1
|
Wagner J, Vredevoogd D, Yu X, Lu D, Peeper DS, Hermanns HM, Wang J, Wajant H, Siegmund D. TRAF2 and RIPK1 redundantly mediate classical NFκB signaling by TNFR1 and CD95-type death receptors. Cell Death Dis 2025; 16:35. [PMID: 39837830 PMCID: PMC11751453 DOI: 10.1038/s41419-024-07325-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 12/05/2024] [Accepted: 12/27/2024] [Indexed: 01/23/2025]
Abstract
This study suggests a modified model of TNFR1-induced complex I-mediated NFκB signaling. Evaluation of a panel of five tumor cell lines (HCT116-PIK3CAmut, SK-MEL-23, HeLa-RIPK3, HT29, D10) with TRAF2 knockout revealed in two cell lines (HT29, HeLa-RIPK3) a sensitizing effect for death receptor-induced necroptosis and in one cell line (D10) a mild sensitization for TNFR1-induced apoptosis. TRAF2 deficiency inhibited death receptor-induced classical NFκB-mediated production of IL-8 only in a subset of cell lines and only partly. TRAF5, furthermore, failed to improve DR-induced NFκB signaling in HCT116-PIK3CAmut and HCT116-PIK3CAmut-TRAF2KO cells. These findings argue for a non-obligatory role of TRAF2 in death receptor-induced classical NFκB signaling. Similar as in TRAF2-deficient cells, TNF- and CD95L-induced NFκB signaling was found to be only poorly affected in RIPK1KO cells and in cells treated with the RIPK1-specific PROTAC LD4172. Intriguingly, however, death receptor-induced NFκB signaling was completely inhibited in HCT116-PIK3CAmut cells double deficient for TRAF2 and RIPK1 and in TRAF2-deficient cells treated with LD4172. Moreover, with exception of recruitment of TRADD, acting upstream to TRAF2 and parallel to RIPK1, TNFR1 signaling complex formation was abrogated in TRAF2-RIPK1 DKO cells. Based on our findings, two distinguishable types of TNFR1-interacting complexes promote TNF-induced NFκB signaling: First, a TRADD-TRAF2/cIAP utilizing complex Ia which becomes evident in RIPK1-deficient cells. Second, a non-modified RIPK1 utilizing complex Ib which acts in TRADD- or TRAF2-deficient cells. Complex Ia and Ib may furthermore interact and cooperate to ubiquitinate RIPK1 resulting in a modified complex Ia/b preventing complex Ia and Ib to convert to the established TNFR1-induced cytotoxic complexes IIa and IIb.
Collapse
Affiliation(s)
- Jennifer Wagner
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Auvera Haus Grombühlstraße 12, 97080, Würzburg, Germany
| | - David Vredevoogd
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherlands
| | - Xin Yu
- The Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Dong Lu
- The Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Daniel S Peeper
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherlands
| | - Heike M Hermanns
- Division of Hepatology, Department of Internal Medicine II, University Hospital Würzburg, Auvera Haus Grombühlstraße 12, 97080, Würzburg, Germany
| | - Jin Wang
- The Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for NextGen Therapeutics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Harald Wajant
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Auvera Haus Grombühlstraße 12, 97080, Würzburg, Germany.
| | - Daniela Siegmund
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Auvera Haus Grombühlstraße 12, 97080, Würzburg, Germany
| |
Collapse
|
2
|
Niu X, You Q, Hou K, Tian Y, Wei P, Zhu Y, Gao B, Ashrafizadeh M, Aref AR, Kalbasi A, Cañadas I, Sethi G, Tergaonkar V, Wang L, Lin Y, Kang D, Klionsky DJ. Autophagy in cancer development, immune evasion, and drug resistance. Drug Resist Updat 2025; 78:101170. [PMID: 39603146 DOI: 10.1016/j.drup.2024.101170] [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: 07/16/2024] [Revised: 10/22/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024]
Abstract
Macroautophagy/autophagy is a highly conserved evolutionary mechanism involving lysosomes for the degradation of cytoplasmic components including organelles. The constitutive, basal level of autophagy is fundamental for preserving cellular homeostasis; however, alterations in autophagy can cause disease pathogenesis, including cancer. The role of autophagy in cancer is particularly complicated, since this process acts both as a tumor suppressor in precancerous stages but facilitates tumor progression during carcinogenesis and later stages of cancer progression. This shift between anti-tumor and pro-tumor roles may be influenced by genetic and environmental factors modulating key pathways such as those involving autophagy-related proteins, the PI3K-AKT-MTOR axis, and AMPK, which often show dysregulation in tumors. Autophagy regulates various cellular functions, including metabolism of glucose, glutamine, and lipids, cell proliferation, metastasis, and several types of cell death (apoptosis, ferroptosis, necroptosis and immunogenic cell death). These multifaceted roles demonstrate the potential of autophagy to affect DNA damage repair, cell death pathways, proliferation and survival, which are critical in determining cancer cells' response to chemotherapy. Therefore, targeting autophagy pathways presents a promising strategy to combat chemoresistance, as one of the major reasons for the failure in cancer patient treatment. Furthermore, autophagy modulates immune evasion and the function of immune cells such as T cells and dendritic cells, influencing the tumor microenvironment and cancer's biological behavior. However, the therapeutic targeting of autophagy is complex due to its dual role in promoting survival and inducing cell death in cancer cells, highlighting the need for strategies that consider both the beneficial and detrimental effects of autophagy modulation in cancer therapy. Hence, both inducers and inhibitors of autophagy have been introduced for the treatment of cancer. This review emphasizes the intricate interplay between autophagy, tumor biology, and immune responses, offering insights into potential therapeutic approaches that deploy autophagy in the cancer suppression.
Collapse
Affiliation(s)
- Xuegang Niu
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Qi You
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Kaijian Hou
- School of Public Health(Long Hu people hospital), Shantou University, Shantou, 515000, Guangdong, China
| | - Yu Tian
- School of Public Health, Benedictine University, Lisle, IL 60532, USA
| | - Penghui Wei
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Yang Zhu
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Bin Gao
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Milad Ashrafizadeh
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China
| | - Amir Reza Aref
- VitroVision Department, DeepkinetiX, Inc, Boston, MA, USA
| | - Alireza Kalbasi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Israel Cañadas
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Gautam Sethi
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore 117600, Singapore
| | - Vinay Tergaonkar
- Laboratory of NF-κB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A⁎STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Lingzhi Wang
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore 117600, Singapore
| | - Yuanxiang Lin
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| | - Dezhi Kang
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| | - Daniel J Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
3
|
Bilsborough JM, Targan SR. TL1A: A model for a precision medicine approach in the treatment of Crohn's disease and ulcerative colitis. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 101:287-299. [PMID: 39521604 DOI: 10.1016/bs.apha.2024.10.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Inflammatory bowel disease (IBD) is a collective term for chronic inflammatory diseases of the intestinal tract. The term IBD encompasses two main forms, Crohn's disease (CD) and Ulcerative colitis (UC). CD is characterized by inflammation throughout the length of the gut, especially the ileum and colon, and is often complicated with fistulae and/or intestinal strictures. Ulcerative colitis (UC) is inflammatory disease restricted to the colon and rectum. In practice however, IBD is a heterogenous disease with CD and UC representing the extremes of a continuum of diseases with varied clinical presentation, including disease location, severity, and manifestation of extraintestinal diseases. This disease heterogeneity poses a challenge to successful and efficacious therapeutic treatment as the etiology driving disease in individual patients is unknown and likely to be multifactorial, including genetic predisposition, environmental factors such as the microbiota, as well as social behaviors such as smoking and diet. Precision medicine provides a strategy to account for disease heterogeneity and diverse etiology to select for patients most likely to respond to a given therapeutic. In this chapter we present an example of the development of a novel antibody therapeutic, Tulisokibart, as a model for a Precision Medicine approach to the successful treatment of patients with IBD.
Collapse
Affiliation(s)
- Janine M Bilsborough
- F. Widjaja Inflammatory Bowel Disease Institute, Los Angeles, CA, United States.
| | - Stephan R Targan
- F. Widjaja Inflammatory Bowel Disease Institute, Los Angeles, CA, United States
| |
Collapse
|
4
|
Liu X, Zhang J, Zhang D, Pan Y, Zeng R, Xu C, Shi S, Xu J, Qi Q, Dong X, Wang J, Liu T, Dong L. Necroptosis plays a role in TL1A-induced airway inflammation and barrier damage in asthma. Respir Res 2024; 25:271. [PMID: 38987753 PMCID: PMC11238433 DOI: 10.1186/s12931-024-02900-4] [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: 01/09/2024] [Accepted: 06/29/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND Airway epithelial cell (AEC) necroptosis contributes to airway allergic inflammation and asthma exacerbation. Targeting the tumor necrosis factor-like ligand 1 A (TL1A)/death receptor 3 (DR3) axis has a therapeutic effect on asthmatic airway inflammation. The role of TL1A in mediating necroptosis of AECs challenged with ovalbumin (OVA) and its contribution to airway inflammation remains unclear. METHODS We evaluated the expression of the receptor-interacting serine/threonine-protein kinase 3(RIPK3) and the mixed lineage kinase domain-like protein (MLKL) in human serum and lung, and histologically verified the level of MLKL phosphorylation in lung tissue from asthmatics and OVA-induced mice. Next, using MLKL knockout mice and the RIPK3 inhibitor GSK872, we investigated the effects of TL1A on airway inflammation and airway barrier function through the activation of necroptosis in experimental asthma. RESULTS High expression of necroptosis marker proteins was observed in the serum of asthmatics, and necroptosis was activated in the airway epithelium of both asthmatics and OVA-induced mice. Blocking necroptosis through MLKL knockout or RIPK3 inhibition effectively attenuated parabronchial inflammation, mucus hypersecretion, and airway collagen fiber accumulation, while also suppressing type 2 inflammatory factors secretion. In addition, TL1A/ DR3 was shown to act as a death trigger for necroptosis in the absence of caspases by silencing or overexpressing TL1A in HBE cells. Furthermore, the recombinant TL1A protein was found to induce necroptosis in vivo, and knockout of MLKL partially reversed the pathological changes induced by TL1A. The necroptosis induced by TL1A disrupted the airway barrier function by decreasing the expression of tight junction proteins zonula occludens-1 (ZO-1) and occludin, possibly through the activation of the NF-κB signaling pathway. CONCLUSIONS TL1A-induced airway epithelial necroptosis plays a significant role in promoting airway inflammation and barrier dysfunction in asthma. Inhibition of the TL1A-induced necroptosis pathway could be a promising therapeutic strategy.
Collapse
Affiliation(s)
- Xiaofei Liu
- Department of Respiratory, Shandong Institute of Respiratory Diseases, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, China
| | - Jintao Zhang
- Department of Respiratory, Shandong Institute of Respiratory Diseases, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, China
| | - Dong Zhang
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Yun Pan
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Rong Zeng
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Changjuan Xu
- Department of Respiratory, Shandong Institute of Respiratory Diseases, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, China
| | - Shuochuan Shi
- Department of Respiratory, Shandong Institute of Respiratory Diseases, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, China
| | - Jiawei Xu
- Department of Respiratory, Shandong Institute of Respiratory Diseases, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, China
| | - Qian Qi
- Department of Respiratory, Shandong Institute of Respiratory Diseases, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, China
| | - Xueli Dong
- Department of Respiratory, Shandong Institute of Respiratory Diseases, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, China
| | - Junfei Wang
- Department of Respiratory and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Tian Liu
- Department of Respiratory and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Liang Dong
- Department of Respiratory, Shandong Institute of Respiratory Diseases, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, China.
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China.
| |
Collapse
|
5
|
Solon M, Ge N, Hambro S, Haller S, Jiang J, Baca M, Preston J, Maltzman A, Wickliffe KE, Liang Y, Reja R, Nickles D, Newton K, Webster JD. ZBP1 and TRIF trigger lethal necroptosis in mice lacking caspase-8 and TNFR1. Cell Death Differ 2024; 31:672-682. [PMID: 38548850 PMCID: PMC11093969 DOI: 10.1038/s41418-024-01286-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 03/13/2024] [Accepted: 03/20/2024] [Indexed: 05/16/2024] Open
Abstract
Necroptosis is a lytic form of cell death that is mediated by the kinase RIPK3 and the pseudokinase MLKL when caspase-8 is inhibited downstream of death receptors, toll-like receptor 3 (TLR3), TLR4, and the intracellular Z-form nucleic acid sensor ZBP1. Oligomerization and activation of RIPK3 is driven by interactions with the kinase RIPK1, the TLR adaptor TRIF, or ZBP1. In this study, we use immunohistochemistry (IHC) and in situ hybridization (ISH) assays to generate a tissue atlas characterizing RIPK1, RIPK3, Mlkl, and ZBP1 expression in mouse tissues. RIPK1, RIPK3, and Mlkl were co-expressed in most immune cell populations, endothelial cells, and many barrier epithelia. ZBP1 was expressed in many immune populations, but had more variable expression in epithelia compared to RIPK1, RIPK3, and Mlkl. Intriguingly, expression of ZBP1 was elevated in Casp8-/- Tnfr1-/- embryos prior to their succumbing to aberrant necroptosis around embryonic day 15 (E15). ZBP1 contributed to this embryonic lethality because rare Casp8-/- Tnfr1-/- Zbp1-/- mice survived until after birth. Necroptosis mediated by TRIF contributed to the demise of Casp8-/- Tnfr1-/- Zbp1-/- pups in the perinatal period. Of note, Casp8-/- Tnfr1-/- Trif-/- Zbp1-/- mice exhibited autoinflammation and morbidity, typically within 5-7 weeks of being born, which is not seen in Casp8-/- Ripk1-/- Trif-/- Zbp1-/-, Casp8-/- Ripk3-/-, or Casp8-/- Mlkl-/- mice. Therefore, after birth, loss of caspase-8 probably unleashes RIPK1-dependent necroptosis driven by death receptors other than TNFR1.
Collapse
Affiliation(s)
- Margaret Solon
- Department of Pathology, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Nianfeng Ge
- Department of Pathology, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Shannon Hambro
- Department of Pathology, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Susan Haller
- Department of Pathology, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Jian Jiang
- Department of Pathology, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Miriam Baca
- Department of Pathology, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Jessica Preston
- Department of Pathology, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Allie Maltzman
- Department of Physiological Chemistry, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Katherine E Wickliffe
- Department of Physiological Chemistry, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Yuxin Liang
- Department of Microchemistry, Proteomics, Lipidomics, and Next Generation Sequencing, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Rohit Reja
- Department of Physiological Chemistry, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
- Department of Oncology Bioinformatics, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Dorothee Nickles
- Department of Oncology Bioinformatics, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
- Department of Translational Oncology, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Kim Newton
- Department of Physiological Chemistry, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA.
| | - Joshua D Webster
- Department of Pathology, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA.
| |
Collapse
|
6
|
Yin C, Wang J, Zhang Y, Zhang X, Zhao W, Shen Y, Liu S, Liu S. Death receptor 3 is involved in preeclampsia through regulating placental trophoblast cell physiology by inactivating the PI3K/AKT pathway. Immun Inflamm Dis 2023; 11:e995. [PMID: 37773709 PMCID: PMC10523955 DOI: 10.1002/iid3.995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 08/09/2023] [Accepted: 08/16/2023] [Indexed: 10/01/2023] Open
Abstract
BACKGROUND Preeclampsia (PE) is a pregnancy related disease that affects about 5% of pregnancies. Death receptor 3 (DR3) expression is significantly elevated in both placental tissue and plasma of PE patients. However, whether DR3 was involved in trophoblasts in pathogenesis of PE are not well elucidated. OBJECTIVE Our research was designed to illustrate the biological roles of DR3 in placental trophoblasts, as well as explain its relevant mechanisms. METHODS HTR-8/SVneo cells viability, migration, invasion, and apoptosis were assessed using MTT, Transwell assay, and flow cytometry analysis, respectively. Levels of DR3, PI3K, and AKT in HTR-8/SVneo cells were analyzed via reverse transcription-quantitative polymerase chain reaction assay. Western blot analysis was utilized to assess DR3, p-PI3K, p-AKT, PI3K, and AKT protein expression. RESULTS Upregulation of DR3 obviously inhibited HTR-8/SVneo cells viability, migration, and invasion, as well as promoted HTR-8/SVneo cells apoptosis, as opposed to the control-plasmid group. We also found that DR3-plasmid enhanced cleaved-caspase3 expression, reduced p-PI3K and p-AKT protein expression, and p-PI3K/PI3K or p-AKT/AKT ratio in HTR-8/SVneo cells. Importantly, IGF-1, a PI3K/AKT signaling pathway agonist, partially reversed the effects of DR3-plasmid on the cell viability, migration, invasion, apoptosis, and PI3K/AKT signal pathway in HTR-8/SVneo cells. CONCLUSION DR3 was involved in PE through regulating placental trophoblast cell physiology via PI3K/AKT pathway, which might be a promising therapeutic target for PE therapy.
Collapse
Affiliation(s)
- Cheng Yin
- Obstetrics DepartmentThe Third Affiliated Hospital of Qiqihar Medical UniversityQiqiharChina
| | - Jiahui Wang
- Obstetrics DepartmentThe Third Affiliated Hospital of Qiqihar Medical UniversityQiqiharChina
| | - Yu Zhang
- Obstetrics DepartmentThe Third Affiliated Hospital of Qiqihar Medical UniversityQiqiharChina
| | - Xinping Zhang
- Obstetrics DepartmentThe Third Affiliated Hospital of Qiqihar Medical UniversityQiqiharChina
| | - Wei Zhao
- Gynecology DepartmentThe Third Affiliated Hospital of Qiqihar Medical UniversityQiqiharChina
| | - Yanxiang Shen
- Cardiovascular Internal Medicine DepartmentThe Third Affiliated Hospital of Qiqihar Medical UniversityQiqiharChina
| | - Shi Liu
- Central LaboratoryThe Third Affiliated Hospital of Qiqihar Medical UniversityQiqiharChina
| | - Su Liu
- Obstetrics DepartmentThe Third Affiliated Hospital of Qiqihar Medical UniversityQiqiharChina
| |
Collapse
|
7
|
Liu X, Liu L, Wang X, Jin Y, Wang S, Xie Q, Jin Y, Zhang M, Liu Y, Li J, Wang Z, Fu X, Jin CY. Necroptosis inhibits autophagy by regulating the formation of RIP3/p62/Keap1 complex in shikonin-induced ROS dependent cell death of human bladder cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 118:154943. [PMID: 37421765 DOI: 10.1016/j.phymed.2023.154943] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 06/02/2023] [Accepted: 06/25/2023] [Indexed: 07/10/2023]
Abstract
BACKGROUND Shikonin, a natural naphthoquinone compound, has a wide range of pharmacological effects, but its anti-tumor effect and underlying mechanisms in bladder cancer remain unclear. PURPOSE We aimed to investigate the role of shikonin in bladder cancer in vitro and in vivo in order to broaden the scope of shikonin's clinical application. STUDY DESIGN AND METHODS We performed MTT and colony formation to detect the inhibiting effect of shikonin on bladder cancer cells. ROS staining and flow cytometry assays were performed to detect the accumulation of ROS. Western blotting, siRNA and immunoprecipitation were used to evaluate the effect of necroptosis in bladder cancer cells. Transmission electron microscopy and immunofluorescence were used to examine the effect of autophagy. Nucleoplasmic separation and other pharmacological experimental methods described were used to explore the Nrf2 signal pathway and the crosstalk with necroptosis and autophagy. We established a subcutaneously implanted tumor model and performed immunohistochemistry assays to study the effects and the underlying mechanisms of shikonin on bladder cancer cells in vivo. RESULTS The results showed that shikonin has a selective inhibitory effect on bladder cancer cells and has no toxicity on normal bladder epithelial cells. Mechanically, shikonin induced necroptosis and impaired autophagic flux via ROS generation. The accumulation of autophagic biomarker p62 elevated p62/Keap1 complex and activated the Nrf2 signaling pathway to fight against ROS. Furthermore, crosstalk between necroptosis and autophagy was present, we found that RIP3 may be involved in autophagosomes and be degraded by autolysosomes. We found for the first time that shikonin-induced activation of RIP3 may disturb the autophagic flux, and inhibiting RIP3 and necroptosis could accelerate the conversion of autophagosome to autolysosome and further activate autophagy. Therefore, on the basis of RIP3/p62/Keap1 complex regulatory system, we further combined shikonin with late autophagy inhibitor(chloroquine) to treat bladder cancer and achieved a better inhibitory effect. CONCLUSION In conclusion, shikonin could induce necroptosis and impaired autophagic flux through RIP3/p62/Keap1 complex regulatory system, necroptosis could inhibit the process of autophagy via RIP3. Combining shikonin with late autophagy inhibitor could further activate necroptosis via disturbing RIP3 degradation in bladder cancer in vitro and in vivo.
Collapse
Affiliation(s)
- Xiaojie Liu
- Department of Microbiology and biochemical pharmacy, College of pharmacy, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan Province, 450001, China
| | - Lu Liu
- Department of Microbiology and biochemical pharmacy, College of pharmacy, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan Province, 450001, China
| | - Xu Wang
- Department of Microbiology and biochemical pharmacy, College of pharmacy, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan Province, 450001, China
| | - Yubin Jin
- The Second Senior High School of Tumen City, Yuegong Street, Tumen, Jilin Province, 137200, China
| | - Shuang Wang
- Department of Microbiology and biochemical pharmacy, College of pharmacy, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan Province, 450001, China
| | - Qin Xie
- Department of Microbiology and biochemical pharmacy, College of pharmacy, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan Province, 450001, China
| | - Yanhe Jin
- Department of Microbiology and biochemical pharmacy, College of pharmacy, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan Province, 450001, China
| | - Mengli Zhang
- Department of Microbiology and biochemical pharmacy, College of pharmacy, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan Province, 450001, China
| | - Yunhe Liu
- Department of Microbiology and biochemical pharmacy, College of pharmacy, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan Province, 450001, China
| | - Jinfeng Li
- Department of Kidney Transplantation, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Road, Erqi District, Zhengzhou, Henan Province, 450001, China
| | - Zhenya Wang
- Department of Microbiology and biochemical pharmacy, College of pharmacy, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan Province, 450001, China
| | - Xiangjing Fu
- Department of Microbiology and biochemical pharmacy, College of pharmacy, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan Province, 450001, China.
| | - Cheng-Yun Jin
- Department of Microbiology and biochemical pharmacy, College of pharmacy, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan Province, 450001, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, 450052, China.
| |
Collapse
|
8
|
Xu WD, Li R, Huang AF. Role of TL1A in Inflammatory Autoimmune Diseases: A Comprehensive Review. Front Immunol 2022; 13:891328. [PMID: 35911746 PMCID: PMC9329929 DOI: 10.3389/fimmu.2022.891328] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/02/2022] [Indexed: 01/10/2023] Open
Abstract
TL1A, also called TNFSF15, is a member of tumor necrosis factor family. It is expressed in different immune cell, such as monocyte, macrophage, dendritic cell, T cell and non-immune cell, for example, synovial fibroblast, endothelial cell. TL1A competitively binds to death receptor 3 or decoy receptor 3, providing stimulatory signal for downstream signaling pathways, and then regulates proliferation, activation, apoptosis of and cytokine, chemokine production in effector cells. Recent findings showed that TL1A was abnormally expressed in autoimmune diseases, including rheumatoid arthritis, inflammatory bowel disease, psoriasis, primary biliary cirrhosis, systemic lupus erythematosus and ankylosing spondylitis. In vivo and in vitro studies further demonstrated that TL1A was involved in development and pathogenesis of these diseases. In this study, we comprehensively discussed the complex immunological function of TL1A and focused on recent findings of the pleiotropic activity conducted by TL1A in inflammatory autoimmune disease. Finish of the study will provide new ideas for developing therapeutic strategies for these diseases by targeting TL1A.
Collapse
Affiliation(s)
- Wang-Dong Xu
- Department of Evidence-Based Medicine, Southwest Medical University, Luzhou, China
| | - Rong Li
- Department of Evidence-Based Medicine, Southwest Medical University, Luzhou, China
| | - An-Fang Huang
- Department of Rheumatology and Immunology, Affiliated Hospital of Southwest Medical University, Luzhou, China
- *Correspondence: An-Fang Huang,
| |
Collapse
|
9
|
Huang Y, Feng Y, Cui L, Yang L, Zhang Q, Zhang J, Jiang X, Zhang X, Lv Y, Jia JZ, Zhang DX, Huang YS. Autophagy-Related LC3 Accumulation Interacted Directly With LIR Containing RIPK1 and RIPK3, Stimulating Necroptosis in Hypoxic Cardiomyocytes. Front Cell Dev Biol 2021; 9:679637. [PMID: 34368130 PMCID: PMC8344065 DOI: 10.3389/fcell.2021.679637] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 06/16/2021] [Indexed: 11/13/2022] Open
Abstract
The exact relationships and detailed mechanisms between autophagy and necroptosis remain obscure. Here, we demonstrated the link between accumulated autophagosome and necroptosis by intervening with autophagic flux. We first confirmed that the LC3 interacting region (LIR) domain is present in the protein sequences of RIPK1 and RIPK3. Mutual effects among LC3, RIPK1, and RIPK3 have been identified in myocardium and cardiomyocytes. Direct LC3-RIPK1 and LC3-RIPK3 interactions were confirmed by pull-down assays, and their interactions were deleted after LIR domain mutation. Moreover, after disrupting autophagic flux under normoxia with bafilomycin A1 treatment, or with LC3 or ATG5 overexpression adenovirus, RIPK1, RIPK3, p-RIPK3, and p-MLKL levels increased, suggesting necroptosis activation. Severe disruptions in autophagic flux were observed under hypoxia and bafilomycin A1 co-treated cardiomyocytes and myocardium and led to more significant activation of necroptosis. Conversely, after alleviating hypoxia-induced autophagic flux impairment with LC3 or ATG5 knockdown adenovirus, the effects of hypoxia on RIPK1 and RIPK3 levels were reduced, which resulted in decreased p-RIPK3 and p-MLKL. Furthermore, necroptosis was inhibited by siRNAs against RIPK1 and RIPK3 under hypoxia or normoxia. Based on our results, LIR domain mediated LC3-RIPK1 and LC3-RIPK3 interaction. Besides, autophagosome accumulation under hypoxia lead to necrosome formation and, in turn, necroptosis, while when autophagic flux was uninterrupted, RIPK1 and RIPK3 were cleared through an autophagy-related pathway which inhibited necroptosis. These findings provide novel insights for the role of LC3 in regulating cardiomyocyte necroptosis, indicating its therapeutic potential in the prevention and treatment of hypoxic myocardial injury and other hypoxia-related diseases.
Collapse
Affiliation(s)
- Yao Huang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burn Research, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yanhai Feng
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burn Research, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Lin Cui
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burn Research, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Lei Yang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burn Research, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Qiong Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burn Research, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Junhui Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burn Research, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xupin Jiang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burn Research, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xingyue Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burn Research, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yanling Lv
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burn Research, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jie-Zhi Jia
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burn Research, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Dong-Xia Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burn Research, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yue-Sheng Huang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burn Research, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Department of Wound Repair, and Institute of Wound Repair, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| |
Collapse
|
10
|
Necroptosis molecular mechanisms: Recent findings regarding novel necroptosis regulators. Exp Mol Med 2021; 53:1007-1017. [PMID: 34075202 PMCID: PMC8166896 DOI: 10.1038/s12276-021-00634-7] [Citation(s) in RCA: 160] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 02/05/2023] Open
Abstract
Necroptosis is a form of programmed necrosis that is mediated by various cytokines and pattern recognition receptors (PRRs). Cells dying by necroptosis show necrotic phenotypes, including swelling and membrane rupture, and release damage-associated molecular patterns (DAMPs), inflammatory cytokines, and chemokines, thereby mediating extreme inflammatory responses. Studies on gene knockout or necroptosis-specific inhibitor treatment in animal models have provided extensive evidence regarding the important roles of necroptosis in inflammatory diseases. The necroptosis signaling pathway is primarily modulated by activation of receptor-interacting protein kinase 3 (RIPK3), which phosphorylates mixed-lineage kinase domain-like protein (MLKL), mediating MLKL oligomerization. In the necroptosis process, these proteins are fine-tuned by posttranslational regulation via phosphorylation, ubiquitination, glycosylation, and protein-protein interactions. Herein, we review recent findings on the molecular regulatory mechanisms of necroptosis.
Collapse
|
11
|
Oriol-Tordera B, Olvera A, Duran-Castells C, Llano A, Mothe B, Massanella M, Dalmau J, Ganoza C, Sanchez J, Calle ML, Clotet B, Martinez-Picado J, Negredo E, Blanco J, Hartigan-O'Connor D, Brander C, Ruiz-Riol M. TL1A-DR3 Plasma Levels Are Predictive of HIV-1 Disease Control, and DR3 Costimulation Boosts HIV-1-Specific T Cell Responses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 205:3348-3357. [PMID: 33177161 PMCID: PMC7725879 DOI: 10.4049/jimmunol.2000933] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/02/2020] [Indexed: 12/12/2022]
Abstract
Relative control of HIV-1 infection has been linked to genetic and immune host factors. In this study, we analyzed 96 plasma proteome arrays from chronic untreated HIV-1-infected individuals using the classificatory random forest approach to discriminate between uncontrolled disease (plasma viral load [pVL] >50,000 RNA copies/ml; CD4 counts 283 cells/mm3, n = 47) and relatively controlled disease (pVL <10,000 RNA copies/ml; CD4 counts 657 cells/mm3, n = 49). Our analysis highlighted the TNF molecule's relevance, in particular, TL1A (TNFSF15) and its cognate DR3 (TNFSRF25), both of which increased in the relative virus control phenotype. DR3 levels (in plasma and PBMCs) were validated in unrelated cohorts (including long-term nonprogressors), thus confirming their independence from CD4 counts and pVL. Further analysis in combined antiretroviral treatment (cART)-treated individuals with a wide range of CD4 counts (137-1835 cells/mm3) indicated that neither TL1A nor DR3 levels reflected recovery of CD4 counts with cART. Interestingly, in cART-treated individuals, plasma TL1A levels correlated with regulatory T cell frequencies, whereas soluble DR3 was strongly associated with the abundance of effector HLA-DR+CD8+ T cells. A positive correlation was also observed between plasma DR3 levels and the HIV-1-specific T cell responses. In vitro, costimulation of PBMC with DR3-specific mAb increased the magnitude of HIV-1-specific responses. Finally, in splenocytes of DNA.HTI-vaccinated mice, costimulation of HTI peptides and a DR3 agonist (4C12) intensified the magnitude of T cell responses by 27%. These data describe the role of the TL1A-DR3 axis in the natural control of HIV-1 infection and point to the use of DR3 agonists in HIV-1 vaccine regimens.
Collapse
Affiliation(s)
- Bruna Oriol-Tordera
- Institut de Recerca de la Sida IrsiCaixa, Hospital Universitari Germans Trias i Pujol, Badalona, 08916 Barcelona, Spain
- Departament de Biologia Cellular, de Fisiologia i d'Immunologia, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193 Barcelona, Spain
| | - Alex Olvera
- Institut de Recerca de la Sida IrsiCaixa, Hospital Universitari Germans Trias i Pujol, Badalona, 08916 Barcelona, Spain
- Universitat de Vic - Universitat Central de Catalunya, Vic, 08500 Barcelona, Spain
| | - Clara Duran-Castells
- Institut de Recerca de la Sida IrsiCaixa, Hospital Universitari Germans Trias i Pujol, Badalona, 08916 Barcelona, Spain
- Departament de Biologia Cellular, de Fisiologia i d'Immunologia, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193 Barcelona, Spain
| | - Anuska Llano
- Institut de Recerca de la Sida IrsiCaixa, Hospital Universitari Germans Trias i Pujol, Badalona, 08916 Barcelona, Spain
| | - Beatriz Mothe
- Institut de Recerca de la Sida IrsiCaixa, Hospital Universitari Germans Trias i Pujol, Badalona, 08916 Barcelona, Spain
- Universitat de Vic - Universitat Central de Catalunya, Vic, 08500 Barcelona, Spain
- Fundació Lluita contra la Sida i les Malalties Infeccioses, Servei de Malalties Infecciones Hospital Universitari Germans Trias i Pujol, Badalona, 08916 Barcelona, Spain
| | - Marta Massanella
- Institut de Recerca de la Sida IrsiCaixa, Hospital Universitari Germans Trias i Pujol, Badalona, 08916 Barcelona, Spain
| | - Judith Dalmau
- Institut de Recerca de la Sida IrsiCaixa, Hospital Universitari Germans Trias i Pujol, Badalona, 08916 Barcelona, Spain
| | - Carmela Ganoza
- Asociación Civil Impacta Salud y Educacion, Lima 15063, Peru
- Facultad de Medicina Alberto Hurtado de la Universidad Peruana Cayetano Heredia, San Martín de Porres, Lima 15102, Peru
| | - Jorge Sanchez
- Asociación Civil Impacta Salud y Educacion, Lima 15063, Peru
- Department of Global Health, University of Washington, Seattle, WA 98195
- Centro de Investigaciones Tecnológicas, Biomédicas y Medioambientales, Bellavista, Lima 07006, Peru
| | - Maria Luz Calle
- Universitat de Vic - Universitat Central de Catalunya, Vic, 08500 Barcelona, Spain
| | - Bonaventura Clotet
- Institut de Recerca de la Sida IrsiCaixa, Hospital Universitari Germans Trias i Pujol, Badalona, 08916 Barcelona, Spain
- Universitat de Vic - Universitat Central de Catalunya, Vic, 08500 Barcelona, Spain
- Fundació Lluita contra la Sida i les Malalties Infeccioses, Servei de Malalties Infecciones Hospital Universitari Germans Trias i Pujol, Badalona, 08916 Barcelona, Spain
| | - Javier Martinez-Picado
- Institut de Recerca de la Sida IrsiCaixa, Hospital Universitari Germans Trias i Pujol, Badalona, 08916 Barcelona, Spain
- Universitat de Vic - Universitat Central de Catalunya, Vic, 08500 Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats, 08010 Barcelona, Spain
| | - Eugènia Negredo
- Universitat de Vic - Universitat Central de Catalunya, Vic, 08500 Barcelona, Spain
- Fundació Lluita contra la Sida i les Malalties Infeccioses, Servei de Malalties Infecciones Hospital Universitari Germans Trias i Pujol, Badalona, 08916 Barcelona, Spain
| | - Julià Blanco
- Institut de Recerca de la Sida IrsiCaixa, Hospital Universitari Germans Trias i Pujol, Badalona, 08916 Barcelona, Spain
- Universitat de Vic - Universitat Central de Catalunya, Vic, 08500 Barcelona, Spain
| | - Dennis Hartigan-O'Connor
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA 95616
- California National Primate Research Center, University of California, Davis, Davis, CA 95616; and
- Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA 94110
| | - Christian Brander
- Institut de Recerca de la Sida IrsiCaixa, Hospital Universitari Germans Trias i Pujol, Badalona, 08916 Barcelona, Spain
- Universitat de Vic - Universitat Central de Catalunya, Vic, 08500 Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats, 08010 Barcelona, Spain
| | - Marta Ruiz-Riol
- Institut de Recerca de la Sida IrsiCaixa, Hospital Universitari Germans Trias i Pujol, Badalona, 08916 Barcelona, Spain;
| |
Collapse
|
12
|
Li Z, Yuan W, Lin Z. Functional roles in cell signaling of adaptor protein TRADD from a structural perspective. Comput Struct Biotechnol J 2020; 18:2867-2876. [PMID: 33163147 PMCID: PMC7593343 DOI: 10.1016/j.csbj.2020.10.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 12/15/2022] Open
Abstract
TRADD participates in various receptor signaling pathways and plays vital roles in many biological activities, including cell survival and apoptosis, in different cellular contexts. TRADD has two distinct functional domains, a TRAF-binding domain at the N-terminus and a death domain (DD) at the C-terminus. The TRAF binding domain of TRADD folds into an α-β plait topology and is mainly responsible for binding TRAF2, while the TRADD-DD can interact with a variety of DD-containing proteins, including receptors and intracellular signaling molecules. After activation of specific receptors such as TNFR1 and DR3, TRADD can bind to the receptor through DD-DD interaction, creating a membrane-proximal platform for the recruitment of downstream molecules to propagate cellular signals. In this review, we highlight recent advances in the studies of the structural mechanism of TRADD adaptor functions for NF-κB activation and apoptosis induction. We also provide suggestions for future structure research related to TRADD-mediated signaling pathways.
Collapse
Affiliation(s)
- Zhen Li
- School of Life Sciences, Tianjin University, Tianjin 300072, PR China.,Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, Tianjin 300072, PR China
| | - Wensu Yuan
- School of Life Sciences, Tianjin University, Tianjin 300072, PR China.,Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, Tianjin 300072, PR China
| | - Zhi Lin
- School of Life Sciences, Tianjin University, Tianjin 300072, PR China.,Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, Tianjin 300072, PR China.,Department of Physiology, National University of Singapore, 117456, Singapore.,Life Sciences Institute, National University of Singapore, 117456, Singapore
| |
Collapse
|
13
|
Bai X, Wang Y, Hu B, Cao Q, Xing M, Song S, Ji A. Fucoidan Induces Apoptosis of HT-29 Cells via the Activation of DR4 and Mitochondrial Pathway. Mar Drugs 2020; 18:E220. [PMID: 32326052 PMCID: PMC7231298 DOI: 10.3390/md18040220] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/04/2020] [Accepted: 04/15/2020] [Indexed: 02/07/2023] Open
Abstract
Fucoidan has a variety of pharmacological activities, but the understanding of the mechanism of fucoidan-induced apoptosis of colorectal cancer cells remains limited. The results of the present study demonstrated that the JNK signaling pathway is involved in the activation of apoptosis in colorectal cancer-derived HT-29 cells, and fucoidan induces apoptosis by activation of the DR4 at the transcriptional and protein levels. The survival rate of HT-29 cells was approximately 40% in the presence of 800 μg/mL of fucoidan, but was increased to 70% after DR4 was silenced by siRNA. Additionally, fucoidan has been shown to reduce the mitochondrial membrane potential and destroy the integrity of mitochondrial membrane. In the presence of an inhibitor of cytochrome C inhibitor and DR4 siRNA or the presence of cytochrome C inhibitor only, the cell survival rate was significantly higher than when cells were treated with DR4 siRNA only. These data indicate that both the DR4 and the mitochondrial pathways contribute to fucoidan-induced apoptosis of HT-29 cells, and the extrinsic pathway is upstream of the intrinsic pathway. In conclusion, the current work identified the mechanism of fucoidan-induced apoptosis and provided a novel theoretical basis for the future development of clinical applications of fucoidan as a drug.
Collapse
Affiliation(s)
- Xu Bai
- Marine College, Shandong University, Weihai 264209, China; (X.B.); (Y.W.); (B.H.); (Q.C.); (M.X.)
| | - Yu Wang
- Marine College, Shandong University, Weihai 264209, China; (X.B.); (Y.W.); (B.H.); (Q.C.); (M.X.)
| | - Bo Hu
- Marine College, Shandong University, Weihai 264209, China; (X.B.); (Y.W.); (B.H.); (Q.C.); (M.X.)
| | - Qi Cao
- Marine College, Shandong University, Weihai 264209, China; (X.B.); (Y.W.); (B.H.); (Q.C.); (M.X.)
| | - Maochen Xing
- Marine College, Shandong University, Weihai 264209, China; (X.B.); (Y.W.); (B.H.); (Q.C.); (M.X.)
| | - Shuliang Song
- Marine College, Shandong University, Weihai 264209, China; (X.B.); (Y.W.); (B.H.); (Q.C.); (M.X.)
| | - Aiguo Ji
- Marine College, Shandong University, Weihai 264209, China; (X.B.); (Y.W.); (B.H.); (Q.C.); (M.X.)
- School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| |
Collapse
|
14
|
Heib M, Rose-John S, Adam D. Necroptosis, ADAM proteases and intestinal (dys)function. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 353:83-152. [PMID: 32381179 DOI: 10.1016/bs.ircmb.2020.02.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Recently, an unexpected connection between necroptosis and members of the a disintegrin and metalloproteinase (ADAM) protease family has been reported. Necroptosis represents an important cell death routine which helps to protect from viral, bacterial, fungal and parasitic infections, maintains adult T cell homeostasis and contributes to the elimination of potentially defective organisms before parturition. Equally important for organismal homeostasis, ADAM proteases control cellular processes such as development and differentiation, immune responses or tissue regeneration. Notably, necroptosis as well as ADAM proteases have been implicated in the control of inflammatory responses in the intestine. In this review, we therefore provide an overview of the physiology and pathophysiology of necroptosis, ADAM proteases and intestinal (dys)function, discuss the contribution of necroptosis and ADAMs to intestinal (dys)function, and review the current knowledge on the role of ADAMs in necroptotic signaling.
Collapse
Affiliation(s)
- Michelle Heib
- Institut für Immunologie, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Stefan Rose-John
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Dieter Adam
- Institut für Immunologie, Christian-Albrechts-Universität zu Kiel, Kiel, Germany.
| |
Collapse
|
15
|
Tougaard P, Martinsen LO, Lützhøft DO, Jensen HE, Flethøj M, Vandenabeele P, Pedersen AE, Skov S, Hansen AK, Hansen CHF. TL1A regulates adipose-resident innate lymphoid immune responses and enables diet-induced obesity in mice. Int J Obes (Lond) 2020; 44:1062-1074. [PMID: 32001795 DOI: 10.1038/s41366-020-0539-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 01/07/2020] [Accepted: 01/16/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND/OBJECTIVES TL1A is a pro-inflammatory cytokine that is homologous to TNFα and connected with the development of several chronic inflammatory disorders. The preliminary results of this study indicated reduced fat accumulation in 9-month-old TL1A-deficient mice at steady state. Thus, the objective was to investigate whether TL1A-deficient mice are resistant to the development of high-fat (HF) diet-induced obesity and to investigate the impact on lymphocyte infiltration in adipose tissue. METHODS TL1A-deficient and TL1A-sufficient male BALB/cJ littermate mice were fed a 60% HF diet or a 10% low-fat control diet for 22 weeks. Mouse body composition and weight were monitored, and tissues were processed and evaluated by flow cytometry, qPCR, and histology. RESULTS In this study, the TL1A-deficient HF-diet-fed mice had reduced whole-body weight gain, which was directly explained by a corresponding fat mass reduction (average 37.2%), compared with that of their TL1A-sufficient littermates. Despite previous data showing marked changes in the gut microbial community, TL1A-deficient GF mice also displayed reduced adiposity. Furthermore, the TL1A-deficient mice were resistant to hepatic steatosis and were shown to have improved glucose tolerance, as determined by oral glucose tolerance test (OGTT), and greater insulin sensitivity. In the epididymal white adipose tissue (eWAT), TL1A deficiency in HF-diet-fed mice resulted in a reduced abundance of IL-18Ra+ type-1 ILCs and γδT cells as well as markedly reduced expression of the mitochondria-regulating genes Ucp1, Ucp2, Ucp3, and Prdm16. Finally, to investigate the link of TL1A to obesity in humans, we identified a noncoding polymorphism (rs4979453) close to the TL1A locus that is associated with waist circumference in men (p = 0.00096, n = 60586). CONCLUSIONS These findings indicate that TL1A plays an important role in regulating adipose tissue mass and that this role is independent of the gut microbiota. Furthermore, we show that TL1A regulates adipose-resident innate lymphocytes and mitochondria-mediated oxidative stress in eWAT.
Collapse
Affiliation(s)
- Peter Tougaard
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, København, Denmark. .,Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, København, Denmark. .,Molecular Signaling and Cell Death Unit, VIB-Ugent Center for Inflammation Research, Flanders Institute for Biotechnology, Ghent, Belgium. .,Department for Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| | - Louise Otterstrøm Martinsen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, København, Denmark
| | - Ditte Olsen Lützhøft
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, København, Denmark
| | - Henrik Elvang Jensen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, København, Denmark
| | - Mette Flethøj
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, København, Denmark
| | - Peter Vandenabeele
- Molecular Signaling and Cell Death Unit, VIB-Ugent Center for Inflammation Research, Flanders Institute for Biotechnology, Ghent, Belgium.,Department for Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Anders Elm Pedersen
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, København, Denmark.,Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, København, Denmark
| | - Søren Skov
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, København, Denmark
| | - Axel Kornerup Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, København, Denmark
| | - Camilla Hartmann Friis Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, København, Denmark
| |
Collapse
|
16
|
Cheng X, Jiang X, Song Y, Gao J, Xue Y, Hassan Z, Gao Q, Zou J. Identification and modulation of expression of a TNF receptor superfamily member 25 homologue in grass carp (Ctenopharyngodon idella). AQUACULTURE AND FISHERIES 2020. [DOI: 10.1016/j.aaf.2019.10.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
17
|
Xi J, Wang L, Yan C, Song J, Song Y, Chen J, Zhu Y, Chen Z, Jin C, Ding J, Zhao C. The Cancer Genome Atlas dataset-based analysis of aberrantly expressed genes by GeneAnalytics in thymoma associated myasthenia gravis: focusing on T cells. J Thorac Dis 2019; 11:2315-2323. [PMID: 31372268 DOI: 10.21037/jtd.2019.06.01] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Background Myasthenia gravis (MG) is a group of autoimmune disease which could be accompanied by thymoma. Many differences have been observed between thymoma-associated MG (TAMG) and non-MG thymoma (NMG). However, the molecular difference between them remained unknown. This study aimed to explore the differentially expressed genes (DEGs) between the two categories and to elucidate the possible pathogenesis of TAMG further. Methods DEGs were calculated using the RNA-Sequencing data from 11 TAMG and 10 NMG in The Cancer Genome Atlas (TCGA) database. GeneAnalytics was performed to characterize the associations between DEGs and tissues and cells, diseases, gene ontology (GO) terms, pathways, phenotypes, and drug/compounds, respectively. Genes related to T cells were sorted out using LifeMapDiscovery Cells and Tissues Database. Genes directly related to the phenotype of autoimmune diseases that were identified by VarElect were validated by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Results The expression level of 169 genes showed a significant difference between the two groups, with 94 up-regulated and 75 down-regulated. Overexpression of six genes (ATM, SFTPB, ANKRD55, BTLA, CCR7, TNFRSF25), which are expressed in T cells and directly related to autoimmune disease through VarElect, was identified. The overexpression of soluble BTLA (sBTLA) (P=0.027), CCR7 (P=0.0018), TNFRSF25 (P=0.0013) and ANKRD55 (P=0.0026) was validated by RT-qPCR in thymoma tissues from our center. Conclusions Overexpression of sBTLA, CCR7, TNFRSF25 and ANKRD55 was identified and validated by RT-qPCR, which could partly explain the underlying pathogenesis in TAMG.
Collapse
Affiliation(s)
- Jianying Xi
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Liang Wang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Chong Yan
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jie Song
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yang Song
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ji Chen
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yongjun Zhu
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Zhiming Chen
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Chun Jin
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Jianyong Ding
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Chongbo Zhao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China.,Department of Neurology, Jing'an District Centre Hospital of Shanghai, Fudan University, Shanghai 200040, China
| |
Collapse
|
18
|
Masvekar R, Wu T, Kosa P, Barbour C, Fossati V, Bielekova B. Cerebrospinal fluid biomarkers link toxic astrogliosis and microglial activation to multiple sclerosis severity. Mult Scler Relat Disord 2019; 28:34-43. [PMID: 30553167 PMCID: PMC6411304 DOI: 10.1016/j.msard.2018.11.032] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/20/2018] [Accepted: 11/29/2018] [Indexed: 11/26/2022]
Abstract
BACKGROUND Once multiple sclerosis (MS) reaches the progressive stage, immunomodulatory treatments have limited efficacy. This suggests that processes other than activation of innate immunity may at least partially underlie disability progression during late stages of MS. Pathology identified these alternative processes as aberrant activation of astrocytes and microglia, and subsequent degeneration of oligodendrocytes and neurons. However, we mostly lack biomarkers that could measure central nervous system (CNS) cell-specific intrathecal processes in living subjects. This prevents differentiating pathogenic processes from an epiphenomenon. Therefore, we sought to develop biomarkers of CNS cell-specific processes and link them to disability progression in MS. METHODS In a blinded manner, we measured over 1000 proteins in the cerebrospinal fluid (CSF) of 431 patients with neuroimmunological diseases and healthy volunteers using modified DNA-aptamers (SOMAscan®). We defined CNS cell type-enriched clusters using variable cluster analysis, combined with in vitro modeling. Differences between diagnostic categories were identified in the training cohort (n = 217) and their correlation to disability measures were assessed; results were validated in an independent validation cohort (n = 214). RESULTS Astrocyte cluster 8 (MMP7, SERPINA3, GZMA and CLIC1) and microglial cluster 2 (DSG2 and TNFRSF25) were reproducibly elevated in MS and had a significant and reproducible correlation with MS severity suggesting their pathogenic role. In vitro studies demonstrated that proteins of astrocyte cluster 8 are noticeably released upon stimulation with proinflammatory stimuli and overlap with the phenotype of recently described neuro-toxic (A1) astrocytes. CONCLUSION Microglial activation and toxic astrogliosis are associated with MS disease process and may partake in CNS tissue destruction. This hypothesis should be tested in new clinical trials.
Collapse
Affiliation(s)
- Ruturaj Masvekar
- Neuroimmunological Diseases Section (NDS), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Building 10, Room 5N248, 10 Center Drive, MSC1444, Bethesda, MD 20892, United States
| | - Tianxia Wu
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Peter Kosa
- Neuroimmunological Diseases Section (NDS), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Building 10, Room 5N248, 10 Center Drive, MSC1444, Bethesda, MD 20892, United States
| | - Christopher Barbour
- Neuroimmunological Diseases Section (NDS), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Building 10, Room 5N248, 10 Center Drive, MSC1444, Bethesda, MD 20892, United States; Department of Mathematical Sciences, Montana State University, Bozeman, MT, United States
| | - Valentina Fossati
- The New York Stem Cell Foundation Research Institute, New York, NY, United States
| | - Bibiana Bielekova
- Neuroimmunological Diseases Section (NDS), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Building 10, Room 5N248, 10 Center Drive, MSC1444, Bethesda, MD 20892, United States.
| |
Collapse
|
19
|
Zhou H, Wang J, Zhu P, Hu S, Ren J. Ripk3 regulates cardiac microvascular reperfusion injury: The role of IP3R-dependent calcium overload, XO-mediated oxidative stress and F-action/filopodia-based cellular migration. Cell Signal 2018; 45:12-22. [PMID: 29413844 DOI: 10.1016/j.cellsig.2018.01.020] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 01/13/2018] [Accepted: 01/21/2018] [Indexed: 01/09/2023]
Abstract
Ripk3-mediated cellular apoptosis is a major contributor to the pathogenesis of myocardial ischemia reperfusion (IR) injury. However, the mechanisms by which Ripk3 influences microvascular homeostasis and endothelial apoptosis are not completely understood. In this study, loss of Ripk3 inhibited endothelial apoptosis, alleviated luminal swelling, maintained microvasculature patency, reduced the expression of adhesion molecules and limited the myocardial inflammatory response. In vitro, Ripk3 deficiency protected endothelial cells from apoptosis and migratory arrest induced by HR injury. Mechanistically, Ripk3 had the ability to migrate onto the endoplasmic reticulum (ER), leading to ER damage, as evidenced by increased IP3R and XO expression. The higher IP3R content was associated with cellular calcium overload, and increased XO expression was involved in cellular oxidative injury. Furthermore, IP3R-mediated calcium overload and XO-dependent oxidative damage were able to initiate cellular apoptosis. More importantly, IP3R and XO also caused F-actin degradation into G-actin via post-transcriptional modification of cofilin, impairing the formation of the filopodia and limiting the migratory response of endothelial cells. Altogether, our data confirmed that Ripk3 was involved in microvascular IR injury via regulation of IP3R-mediated calcium overload, XO-dependent oxidative damage and filopodia-related cellular migration, ultimately leading to endothelial apoptosis and migratory inhibition. These findings provide a potential target for treating cardiac microcirculatory IR injury.
Collapse
Affiliation(s)
- Hao Zhou
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China; Center for Cardiovascular Research and Alternative Medicine, Wyoming University, Laramie, WY 82071, USA.
| | - Jin Wang
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China
| | - Pingjun Zhu
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China
| | - Shunying Hu
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China
| | - Jun Ren
- Center for Cardiovascular Research and Alternative Medicine, Wyoming University, Laramie, WY 82071, USA
| |
Collapse
|
20
|
Kondylis V, Kumari S, Vlantis K, Pasparakis M. The interplay of IKK, NF-κB and RIPK1 signaling in the regulation of cell death, tissue homeostasis and inflammation. Immunol Rev 2018; 277:113-127. [PMID: 28462531 DOI: 10.1111/imr.12550] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 03/07/2017] [Indexed: 12/12/2022]
Abstract
Regulated cell death pathways have important functions in host defense and tissue homeostasis. Studies in genetic mouse models provided evidence that cell death could cause inflammation in different tissues. Inhibition of RIPK3-MLKL-dependent necroptosis by FADD and caspase-8 was identified as a key mechanism preventing inflammation in epithelial barriers. Moreover, the interplay between IKK/NF-κB and RIPK1 signaling was recognized as a critical determinant of tissue homeostasis and inflammation. NEMO was shown to regulate RIPK1 kinase activity-mediated apoptosis by NF-κB-dependent and -independent functions, which are critical for averting chronic tissue injury and inflammation in the intestine and the liver. In addition, RIPK1 was shown to exhibit kinase activity-independent functions that are essential for preventing cell death, maintaining tissue architecture and inhibiting inflammation. In the intestine, RIPK1 acts as a scaffold to prevent epithelial cell apoptosis and preserve tissue integrity. In the skin, RIPK1 functions via its RHIM to counteract ZBP1/DAI-dependent activation of RIPK3-MLKL-dependent necroptosis and inflammation. Collectively, these studies provided evidence that the regulation of cell death signaling plays an important role in the maintenance of tissue homeostasis, and suggested that cell death could be causally involved in the pathogenesis of inflammatory diseases.
Collapse
Affiliation(s)
- Vangelis Kondylis
- Institute for Genetics, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Snehlata Kumari
- Institute for Genetics, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Katerina Vlantis
- Institute for Genetics, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Manolis Pasparakis
- Institute for Genetics, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| |
Collapse
|
21
|
Fuchslocher Chico J, Saggau C, Adam D. Proteolytic control of regulated necrosis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:2147-2161. [DOI: 10.1016/j.bbamcr.2017.05.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 05/27/2017] [Accepted: 05/30/2017] [Indexed: 12/20/2022]
|
22
|
Bittner S, Ehrenschwender M. Multifaceted death receptor 3 signaling-promoting survival and triggering death. FEBS Lett 2017; 591:2543-2555. [DOI: 10.1002/1873-3468.12747] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 06/24/2017] [Accepted: 07/03/2017] [Indexed: 02/06/2023]
Affiliation(s)
- Sebastian Bittner
- Institute of Clinical Microbiology and Hygiene; University Hospital Regensburg; Germany
| | - Martin Ehrenschwender
- Institute of Clinical Microbiology and Hygiene; University Hospital Regensburg; Germany
| |
Collapse
|