, Yang W, Anderson BJ, Holena DN, Lanken PN, Christie JD, Meyer NJ, Mangalmurti NS. Plasma receptor interacting protein kinase-3 levels are associated with acute respiratory distress syndrome in sepsis and trauma: a cohort study. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2019; 23:235. [PMID: 31253195 PMCID: PMC6599265 DOI: 10.1186/s13054-019-2482-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 05/19/2019] [Indexed: 02/06/2023]
Abstract
Background Necroptosis, a form of programmed cell death mediated by receptor interacting serine/threonine-protein kinase-3 (RIPK3), is implicated in murine models of acute respiratory distress syndrome (ARDS). We hypothesized that plasma RIPK3 concentrations in sepsis and trauma would be associated with ARDS development and that plasma RIPK3 would reflect changes in lung tissue RIPK3 in a murine model of systemic inflammation. Methods We utilized prospective cohort studies of critically ill sepsis (n = 120) and trauma (n = 180) patients and measured plasma RIPK3 at presentation and 48 h. Patients were followed for 6 days for ARDS by the Berlin definition. We used multivariable logistic regression to determine the association of plasma RIPK3 with ARDS in each cohort, adjusting for confounders. In mice, we determined whether plasma and lung tissue RIPK3 levels rise concomitantly 4 h after injection with lipopolysaccharide and ZVAD-FMK, an apoptosis inhibitor. Results The change in plasma RIPK3 from presentation to 48 h (ΔRIPK3) was associated with ARDS in sepsis (OR 1.30, 95% CI 1.03–1.63, per ½ standard deviation) and trauma (OR 1.79, 95% CI 1.33–2.40). This association was not evident for presentation RIPK3 levels. Secondary analyses showed similar findings for the association of ΔRIPK3 with acute kidney injury and 30-day mortality. Mice injected with lipopolysaccharide and ZVAD-FMK had significantly higher plasma (p < 0.001) and lung (p = 0.005) RIPK3 than control mice. Conclusions The change in plasma RIPK3 from presentation to 48 h in both sepsis and trauma patients is independently associated with ARDS, and plasma RIPK3 may reflect RIPK3 activity in lung tissue. Electronic supplementary material The online version of this article (10.1186/s13054-019-2482-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Michael G S Shashaty
- Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, 5039 W Gates Building, 3600 Spruce Street, Philadelphia, PA, 19104, USA. .,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, USA. .,Center for Translational Lung Biology, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, USA.
| | - John P Reilly
- Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, 5039 W Gates Building, 3600 Spruce Street, Philadelphia, PA, 19104, USA.,Center for Translational Lung Biology, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, USA
| | - Hilary E Faust
- Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, 5039 W Gates Building, 3600 Spruce Street, Philadelphia, PA, 19104, USA.,Center for Translational Lung Biology, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, USA
| | - Caitlin M Forker
- Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, 5039 W Gates Building, 3600 Spruce Street, Philadelphia, PA, 19104, USA
| | - Caroline A G Ittner
- Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, 5039 W Gates Building, 3600 Spruce Street, Philadelphia, PA, 19104, USA
| | - Peggy X Zhang
- Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, 5039 W Gates Building, 3600 Spruce Street, Philadelphia, PA, 19104, USA
| | - Meghan J Hotz
- Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, 5039 W Gates Building, 3600 Spruce Street, Philadelphia, PA, 19104, USA
| | - David Fitzgerald
- Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, 5039 W Gates Building, 3600 Spruce Street, Philadelphia, PA, 19104, USA
| | - Wei Yang
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, USA
| | - Brian J Anderson
- Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, 5039 W Gates Building, 3600 Spruce Street, Philadelphia, PA, 19104, USA.,Center for Translational Lung Biology, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, USA
| | - Daniel N Holena
- Division of Traumatology, Surgical Critical Care, and Emergency Surgery, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, USA
| | - Paul N Lanken
- Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, 5039 W Gates Building, 3600 Spruce Street, Philadelphia, PA, 19104, USA
| | - Jason D Christie
- Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, 5039 W Gates Building, 3600 Spruce Street, Philadelphia, PA, 19104, USA.,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, USA.,Center for Translational Lung Biology, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, USA
| | - Nuala J Meyer
- Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, 5039 W Gates Building, 3600 Spruce Street, Philadelphia, PA, 19104, USA.,Center for Translational Lung Biology, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, USA
| | - Nilam S Mangalmurti
- Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, 5039 W Gates Building, 3600 Spruce Street, Philadelphia, PA, 19104, USA.,Center for Translational Lung Biology, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, USA
| |
Collapse
|