1
|
Fu J, Wang H, Chen Y, Zhang C, Zou Y. The Multifaceted Ubiquitination of BIK1 During Plant Immunity in Arabidopsis thaliana. Int J Mol Sci 2024; 25:12187. [PMID: 39596247 PMCID: PMC11594851 DOI: 10.3390/ijms252212187] [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: 10/20/2024] [Revised: 11/10/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
As sessile organisms, the plant immune system plays a vital role in protecting plants from the widespread pathogens in the environment. The Arabidopsis thaliana (Arabidopsis) receptor-like cytoplasmic kinase BOTRYTIS-INDUCED KINASE1 (BIK1) acts as a central regulator during plant immunity. As such, not only the BIK1 protein accumulation but also the attenuation is tightly regulated to ensure effective immune responses. Recent studies have highlighted the critical roles of ubiquitination in maintaining BIK1 homeostasis. Here, we review the latest advances in the ubiquitination of BIK1 in plant immunity, which is mediated by ubiquitin ligases PUB25/26, RHA3A/B, RGLG1/2, and PUB4. Additionally, we summarize and discuss the sites and types of BIK1 ubiquitination. Collectively, these analyses not only illustrate that the differential modifications on BIK1 by multiple ubiquitin ligases hold a crucial position in plant immunity but also provide a good example for future studies on ubiquitin-mediated modifications in plants.
Collapse
Affiliation(s)
| | | | | | | | - Yanmin Zou
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaboration Innovation Center for Cell Signaling and Environmental Adaptation, Hebei Key Laboratory of Molecular and Cellular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, China
| |
Collapse
|
2
|
Cui L, Yang L, Lai B, Luo L, Deng H, Chen Z, Wang Z. Integrative and comprehensive pan-cancer analysis of ubiquitin specific peptidase 11 ( USP11) as a prognostic and immunological biomarker. Heliyon 2024; 10:e34523. [PMID: 39114046 PMCID: PMC11305246 DOI: 10.1016/j.heliyon.2024.e34523] [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: 02/03/2024] [Revised: 07/02/2024] [Accepted: 07/10/2024] [Indexed: 08/10/2024] Open
Abstract
The significance of USP11 as a critical regulator in cancer has garnered substantial attention, primarily due to its catalytic activity as a deubiquitinating enzyme. Nonetheless, a thorough evaluation of USP11 across various cancer types in pan-cancer studies remains absent. Our analysis integrates data from a variety of sources, including five immunotherapy cohorts, thirty-three cohorts from The Cancer Genome Atlas (TCGA), and sixteen cohorts from the Gene Expression Omnibus (GEO), two of which involve single-cell transcriptomic data. Our findings indicate that aberrant USP11 expression is predictive of survival outcomes across various cancer types. The highest frequency of genomic alterations was observed in uterine corpus endometrial carcinoma (UCEC), with single-cell transcriptome analysis revealing significantly higher USP11 expression in plasmacytoid dendritic cells and mast cells. Notably, USP11 expression was associated with the infiltration levels of CD8+ T cells and natural killer (NK) activated cells. Additionally, in the skin cutaneous melanoma (SKCM) phs000452 cohort, patients with higher USP11 mRNA levels during immunotherapy experienced a significantly shorter median progression-free survival. USP11 emerges as a promising molecular biomarker with significant potential for predicting patient prognosis and immunoreactivity across various cancer types.
Collapse
Affiliation(s)
- Lijuan Cui
- Pathology Department, Suining Central Hospital, Suining, Sichuan, 629000, China
| | - Ling Yang
- Pathology Department, Suining Central Hospital, Suining, Sichuan, 629000, China
| | - Boan Lai
- Pathology Department, Suining Central Hospital, Suining, Sichuan, 629000, China
| | - Lingzhi Luo
- Pathology Department, Suining Central Hospital, Suining, Sichuan, 629000, China
| | - Haoyue Deng
- Pathology Department, Suining Central Hospital, Suining, Sichuan, 629000, China
| | - Zhongyi Chen
- Pathology Department, Suining Central Hospital, Suining, Sichuan, 629000, China
| | - Zixing Wang
- Pathology Department, Suining Central Hospital, Suining, Sichuan, 629000, China
| |
Collapse
|
3
|
Zhang J, Tu H, Zheng Z, Zhao X, Lin X. RNF31 promotes tumorigenesis via inhibiting RIPK1 kinase-dependent apoptosis. Oncogene 2023; 42:1585-1596. [PMID: 36997719 DOI: 10.1038/s41388-023-02669-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 03/06/2023] [Accepted: 03/13/2023] [Indexed: 04/01/2023]
Abstract
It is well established that interferon (IFN) and tumor necrosis factor (TNF) could synergistically promote antitumor toxicity and avoid resistance of antigen-negative tumors during cancer immunotherapy. The linear ubiquitin chain assembly complex (LUBAC) has been widely known to regulate receptor-interacting protein kinase-1(RIPK1) kinase activity and TNF-mediated cell death during inflammation and embryogenesis. However, whether LUBAC and RIPK1 kinase activity in tumor microenvironment could regulate antitumor immunity are still not very clear. Here, we demonstrated a cancer cell-intrinsic role of LUBAC complex in tumor microenvironment to promote tumorigenesis. Lacking LUBAC component RNF31 in B16 melanoma cells but not immune cells including macrophages or dendritic cells greatly impaired tumor growth by increasing intratumoral CD8+ T cells infiltration. Mechanistically, we found that tumor cells without RNF31 shown severe apoptosis-mediated cell death caused by TNFα/IFNγ in the tumor microenvironment. Most importantly, we found that RNF31 could limit RIPK1 kinase activity and further prevent tumor cell death in a transcription-independent manner, suggesting a crucial role of RIPK1 kinase activity in tumorigenesis. Together, our results demonstrate an essential role of RNF31 and RIPK1 kinase activity in tumorigenesis and imply that RNF31 inhibition could be harnessed to enhance antitumor toxicity during tumor immunotherapy.
Collapse
Affiliation(s)
- Jie Zhang
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, Tsinghua University School of Medicine, Beijing, 100084, China
| | - Hailin Tu
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, Tsinghua University School of Medicine, Beijing, 100084, China
| | - Zheyu Zheng
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, Tsinghua University School of Medicine, Beijing, 100084, China
| | - Xueqiang Zhao
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, Tsinghua University School of Medicine, Beijing, 100084, China
| | - Xin Lin
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China.
- Department of Basic Medical Sciences, Tsinghua University School of Medicine, Beijing, 100084, China.
| |
Collapse
|
4
|
Mussbacher M, Derler M, Basílio J, Schmid JA. NF-κB in monocytes and macrophages - an inflammatory master regulator in multitalented immune cells. Front Immunol 2023; 14:1134661. [PMID: 36911661 PMCID: PMC9995663 DOI: 10.3389/fimmu.2023.1134661] [Citation(s) in RCA: 108] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 02/07/2023] [Indexed: 02/25/2023] Open
Abstract
Nuclear factor κB (NF-κB) is a dimeric transcription factor constituted by two of five protein family members. It plays an essential role in inflammation and immunity by regulating the expression of numerous chemokines, cytokines, transcription factors, and regulatory proteins. Since NF-κB is expressed in almost all human cells, it is important to understand its cell type-, tissue-, and stimulus-specific roles as well as its temporal dynamics and disease-specific context. Although NF-κB was discovered more than 35 years ago, many questions are still unanswered, and with the availability of novel technologies such as single-cell sequencing and cell fate-mapping, new fascinating questions arose. In this review, we will summarize current findings on the role of NF-κB in monocytes and macrophages. These innate immune cells show high plasticity and dynamically adjust their effector functions against invading pathogens and environmental cues. Their versatile functions can range from antimicrobial defense and antitumor immune responses to foam cell formation and wound healing. NF-κB is crucial for their activation and balances their phenotypes by finely coordinating transcriptional and epigenomic programs. Thereby, NF-κB is critically involved in inflammasome activation, cytokine release, and cell survival. Macrophage-specific NF-κB activation has far-reaching implications in the development and progression of numerous inflammatory diseases. Moreover, recent findings highlighted the temporal dynamics of myeloid NF-κB activation and underlined the complexity of this inflammatory master regulator. This review will provide an overview of the complex roles of NF-κB in macrophage signal transduction, polarization, inflammasome activation, and cell survival.
Collapse
Affiliation(s)
- Marion Mussbacher
- Department of Pharmacology and Toxicology, Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria
| | - Martina Derler
- Department of Pharmacology and Toxicology, Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria
| | - José Basílio
- Department of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
- INESC ID–Instituto de Engenharia de Sistemas e Computadores, Investigação e Desenvolvimento em Lisboa, Universidade de Lisboa, Lisboa, Portugal
| | - Johannes A. Schmid
- Department of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
5
|
Zinatizadeh MR, Schock B, Chalbatani GM, Zarandi PK, Jalali SA, Miri SR. The Nuclear Factor Kappa B (NF-kB) signaling in cancer development and immune diseases. Genes Dis 2021; 8:287-297. [PMID: 33997176 PMCID: PMC8093649 DOI: 10.1016/j.gendis.2020.06.005] [Citation(s) in RCA: 285] [Impact Index Per Article: 71.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 05/26/2020] [Accepted: 06/12/2020] [Indexed: 02/07/2023] Open
Abstract
The nuclear factor kappa B (NF-kB) family of transcription factors plays an essential role as stressors in the cellular environment, and controls the expression of important regulatory genes such as immunity, inflammation, death, and cell proliferation. NF-kB protein is located in the cytoplasm, and can be activated by various cellular stimuli. There are two pathways for NF-kB activation, as the canonical and non-canonical pathways, which require complex molecular interactions with adapter proteins and phosphorylation and ubiquitinase enzymes. Accordingly, this increases NF-kB translocation in the nucleus and regulates gene expression. In this study, the concepts that emerge in different cellular systems allow the design of NF-kB function in humans. This would not only allow the development for rare diseases associated with NF-kB, but would also be used as a source of useful information to eliminate widespread consequences such as cancer or inflammatory/immune diseases.
Collapse
Affiliation(s)
| | - Bettina Schock
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, BT7 1NN, United Kingdom
| | - Ghanbar Mahmoodi Chalbatani
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, 1336616357, Iran
- Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Science, Tehran, 1336616357, Iran
| | | | - Seyed Amir Jalali
- Department of Medical Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 1336616357, Iran
| | - Seyed Rouhollah Miri
- Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Science, Tehran, 1336616357, Iran
| |
Collapse
|
6
|
Fischer R, Kontermann RE, Pfizenmaier K. Selective Targeting of TNF Receptors as a Novel Therapeutic Approach. Front Cell Dev Biol 2020; 8:401. [PMID: 32528961 PMCID: PMC7264106 DOI: 10.3389/fcell.2020.00401] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 05/01/2020] [Indexed: 12/14/2022] Open
Abstract
Tumor necrosis factor (TNF) is a central regulator of immunity. Due to its dominant pro-inflammatory effects, drugs that neutralize TNF were developed and are clinically used to treat inflammatory and autoimmune diseases, such as rheumatoid arthritis, inflammatory bowel disease and psoriasis. However, despite their clinical success the use of anti-TNF drugs is limited, in part due to unwanted, severe side effects and in some diseases its use even is contraindicative. With gaining knowledge about the signaling mechanisms of TNF and the differential role of the two TNF receptors (TNFR), alternative therapeutic concepts based on receptor selective intervention have led to the development of novel protein therapeutics targeting TNFR1 with antagonists and TNFR2 with agonists. These antibodies and bio-engineered ligands are currently in preclinical and early clinical stages of development. Preclinical data obtained in different disease models show that selective targeting of TNFRs has therapeutic potential and may be superior to global TNF blockade in several disease indications.
Collapse
Affiliation(s)
- Roman Fischer
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Roland E Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Klaus Pfizenmaier
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| |
Collapse
|
7
|
Asmamaw MD, Liu Y, Zheng YC, Shi XJ, Liu HM. Skp2 in the ubiquitin-proteasome system: A comprehensive review. Med Res Rev 2020; 40:1920-1949. [PMID: 32391596 DOI: 10.1002/med.21675] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 03/26/2020] [Accepted: 04/27/2020] [Indexed: 12/19/2022]
Abstract
The ubiquitin-proteasome system (UPS) is a complex process that regulates protein stability and activity by the sequential actions of E1, E2 and E3 enzymes to influence diverse aspects of eukaryotic cells. However, due to the diversity of proteins in cells, substrate selection is a highly critical part of the process. As a key player in UPS, E3 ubiquitin ligases recruit substrates for ubiquitination specifically. Among them, RING E3 ubiquitin ligases which are the most abundant E3 ubiquitin ligases contribute to diverse cellular processes. The multisubunit cullin-RING ligases (CRLs) are the largest family of RING E3 ubiquitin ligases with tremendous plasticity in substrate specificity and regulate a vast array of cellular functions. The F-box protein Skp2 is a component of CRL1 (the prototype of CRLs) which is expressed in many tissues and participates in multiple cellular functions such as cell proliferation, metabolism, and tumorigenesis by contributing to the ubiquitination and subsequent degradation of several specific tumor suppressors. Most importantly, Skp2 plays a pivotal role in a plethora of cancer-associated signaling pathways. It enhances cell growth, accelerates cell cycle progression, promotes migration and invasion, and inhibits cell apoptosis among others. Hence, targeting Skp2 may represent a novel and attractive strategy for the treatment of different human cancers overexpressing this oncogene. In this review article, we summarized the known roles of Skp2 both in health and disease states in relation to the UPS.
Collapse
Affiliation(s)
- Moges Dessale Asmamaw
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Henan Key Laboratory of Drug Quality Control & Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Ministry of Education of China, Zhengzhou, Henan, China
| | - Ying Liu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Henan Key Laboratory of Drug Quality Control & Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Ministry of Education of China, Zhengzhou, Henan, China
| | - Yi-Chao Zheng
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Henan Key Laboratory of Drug Quality Control & Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Ministry of Education of China, Zhengzhou, Henan, China
| | - Xiao-Jing Shi
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Henan Key Laboratory of Drug Quality Control & Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Ministry of Education of China, Zhengzhou, Henan, China
| | - Hong-Min Liu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Henan Key Laboratory of Drug Quality Control & Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Ministry of Education of China, Zhengzhou, Henan, China
| |
Collapse
|
8
|
Iskender H, Yenice G, Dokumacioglu E, Hayirli A, Sevim C, Dokumacioglu A, Terim Kapakin KA. Astaxanthin alleviates renal damage of rats on high fructose diet through modulating NFκB/SIRT1 pathway and mitigating oxidative stress. Arch Physiol Biochem 2020; 126:89-93. [PMID: 30081678 DOI: 10.1080/13813455.2018.1493609] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
This study was conducted to determine the effect of astaxanthin (ASX) treatment on alleviation of renal damage in high fructose induced nephrotoxicity in rats. Treatments were arranged in a 2 × 2 factorial fashion: administrations of fructose (30%, via drinking water) and ASX (1 mg/kg/day, within 0.2 ml olive oil) for 8 weeks. Data were analyzed by two-way ANOVA. The ASX treatment decreased serum urea (p < .01) and blood urea-N concentrations (p < .02) at a lower extent in rats receiving fructose than those not receiving fructose. Moreover, the ASX treatment reversed the increases in malondialdehyde (MDA) (p < .0001) and nuclear factor kappa B (NF-κB) (p < .0003) levels and the decreases in superoxide dismutase (SOD) activity (p < .0001) and sirtuin-1 (SIRT1) level (p < .0004), in the kidney upon high fructose consumption. The data suggest that ASX supplementation alleviates renal damage induced by high fructose consumption through modulating NF-κB/SIRT1 pathway and mitigating oxidative stress.
Collapse
Affiliation(s)
- Hatice Iskender
- Department of Nutrition and Dietetics, Faculty of Healthy Sciences, Artvin Coruh University, Artvin, Turkey
| | - Guler Yenice
- Department of Animal Nutrition and Nutritional Disorders, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
| | - Eda Dokumacioglu
- Department of Nutrition and Dietetics, Faculty of Healthy Sciences, Artvin Coruh University, Artvin, Turkey
| | - Armagan Hayirli
- Department of Animal Nutrition and Nutritional Disorders, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
| | - Cigdem Sevim
- Department of Pharmacology, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
| | - Ali Dokumacioglu
- Department of Medical Biochemistry, Hopa Government Hospital, Artvin, Turkey
| | | |
Collapse
|
9
|
Gâtel P, Piechaczyk M, Bossis G. Ubiquitin, SUMO, and Nedd8 as Therapeutic Targets in Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1233:29-54. [PMID: 32274752 DOI: 10.1007/978-3-030-38266-7_2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Ubiquitin defines a family of approximately 20 peptidic posttranslational modifiers collectively called the Ubiquitin-like (UbLs). They are conjugated to thousands of proteins, modifying their function and fate in many ways. Dysregulation of these modifications has been implicated in a variety of pathologies, in particular cancer. Ubiquitin, SUMO (-1 to -3), and Nedd8 are the best-characterized UbLs. They have been involved in the regulation of the activity and/or the stability of diverse components of various oncogenic or tumor suppressor pathways. Moreover, the dysregulation of enzymes responsible for their conjugation/deconjugation has also been associated with tumorigenesis and cancer resistance to therapies. The UbL system therefore constitutes an attractive target for developing novel anticancer therapeutic strategies. Here, we review the roles and dysregulations of Ubiquitin, SUMO, and Nedd8 pathways in tumorigenesis, as well as recent advances in the identification of small molecules targeting their conjugating machineries for potential application in the fight against cancer.
Collapse
Affiliation(s)
- Pierre Gâtel
- Equipe Labellisée Ligue Contre le Cancer, IGMM, Univ Montpellier, CNRS, Montpellier, France
| | - Marc Piechaczyk
- Equipe Labellisée Ligue Contre le Cancer, IGMM, Univ Montpellier, CNRS, Montpellier, France
| | - Guillaume Bossis
- Equipe Labellisée Ligue Contre le Cancer, IGMM, Univ Montpellier, CNRS, Montpellier, France.
| |
Collapse
|
10
|
Tian L, Wang K, Liu H, Li K, Lin B, Fang Z, Han J, Li N, Yang H, Bian L, Liu X, Xi Z. UCH-L1 mitigates neurotoxicity induced by ZnO particles via stabilizing the inhibitor of NF-kappa B signaling, IκB-α. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 180:259-268. [PMID: 31096129 DOI: 10.1016/j.ecoenv.2019.03.058] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 03/07/2019] [Accepted: 03/14/2019] [Indexed: 06/09/2023]
Abstract
Our study determined the toxic effects of zinc oxide (ZnO) particles with different diameters on dopaminergic (DA) neurons, the role of ubiquitin C-terminal hydrolase L1 (UCH-L1) for ZnO particles-induced neurotoxicity, and corresponding molecular mechanisms. We constructed an in vitro cell injury model for DA neurons to analyze the cytotoxicity of ZnO particles using SH-SY5Y cells. Following cell viability assays and flow cytometry, we found that the cytotoxicity of ZnO particles was affected by particle size, time, and dose of exposure. For example, the toxicity of ZnO particles with 50 nm or 100 nm diameter was stronger than that of ZnO particles with 1000 nm diameter. Furthermore, ZnO particles exposure resulted in a significant decrease in UCH-L1 expression in SH-SY5Y; whereas UCH-L1 overexpression led to a significant increase in cell viability and a sharp decrease in ROS level. Western blotting and adenovirus transfection found that exposure to ZnO particles with different diameters all activate the NF-κB signaling in SH-SY5Y cells; whereas UCH-L1 over-expression resulted in increased levels of IκBα, an endogenous inhibitor of NF-κB signaling pathway. ZnO particles with different diameters all induced cytotoxicity in DA neurons, which may be related to the free Zn2+ in the suspension. Regarding the neurotoxic effect of ZnO particles, UCH-L1 protects against and/or alleviates neuronal damage, possibly by deubiquitination of the endogenous inhibitor, IκBα, which leads to activation of NF-κB signaling. Therefore, one possible mechanism for ZnO particle-induced neurotoxicity may be mediated via the down-regulation of UCH-L1 expression in DA cells.
Collapse
Affiliation(s)
- Lei Tian
- Tianjin Institute of Environmental and Operational Medicine, No. 1 Dali Road, Heping District, Tianjin, 300050, China.
| | - Kun Wang
- Tianjin Institute of Environmental and Operational Medicine, No. 1 Dali Road, Heping District, Tianjin, 300050, China.
| | - Huanliang Liu
- Tianjin Institute of Environmental and Operational Medicine, No. 1 Dali Road, Heping District, Tianjin, 300050, China.
| | - Kang Li
- Tianjin Institute of Environmental and Operational Medicine, No. 1 Dali Road, Heping District, Tianjin, 300050, China.
| | - Bencheng Lin
- Tianjin Institute of Environmental and Operational Medicine, No. 1 Dali Road, Heping District, Tianjin, 300050, China.
| | - Zhen Fang
- Tianjin Institute of Environmental and Operational Medicine, No. 1 Dali Road, Heping District, Tianjin, 300050, China; Binzhou Medical College, Yantai 264000, China.
| | - Jie Han
- Tianjin Institute of Environmental and Operational Medicine, No. 1 Dali Road, Heping District, Tianjin, 300050, China; Tianjin University of Sport, No.16 Donghai Road, Jinghai District, Tianjin, 301617, China.
| | - Ning Li
- Tianjin Institute of Environmental and Operational Medicine, No. 1 Dali Road, Heping District, Tianjin, 300050, China; Binzhou Medical College, Yantai 264000, China.
| | - Hu Yang
- Tianjin Institute of Environmental and Operational Medicine, No. 1 Dali Road, Heping District, Tianjin, 300050, China; The Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, China.
| | - Liping Bian
- Tianjin Institute of Environmental and Operational Medicine, No. 1 Dali Road, Heping District, Tianjin, 300050, China.
| | - Xiaohua Liu
- Tianjin Institute of Environmental and Operational Medicine, No. 1 Dali Road, Heping District, Tianjin, 300050, China; Tianjin University of Sport, No.16 Donghai Road, Jinghai District, Tianjin, 301617, China.
| | - Zhuge Xi
- Tianjin Institute of Environmental and Operational Medicine, No. 1 Dali Road, Heping District, Tianjin, 300050, China.
| |
Collapse
|
11
|
Seleznik GM, Reding T, Peter L, Gupta A, Steiner SG, Sonda S, Verbeke CS, Dejardin E, Khatkov I, Segerer S, Heikenwalder M, Graf R. Development of autoimmune pancreatitis is independent of CDKN1A/p21-mediated pancreatic inflammation. Gut 2018; 67:1663-1673. [PMID: 28774888 DOI: 10.1136/gutjnl-2016-313458] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 06/07/2017] [Accepted: 06/19/2017] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Chronic pancreatitis (CP) and autoimmune pancreatitis (AIP) are characterised by different inflammatory processes. If pancreatic inflammation is a prerequisite for autoimmunity is still unclear. AIP is considered mostly a T cell-mediated disease; however, in induction of CP, macrophages play a pivotal role. p21-a member of cyclin-dependent kinase inhibitors-can influence inflammatory processes, in particular can regulate T cell activation and promote macrophage development. We therefore examined the role of p21-mediated inflammation in AIP. DESIGN We intercrossed lymphotoxin (LT) overexpressing mice (Tg(Ela1-LTa,b))-a model to study AIP development-with p21-deficient mice. Furthermore, we characterised p21 expression in human AIP and non-AIP specimens. RESULTS p21 deficiency in LT mice (LTp21-/-) prevented early pancreatic injury and reduced inflammation. In acinar cells, diminished proliferation and abrogated activation of non-canonical nuclear factor kappa-light-chain-enhancer of activated B cell (NF-κB) pathway was observed. In contrast, 12-month-old LT mice with and without p21 had similar inflammatory signatures and T-B cell infiltration. Interestingly, LT and LTp21-/- mice had comparable tertiary lymphoid organs (TLOs), autoantibodies and elevated IgG levels. However, acinar cell proliferation, acinar-to-ductal metaplasia and acinar non-canonical NF-κB pathway activation remained impaired in LTp21-/- pancreata. CONCLUSIONS Our findings indicate that p21 is crucial for pancreatic inflammation in LT-driven pancreatic injury. p21 is involved in early acinar secretion of inflammatory mediators that attract innate immune cells. However, p21 is not essential for humoral immune response, accountable for autoimmunity. Remarkably, p21 renders acinar cells less susceptible to proliferation and transdifferentiation. We therefore suggest that AIP can also develop independent of chronic inflammatory processes.
Collapse
Affiliation(s)
- Gitta M Seleznik
- Department of Visceral and Transplantation Surgery, Swiss HPB Centre, University Hospital Zurich, Zurich, Switzerland
| | - Theresia Reding
- Department of Visceral and Transplantation Surgery, Swiss HPB Centre, University Hospital Zurich, Zurich, Switzerland
| | - Lukas Peter
- Department of Visceral and Transplantation Surgery, Swiss HPB Centre, University Hospital Zurich, Zurich, Switzerland
| | - Anurag Gupta
- Department of Visceral and Transplantation Surgery, Swiss HPB Centre, University Hospital Zurich, Zurich, Switzerland
| | - Sabrina G Steiner
- Department of Visceral and Transplantation Surgery, Swiss HPB Centre, University Hospital Zurich, Zurich, Switzerland
| | - Sabrina Sonda
- Department of Visceral and Transplantation Surgery, Swiss HPB Centre, University Hospital Zurich, Zurich, Switzerland
| | - Caroline S Verbeke
- Department of Pathology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Emmanuel Dejardin
- Laboratory of Molecular Immunology and Signal Transduction, GIGA-Research, University of Liège, Liège, Belgium
| | - Igor Khatkov
- Department of Medicine and Dentistry, Moscow State University, Moscow, Russia
| | - Stephan Segerer
- Division of Nephrology, University Hospital, Zurich, Switzerland.,Division of Nephrology, Dialysis and Transplantation, Kantonsspital Aarau, Aarau, Switzerland
| | - Mathias Heikenwalder
- School of Medicine, Institute of Virology, TUM-Helmholtz Zentrum Munich, Munich, Germany.,Department of Chronic Inflammation and Cancer, German Cancer Center (DKFZ), Heidelberg, Germany
| | - Rolf Graf
- Department of Visceral and Transplantation Surgery, Swiss HPB Centre, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
12
|
Aalto AL, Mohan AK, Schwintzer L, Kupka S, Kietz C, Walczak H, Broemer M, Meinander A. M1-linked ubiquitination by LUBEL is required for inflammatory responses to oral infection in Drosophila. Cell Death Differ 2018; 26:860-876. [PMID: 30026495 PMCID: PMC6462001 DOI: 10.1038/s41418-018-0164-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 06/20/2018] [Accepted: 07/02/2018] [Indexed: 01/04/2023] Open
Abstract
Post-translational modifications such as ubiquitination play a key role in regulation of inflammatory nuclear factor-κB (NF-κB) signalling. The Drosophila IκB kinase γ (IKKγ) Kenny is a central regulator of the Drosophila Imd pathway responsible for activation of the NF-κB Relish. We found the Drosophila E3 ligase and HOIL-1L interacting protein (HOIP) orthologue linear ubiquitin E3 ligase (LUBEL) to catalyse formation of M1-linked linear ubiquitin (M1-Ub) chains in flies in a signal-dependent manner upon bacterial infection. Upon activation of the Imd pathway, LUBEL modifies Kenny with M1-Ub chains. Interestingly, the LUBEL-mediated M1-Ub chains seem to be targeted both directly to Kenny and to K63-linked ubiquitin chains conjugated to Kenny by DIAP2. This suggests that DIAP2 and LUBEL work together to promote Kenny-mediated activation of Relish. We found LUBEL-mediated M1-Ub chain formation to be required for flies to survive oral infection with Gram-negative bacteria, for activation of Relish-mediated expression of antimicrobial peptide genes and for pathogen clearance during oral infection. Interestingly, LUBEL is not required for mounting an immune response against systemic infection, as Relish-mediated antimicrobial peptide genes can be expressed in the absence of LUBEL during septic injury. Finally, transgenic induction of LUBEL-mediated M1-Ub drives expression of antimicrobial peptide genes and hyperplasia in the midgut in the absence of infection. This suggests that M1-Ub chains are important for Imd signalling and immune responses in the intestinal epithelia, and that enhanced M1-Ub chain formation is able to drive chronic intestinal inflammation in flies.
Collapse
Affiliation(s)
- Anna L Aalto
- Department of Cell Biology, Faculty of Science and Engineering, BioCity, Åbo Akademi University, 20520, Turku, Finland
| | - Aravind K Mohan
- Department of Cell Biology, Faculty of Science and Engineering, BioCity, Åbo Akademi University, 20520, Turku, Finland
| | - Lukas Schwintzer
- German Center for Neurodegenerative Diseases (DZNE), 53127, Bonn, Germany
| | - Sebastian Kupka
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, London, WC1E 6BT, UK
| | - Christa Kietz
- Department of Cell Biology, Faculty of Science and Engineering, BioCity, Åbo Akademi University, 20520, Turku, Finland
| | - Henning Walczak
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, London, WC1E 6BT, UK
| | - Meike Broemer
- German Center for Neurodegenerative Diseases (DZNE), 53127, Bonn, Germany
| | - Annika Meinander
- Department of Cell Biology, Faculty of Science and Engineering, BioCity, Åbo Akademi University, 20520, Turku, Finland.
| |
Collapse
|
13
|
Begalli F, Bennett J, Capece D, Verzella D, D'Andrea D, Tornatore L, Franzoso G. Unlocking the NF-κB Conundrum: Embracing Complexity to Achieve Specificity. Biomedicines 2017; 5:E50. [PMID: 28829404 PMCID: PMC5618308 DOI: 10.3390/biomedicines5030050] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/04/2017] [Accepted: 08/10/2017] [Indexed: 12/12/2022] Open
Abstract
Transcription factors of the nuclear factor κB (NF-κB) family are central coordinating regulators of the host defence responses to stress, injury and infection. Aberrant NF-κB activation also contributes to the pathogenesis of some of the most common current threats to global human health, including chronic inflammatory diseases, autoimmune disorders, diabetes, vascular diseases and the majority of cancers. Accordingly, the NF-κB pathway is widely considered an attractive therapeutic target in a broad range of malignant and non-malignant diseases. Yet, despite the aggressive efforts by the pharmaceutical industry to develop a specific NF-κB inhibitor, none has been clinically approved, due to the dose-limiting toxicities associated with the global suppression of NF-κB. In this review, we summarise the main strategies historically adopted to therapeutically target the NF-κB pathway with an emphasis on oncology, and some of the emerging strategies and newer agents being developed to pharmacologically inhibit this pathway.
Collapse
Affiliation(s)
- Federica Begalli
- Centre for Cell Signalling and Inflammation, Department of Medicine, Imperial College London, London W12 0NN, UK.
| | - Jason Bennett
- Centre for Cell Signalling and Inflammation, Department of Medicine, Imperial College London, London W12 0NN, UK.
| | - Daria Capece
- Centre for Cell Signalling and Inflammation, Department of Medicine, Imperial College London, London W12 0NN, UK.
| | - Daniela Verzella
- Centre for Cell Signalling and Inflammation, Department of Medicine, Imperial College London, London W12 0NN, UK.
| | - Daniel D'Andrea
- Centre for Cell Signalling and Inflammation, Department of Medicine, Imperial College London, London W12 0NN, UK.
| | - Laura Tornatore
- Centre for Cell Signalling and Inflammation, Department of Medicine, Imperial College London, London W12 0NN, UK.
| | - Guido Franzoso
- Centre for Cell Signalling and Inflammation, Department of Medicine, Imperial College London, London W12 0NN, UK.
| |
Collapse
|
14
|
Mitsuuchi Y, Benetatos CA, Deng Y, Haimowitz T, Beck SC, Arnone MR, Kapoor GS, Seipel ME, Chunduru SK, McKinlay MA, Begley CG, Condon SM. Bivalent IAP antagonists, but not monovalent IAP antagonists, inhibit TNF-mediated NF- κB signaling by degrading TRAF2-associated cIAP1 in cancer cells. Cell Death Discov 2017; 3:16046. [PMID: 28149532 PMCID: PMC5238498 DOI: 10.1038/cddiscovery.2016.46] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 05/15/2016] [Indexed: 01/05/2023] Open
Abstract
The inhibitor of apoptosis (IAP) proteins have pivotal roles in cell proliferation and differentiation, and antagonizing IAPs in certain cancer cell lines results in induction of cell death. A variety of IAP antagonist compounds targeting the baculovirus IAP protein repeat 3 (BIR3) domain of cIAP1have advanced into clinical trials. Here we sought to compare and contrast the biochemical activities of selected monovalent and bivalent IAP antagonists with the intent of identifying functional differences between these two classes of IAP antagonist drug candidates. The anti-cellular IAP1 (cIAP1) and pro-apoptotic activities of monovalent IAP antagonists were increased by using a single covalent bond to combine the monovalent moieties at the P4 position. In addition, regardless of drug concentration, treatment with monovalent compounds resulted in consistently higher levels of residual cIAP1 compared with that seen following bivalent compound treatment. We found that the remaining residual cIAP1 following monovalent compound treatment was predominantly tumor necrosis factor (TNF) receptor-associated factor 2 (TRAF2)-associated cIAP1. As a consequence, bivalent compounds were more effective at inhibiting TNF-induced activation of p65/NF-κB compared with monovalent compounds. Moreover, extension of the linker chain at the P4 position of bivalent compounds resulted in a decreased ability to degrade TRAF2-associated cIAP1 in a manner similar to monovalent compounds. This result implied that specific bivalent IAP antagonists but not monovalent compounds were capable of inducing formation of a cIAP1 E3 ubiquitin ligase complex with the capacity to effectively degrade TRAF2-associated cIAP1. These results further suggested that only certain bivalent IAP antagonists are preferred for the targeting of TNF-dependent signaling for the treatment of cancer or infectious diseases.
Collapse
Affiliation(s)
- Y Mitsuuchi
- TetraLogic Pharmaceuticals Corporation , 343 Phoenixville Pike, Malvern, PA 19355, USA
| | - C A Benetatos
- TetraLogic Pharmaceuticals Corporation , 343 Phoenixville Pike, Malvern, PA 19355, USA
| | - Y Deng
- TetraLogic Pharmaceuticals Corporation , 343 Phoenixville Pike, Malvern, PA 19355, USA
| | - T Haimowitz
- TetraLogic Pharmaceuticals Corporation , 343 Phoenixville Pike, Malvern, PA 19355, USA
| | - S C Beck
- TetraLogic Pharmaceuticals Corporation , 343 Phoenixville Pike, Malvern, PA 19355, USA
| | - M R Arnone
- TetraLogic Pharmaceuticals Corporation , 343 Phoenixville Pike, Malvern, PA 19355, USA
| | - G S Kapoor
- TetraLogic Pharmaceuticals Corporation , 343 Phoenixville Pike, Malvern, PA 19355, USA
| | - M E Seipel
- TetraLogic Pharmaceuticals Corporation , 343 Phoenixville Pike, Malvern, PA 19355, USA
| | - S K Chunduru
- TetraLogic Pharmaceuticals Corporation , 343 Phoenixville Pike, Malvern, PA 19355, USA
| | - M A McKinlay
- TetraLogic Pharmaceuticals Corporation , 343 Phoenixville Pike, Malvern, PA 19355, USA
| | - C G Begley
- TetraLogic Pharmaceuticals Corporation , 343 Phoenixville Pike, Malvern, PA 19355, USA
| | - S M Condon
- TetraLogic Pharmaceuticals Corporation , 343 Phoenixville Pike, Malvern, PA 19355, USA
| |
Collapse
|
15
|
Abstract
Linear ubiquitination is a post‐translational protein modification recently discovered to be crucial for innate and adaptive immune signaling. The function of linear ubiquitin chains is regulated at multiple levels: generation, recognition, and removal. These chains are generated by the linear ubiquitin chain assembly complex (LUBAC), the only known ubiquitin E3 capable of forming the linear ubiquitin linkage de novo. LUBAC is not only relevant for activation of nuclear factor‐κB (NF‐κB) and mitogen‐activated protein kinases (MAPKs) in various signaling pathways, but importantly, it also regulates cell death downstream of immune receptors capable of inducing this response. Recognition of the linear ubiquitin linkage is specifically mediated by certain ubiquitin receptors, which is crucial for translation into the intended signaling outputs. LUBAC deficiency results in attenuated gene activation and increased cell death, causing pathologic conditions in both, mice, and humans. Removal of ubiquitin chains is mediated by deubiquitinases (DUBs). Two of them, OTULIN and CYLD, are constitutively associated with LUBAC. Here, we review the current knowledge on linear ubiquitination in immune signaling pathways and the biochemical mechanisms as to how linear polyubiquitin exerts its functions distinctly from those of other ubiquitin linkage types.
Collapse
Affiliation(s)
- Yutaka Shimizu
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, London, UK
| | - Lucia Taraborrelli
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, London, UK
| | - Henning Walczak
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, London, UK
| |
Collapse
|
16
|
Moriwaki K, Chan FKM. Necroptosis-independent signaling by the RIP kinases in inflammation. Cell Mol Life Sci 2016; 73:2325-34. [PMID: 27048814 PMCID: PMC4889460 DOI: 10.1007/s00018-016-2203-4] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 03/18/2016] [Indexed: 10/22/2022]
Abstract
Recent advances have identified a signaling cascade involving receptor interacting protein kinase 1 (RIPK1), RIPK3 and the pseudokinase mixed lineage kinase domain-like (MLKL) that is crucial for induction of necroptosis, a non-apoptotic form of cell death. RIPK1-RIPK3-MLKL-mediated necroptosis has been attributed to cause many inflammatory diseases through the release of cellular damage-associated molecular patterns (DAMPs). In addition to necroptosis, emerging evidence suggests that these necroptosis signal adaptors can also facilitate inflammation independent of cell death. In particular, the RIP kinases can drive NF-κB and inflammasome activation independent of cell death. In this review, we will discuss recent discoveries that led to this realization and present arguments why cell death-independent signaling by the RIP kinases may have a more important role in inflammation than necroptosis.
Collapse
Affiliation(s)
- Kenta Moriwaki
- Department of Pathology, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA, 01605, USA
| | - Francis Ka-Ming Chan
- Department of Pathology, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA, 01605, USA.
| |
Collapse
|
17
|
Peltzer N, Darding M, Walczak H. Holding RIPK1 on the Ubiquitin Leash in TNFR1 Signaling. Trends Cell Biol 2016; 26:445-461. [DOI: 10.1016/j.tcb.2016.01.006] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 01/13/2016] [Accepted: 01/19/2016] [Indexed: 12/22/2022]
|
18
|
Baud V, Collares D. Post-Translational Modifications of RelB NF-κB Subunit and Associated Functions. Cells 2016; 5:cells5020022. [PMID: 27153093 PMCID: PMC4931671 DOI: 10.3390/cells5020022] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 04/19/2016] [Accepted: 04/26/2016] [Indexed: 01/02/2023] Open
Abstract
The family of NF-κB transcription factors plays a key role in diverse biological processes, such as inflammatory and immune responses, cell survival and tumor development. Beyond the classical NF-κB activation pathway, a second NF-κB pathway has more recently been uncovered, the so-called alternative NF-κB activation pathway. It has been shown that this pathway mainly controls the activity of RelB, a member of the NF-κB family. Post-translational modifications, such as phosphorylation, acetylation, methylation, ubiquitination and SUMOylation, have recently emerged as a strategy for the fine-tuned regulation of NF-κB. Our review discusses recent progress in the understanding of RelB regulation by post-translational modifications and the associated functions in normal and pathological conditions.
Collapse
Affiliation(s)
- Véronique Baud
- NF-κB, Differentiation and Cancer, Université Paris Descartes, Sorbonne Paris Cité, 75014 Paris, France.
| | - Davi Collares
- NF-κB, Differentiation and Cancer, Université Paris Descartes, Sorbonne Paris Cité, 75014 Paris, France
| |
Collapse
|
19
|
Kupka S, Reichert M, Draber P, Walczak H. Formation and removal of poly-ubiquitin chains in the regulation of tumor necrosis factor-induced gene activation and cell death. FEBS J 2016; 283:2626-39. [PMID: 26749412 DOI: 10.1111/febs.13644] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 12/21/2015] [Accepted: 01/05/2016] [Indexed: 12/17/2022]
Abstract
Tumor necrosis factor (TNF) is a potent cytokine known for its involvement in inflammation, repression of tumorigenesis and activation of immune cells. Consequently, accurate regulation of the TNF signaling pathway is crucial for preventing the potent noxious effects of TNF. These pathological conditions include chronic inflammation, septic shock, cachexia and cancer. The TNF signaling cascade utilizes a complex network of post-translational modifications to control the cellular response following its activation. Next to phosphorylation, the ubiquitination of signaling complex components is probably the most important modification. This process is mediated by a specialist class of enzymes, the ubiquitin ligases. Equally important is the class of dedicated ubiquitin-specific proteases, the deubiquitinases. Together with ubiquitin binding proteins, this ubiquitination-deubiquitination system enables the dynamics of signaling complexes. In TNF signaling, these dynamics translate into the precise regulation of the induction of gene activation or cell death. Here, we review and discuss current knowledge of TNF signaling regulation by the ubiquitin system.
Collapse
Affiliation(s)
- Sebastian Kupka
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, London, UK
| | - Matthias Reichert
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, London, UK
| | - Peter Draber
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, London, UK
| | - Henning Walczak
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, London, UK
| |
Collapse
|
20
|
AGUIRIANO-MOSER VICTOR, SVEJDA BERNHARD, LI ZENGXIA, STURM SONJA, STUPPNER HERMANN, INGOLIC ELISABETH, HÖGER HARALD, SIEGL VERONIKA, MEIER-ALLARD NATHALIE, SADJAK ANTON, PFRAGNER ROSWITHA. Ursolic acid from Trailliaedoxa gracilis induces apoptosis in medullary thyroid carcinoma cells. Mol Med Rep 2015; 12:5003-11. [PMID: 26151624 PMCID: PMC4581794 DOI: 10.3892/mmr.2015.4053] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 02/10/2015] [Indexed: 01/08/2023] Open
Abstract
Medullary thyroid carcinoma (MTC) originates from the C‑cells of the thyroid and is not sensitive to radiation or chemotherapy. Therefore, surgical removal of the tumor tissue in its entirety is the only curative treatment for MTC. The present study aimed to examine the potential mechanisms of action of extracts of Trailliaedoxa gracilis (TG; WW Smith & Forrest), a plant from the province of Sichuan, China, and of ursolic acid (UA), a pentacyclic triterpen present in TG, on the MTC‑SK MTC cell line. A total of 13 TG fractions and UA were examined in vitro for their effects on cell morphology, cell number, proliferation and rates of apoptosis. Reverse transcription‑quantitative polymerase chain reaction of nuclear factor‑κB essential modifier (NEMO) was performed to delineate the role of the apoptotic pathway following treatment with UA. TG and UA were examined in vivo in xenotransplanted MTC‑bearing severe combined immunodeficient mice. The TG fractions exhibited antiproliferative effects, with inhibition of mitochondrial activity in the tumor cells at concentrations, which caused no impairment of the normal control cells. The apoptotic rates of the MTC‑SK cells treated with the TG fractions and UA were determined, in which no marked tumor inhibition was observed in the treated MTC‑mice, and no change in the expression of NEMO was detected in the treated MTC‑SK cells. The observation of early‑onset activation of caspase 8 suggested that the responsible factor was linked to NEMO, an anti‑apoptotic protein. However, no differences in the mRNA transcription levels of NEMO were detected in MTC‑SK cells treated with UA, suggesting that this protein was not associated with the signal transducer and activator of transcription 3 pathway.
Collapse
Affiliation(s)
- VICTOR AGUIRIANO-MOSER
- Department of Pathophysiology and Immunology, Center of Molecular Medicine, Medical University of Graz, Graz A-8010, Austria
| | - BERNHARD SVEJDA
- Department of Pathophysiology and Immunology, Center of Molecular Medicine, Medical University of Graz, Graz A-8010, Austria
| | - ZENG-XIA LI
- Department of Pathophysiology and Immunology, Center of Molecular Medicine, Medical University of Graz, Graz A-8010, Austria
- Department of Biochemistry & Molecular Biology, Shanghai Medical School, Fudan University, Shanghai 200433, P.R. China
| | - SONJA STURM
- Department of Pharmacognosy, Institute of Pharmacy, Center of Molecular Biosciences, Leopold Franzens University of Innsbruck, Innsbruck A-6010, Austria
| | - HERMANN STUPPNER
- Department of Pharmacognosy, Institute of Pharmacy, Center of Molecular Biosciences, Leopold Franzens University of Innsbruck, Innsbruck A-6010, Austria
| | - ELISABETH INGOLIC
- Core Unit of Biomedical Research, ivision of Laboratory Animal Science and Genetics, Medical University of Vienna, Himberg A-2325, Austria
| | - HARALD HÖGER
- Research Institute for Electron Microscopy and Fine Structure Research, University of Technology Graz, Graz A-8010, Austria
| | - VERONIKA SIEGL
- Department of Pathophysiology and Immunology, Center of Molecular Medicine, Medical University of Graz, Graz A-8010, Austria
| | - NATHALIE MEIER-ALLARD
- Department of Pathophysiology and Immunology, Center of Molecular Medicine, Medical University of Graz, Graz A-8010, Austria
| | - ANTON SADJAK
- Department of Pathophysiology and Immunology, Center of Molecular Medicine, Medical University of Graz, Graz A-8010, Austria
| | - ROSWITHA PFRAGNER
- Department of Pathophysiology and Immunology, Center of Molecular Medicine, Medical University of Graz, Graz A-8010, Austria
| |
Collapse
|
21
|
An apoptosis-enhancing drug overcomes platinum resistance in a tumour-initiating subpopulation of ovarian cancer. Nat Commun 2015; 6:7956. [PMID: 26234182 PMCID: PMC4532886 DOI: 10.1038/ncomms8956] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 06/29/2015] [Indexed: 02/07/2023] Open
Abstract
High-grade serous ovarian cancers (HGSCs) are deadly malignancies that relapse despite carboplatin chemotherapy. Here we show that 16 independent primary HGSC samples contain a CA125-negative population enriched for carboplatin-resistant cancer initiating cells. Transcriptome analysis reveals upregulation of homologous recombination DNA repair and anti-apoptotic signals in this population. While treatment with carboplatin enriches for CA125-negative cells, co-treatment with carboplatin and birinapant eliminates these cells in HGSCs expressing high levels of the inhibitor of apoptosis protein cIAP in the CA125-negative population. Birinapant sensitizes CA125-negative cells to carboplatin by mediating degradation of cIAP causing cleavage of caspase 8 and restoration of apoptosis. This co-therapy significantly improves disease-free survival in vivo compared with either therapy alone in tumour-bearing mice. These findings suggest that therapeutic strategies that target CA125-negative cells may be useful in the treatment of HGSC. Despite normalization of the CA125 serum biomarker at the completion of carboplatin therapy the vast majority of patients with high grade serous ovarian cancers relapse. Here, Janzen et al., identify a sub-population of tumor cells that are CA125 negative, cancer initiating and platinum resistant but readily eliminated with the addition of apoptosis enhancing drugs to carboplatin.
Collapse
|
22
|
Catching a DUB in the act: novel ubiquitin-based active site directed probes. Curr Opin Chem Biol 2015; 23:63-70. [PMID: 25461387 PMCID: PMC7185813 DOI: 10.1016/j.cbpa.2014.10.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 09/30/2014] [Accepted: 10/08/2014] [Indexed: 01/21/2023]
Abstract
Activity-based probes used to probe DUB inhibitor specificity. Developments in DUB activity-based probe use. Determination of linkage preference of DUBs using activity-based probes. Outlook for development in DUB probe design and current challenges.
Protein ubiquitylation is an important regulator of protein function, localization and half-life. It plays a key role in most cellular processes including immune signaling. Deregulation of this process is a major causative factor for many diseases. A major advancement in the identification and characterization of the enzymes that remove ubiquitin, deubiquitylases (DUBs) was made by the development of activity-based probes (ABPs). Recent advances in chemical protein synthesis and ligation methodology has yielded novel reagents for use in ubiquitylation research. We describe recent advances and discuss future directions in reagent development for studying DUBs.
Collapse
|
23
|
Greenfeld H, Takasaki K, Walsh MJ, Ersing I, Bernhardt K, Ma Y, Fu B, Ashbaugh CW, Cabo J, Mollo SB, Zhou H, Li S, Gewurz BE. TRAF1 Coordinates Polyubiquitin Signaling to Enhance Epstein-Barr Virus LMP1-Mediated Growth and Survival Pathway Activation. PLoS Pathog 2015; 11:e1004890. [PMID: 25996949 PMCID: PMC4440769 DOI: 10.1371/journal.ppat.1004890] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 04/17/2015] [Indexed: 11/25/2022] Open
Abstract
The Epstein-Barr virus (EBV) encoded oncoprotein Latent Membrane Protein 1 (LMP1) signals through two C-terminal tail domains to drive cell growth, survival and transformation. The LMP1 membrane-proximal TES1/CTAR1 domain recruits TRAFs to activate MAP kinase, non-canonical and canonical NF-kB pathways, and is critical for EBV-mediated B-cell transformation. TRAF1 is amongst the most highly TES1-induced target genes and is abundantly expressed in EBV-associated lymphoproliferative disorders. We found that TRAF1 expression enhanced LMP1 TES1 domain-mediated activation of the p38, JNK, ERK and canonical NF-kB pathways, but not non-canonical NF-kB pathway activity. To gain insights into how TRAF1 amplifies LMP1 TES1 MAP kinase and canonical NF-kB pathways, we performed proteomic analysis of TRAF1 complexes immuno-purified from cells uninduced or induced for LMP1 TES1 signaling. Unexpectedly, we found that LMP1 TES1 domain signaling induced an association between TRAF1 and the linear ubiquitin chain assembly complex (LUBAC), and stimulated linear (M1)-linked polyubiquitin chain attachment to TRAF1 complexes. LMP1 or TRAF1 complexes isolated from EBV-transformed lymphoblastoid B cell lines (LCLs) were highly modified by M1-linked polyubiqutin chains. The M1-ubiquitin binding proteins IKK-gamma/NEMO, A20 and ABIN1 each associate with TRAF1 in cells that express LMP1. TRAF2, but not the cIAP1 or cIAP2 ubiquitin ligases, plays a key role in LUBAC recruitment and M1-chain attachment to TRAF1 complexes, implicating the TRAF1:TRAF2 heterotrimer in LMP1 TES1-dependent LUBAC activation. Depletion of either TRAF1, or the LUBAC ubiquitin E3 ligase subunit HOIP, markedly impaired LCL growth. Likewise, LMP1 or TRAF1 complexes purified from LCLs were decorated by lysine 63 (K63)-linked polyubiqutin chains. LMP1 TES1 signaling induced K63-polyubiquitin chain attachment to TRAF1 complexes, and TRAF2 was identified as K63-Ub chain target. Co-localization of M1- and K63-linked polyubiquitin chains on LMP1 complexes may facilitate downstream canonical NF-kB pathway activation. Our results highlight LUBAC as a novel potential therapeutic target in EBV-associated lymphoproliferative disorders. The linear ubiquitin assembly complex (LUBAC) plays crucial roles in immune receptor-mediated NF-kB and MAP kinase pathway activation. Comparatively little is known about the extent to which microbial pathogens use LUBAC to activate downstream pathways. We demonstrate that TRAF1 enhances EBV oncoprotein LMP1 TES1/CTAR1 domain mediated MAP kinase and canonical NF-kB activation. LMP1 TES1 signaling induces association between TRAF1 and LUBAC, and triggers M1-polyubiquitin chain attachment to TRAF1 complexes. TRAF1 and LMP1 complexes are decorated by M1-polyubiquitin chains in LCL extracts. TRAF2 plays a key role in LMP1-induced LUBAC recruitment and M1-chain attachment to TRAF1 complexes. TRAF1 and LMP1 complexes are modified by lysine 63-linked polyubiquitin chains in LCL extracts, and TRAF2 is a target of LMP1-induced K63-ubiquitin chain attachment. Thus, the TRAF1:TRAF2 heterotrimer may coordinate ubiquitin signaling downstream of TES1. Depletion of TRAF1 or the LUBAC subunit HOIP impairs LCL growth and survival. Thus, although TRAF1 is the only TRAF without a RING finger ubiquitin ligase domain, TRAF1 nonetheless has important roles in ubiqutin-mediated signal transduction downstream of LMP1. Our work suggests that LUBAC is important for EBV-driven B-cell proliferation, and suggests that LUBAC may be a novel therapeutic target in EBV-associated lymphoproliferative disorders.
Collapse
Affiliation(s)
- Hannah Greenfeld
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Kaoru Takasaki
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Michael J. Walsh
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Ina Ersing
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Katharina Bernhardt
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Yijie Ma
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Bishi Fu
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Camille W. Ashbaugh
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Jackson Cabo
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Sarah B. Mollo
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Hufeng Zhou
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Shitao Li
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Benjamin E. Gewurz
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
24
|
Vaden JH, Watson JA, Howard AD, Chen PC, Wilson JA, Wilson SM. Distinct effects of ubiquitin overexpression on NMJ structure and motor performance in mice expressing catalytically inactive USP14. Front Mol Neurosci 2015; 8:11. [PMID: 25954152 PMCID: PMC4407586 DOI: 10.3389/fnmol.2015.00011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 04/06/2015] [Indexed: 01/24/2023] Open
Abstract
Ubiquitin-specific protease 14 (USP14) is a major deubiquitinating enzyme and a key determinant of neuromuscular junction (NMJ) structure and function. We have previously reported dramatic ubiquitin depletion in the nervous systems of the USP14-deficient ataxia (axJ) mice and demonstrated that transgenic ubiquitin overexpression partially rescues the axJ neuromuscular phenotype. However, later work has shown that ubiquitin overexpression does not correct the axJ deficits in hippocampal short term plasticity, and that transgenic expression of a catalytically inactive form of USP14 in the nervous system mimics the neuromuscular phenotype observed in the axJ mice, but causes a only a modest reduction of free ubiquitin. Instead, increased ubiquitin conjugates and aberrant activation of pJNK are observed in the nervous systems of the USP14 catalytic mutant mice. In this report, we demonstrate that restoring free ubiquitin levels in the USP14 catalytic mutant mice improved NMJ structure and reduced pJNK accumulation in motor neuron terminals, but had a negative impact on measures of NMJ function, such as motor performance and muscle development. Transgenic expression of ubiquitin had a dose-dependent effect on NMJ function in wild type mice: moderate levels of overexpression improved NMJ function while more robust ubiquitin overexpression reduced muscle development and motor coordination. Combined, these results suggest that maintenance of free ubiquitin levels by USP14 contributes to NMJ structure, but that USP14 regulates NMJ function through a separate pathway.
Collapse
Affiliation(s)
- Jada H Vaden
- Evelyn F. McKnight Brain Institute, Department of Neurobiology and Civitan International Research Center, University of Alabama at Birmingham Birmingham, AL, USA
| | - Jennifer A Watson
- Evelyn F. McKnight Brain Institute, Department of Neurobiology and Civitan International Research Center, University of Alabama at Birmingham Birmingham, AL, USA
| | - Alan D Howard
- Evelyn F. McKnight Brain Institute, Department of Neurobiology and Civitan International Research Center, University of Alabama at Birmingham Birmingham, AL, USA
| | - Ping-Chung Chen
- Department of Structural Biology, St. Jude Children's Research Hospital Memphis, TN, USA
| | - Julie A Wilson
- Evelyn F. McKnight Brain Institute, Department of Neurobiology and Civitan International Research Center, University of Alabama at Birmingham Birmingham, AL, USA
| | - Scott M Wilson
- Evelyn F. McKnight Brain Institute, Department of Neurobiology and Civitan International Research Center, University of Alabama at Birmingham Birmingham, AL, USA
| |
Collapse
|
25
|
Sorokin AV, Nair BC, Wei Y, Aziz KE, Evdokimova V, Hung MC, Chen J. Aberrant Expression of proPTPRN2 in Cancer Cells Confers Resistance to Apoptosis. Cancer Res 2015; 75:1846-58. [PMID: 25877877 DOI: 10.1158/0008-5472.can-14-2718] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 02/01/2015] [Indexed: 01/09/2023]
Abstract
The protein tyrosine phosphatase receptor PTPRN2 is expressed predominantly in endocrine and neuronal cells, where it functions in exocytosis. We found that its immature isoform proPTPRN2 is overexpressed in various cancers, including breast cancer. High proPTPRN2 expression was associated strongly with lymph node-positive breast cancer and poor clinical outcome. Loss of proPTPRN2 in breast cancer cells promoted apoptosis and blocked tumor formation in mice, whereas enforced expression of proPTPRN2 in nontransformed human mammary epithelial cells exerted a converse effect. Mechanistic investigations suggested that ProPTPRN2 elicited these effects through direct interaction with TRAF2, a hub scaffold protein for multiple kinase cascades, including ones that activate NF-κB. Overall, our results suggest PTPRN2 as a novel candidate biomarker and therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Alexey V Sorokin
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Binoj C Nair
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yongkun Wei
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kathryn E Aziz
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Valentina Evdokimova
- Department of Genomics, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Center for Molecular Medicine and Graduate Institute of Cancer Biology, China Medical University, Taichung, Taiwan
| | - Junjie Chen
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
26
|
Dowling JP, Nair A, Zhang J. A novel function of RIP1 in postnatal development and immune homeostasis by protecting against RIP3-dependent necroptosis and FADD-mediated apoptosis. Front Cell Dev Biol 2015; 3:12. [PMID: 25767797 PMCID: PMC4341114 DOI: 10.3389/fcell.2015.00012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 02/10/2015] [Indexed: 01/10/2023] Open
Abstract
RIP1 is an adaptor kinase originally identified as being able to associate with TNFR1 and Fas, and is later shown to be involved in signaling induced by TLRs. Major signaling pathways regulated by RIP1 include necroptosis, apoptosis, and pro-survival/inflammation NF-κB activation. Previous studies show that RIP1 deficiency has no effect on mouse embryogenesis, but blocks postnatal development. This phenotype could not readily be explained, since mice lacking TNFR1, Fas, or TLRs show no apparent developmental defect. Certain types of RIP1-deficient cells are hypersensitive to TNF-induced apoptosis. However, in our previous study, deletion of the apoptotic adaptor protein, FADD, provides marginal improvement of postnatal development of rip1−/− mice. Remarkably, the current data shows that haploid insufficiency of RIP3, a known mediator of necroptosis, allowed survival of rip1−/−fadd−/− mice beyond weaning age, although the resulting rip1−/−fadd−/− rip3+/− mice were significant smaller in size and weight. Moreover, complete absence of RIP3 further improved postnatal development of the resulting rip1−/−fadd−/−rip3−/− mice, which display normal size and weight. In such triple knockout (TKO) mice, lymphocytes underwent normal development, but progressively accumulated as mice age. This lymphoproliferative (lpr) disease in TKO mice is, however, less severe than that of fadd−/−rip3−/− double knockout mice. In total, the data show that the postnatal developmental defect in rip1−/− mice is due in part to FADD-mediated apoptosis as well as RIP3-dependent necroptosis. Moreover, the function of RIP1 contributes to development of lpr diseases.
Collapse
Affiliation(s)
- John P Dowling
- Department of Microbiology and Immunology, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College, Thomas Jefferson University Philadelphia, PA, USA
| | - Anirudh Nair
- Department of Microbiology and Immunology, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College, Thomas Jefferson University Philadelphia, PA, USA
| | - Jianke Zhang
- Department of Microbiology and Immunology, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College, Thomas Jefferson University Philadelphia, PA, USA
| |
Collapse
|
27
|
Enesa K, Moll HP, Luong L, Ferran C, Evans PC. A20 suppresses vascular inflammation by recruiting proinflammatory signaling molecules to intracellular aggresomes. FASEB J 2015; 29:1869-78. [PMID: 25667218 DOI: 10.1096/fj.14-258533] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 12/24/2014] [Indexed: 12/16/2022]
Abstract
A20 protects against pathologic vascular remodeling by inhibiting the inflammatory transcription factor NF-κB. A20's function has been attributed to ubiquitin editing of receptor-interacting protein 1 (RIP1) to influence activity/stability. The validity of this mechanism was tested using a murine model of transplant vasculopathy and human cells. Mouse C57BL/6 aortae transduced with adenoviruses containing A20 (or β-galactosidase as a control) were allografted into major histocompatibility complex-mismatched BALB/c mice. Primary endothelial cells, smooth muscle cells, or transformed epithelial cells (all human) were transfected with wild-type A20 or with catalytically inactive mutants as a control. NF-κB activity and intracellular localization of RIP1 was monitored by reporter gene assay, immunofluorescent staining, and Western blotting. Native and catalytically inactive versions of A20 had similar inhibitory effects on NF-κB activity (-70% vs. -76%; P > 0.05). A20 promoted localization of RIP1 to insoluble aggresomes in murine vascular allografts and in human cells (53% vs. 0%) without altering RIP1 expression, and this process was increased by the assembly of polyubiquitin chains (87% vs. 28%; P < 0.05). A20 captures polyubiquitinated signaling intermediaries in insoluble aggresomes, thus reducing their bioavailability for downstream NF-κB signaling. This novel mechanism contributes to protection from vasculopathy in transplanted organs treated with exogenous A20.
Collapse
Affiliation(s)
- Karine Enesa
- *British Heart Foundation Cardiovascular Sciences Unit, Imperial College London, London, United Kingdom; Division of Vascular and Endovascular Surgery, Center for Vascular Biology Research and the Transplant Institute, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Cambridge, Massachusetts, USA; and Department of Cardiovascular Sciences and INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, United Kingdom
| | - Herwig P Moll
- *British Heart Foundation Cardiovascular Sciences Unit, Imperial College London, London, United Kingdom; Division of Vascular and Endovascular Surgery, Center for Vascular Biology Research and the Transplant Institute, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Cambridge, Massachusetts, USA; and Department of Cardiovascular Sciences and INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, United Kingdom
| | - Le Luong
- *British Heart Foundation Cardiovascular Sciences Unit, Imperial College London, London, United Kingdom; Division of Vascular and Endovascular Surgery, Center for Vascular Biology Research and the Transplant Institute, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Cambridge, Massachusetts, USA; and Department of Cardiovascular Sciences and INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, United Kingdom
| | - Christiane Ferran
- *British Heart Foundation Cardiovascular Sciences Unit, Imperial College London, London, United Kingdom; Division of Vascular and Endovascular Surgery, Center for Vascular Biology Research and the Transplant Institute, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Cambridge, Massachusetts, USA; and Department of Cardiovascular Sciences and INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, United Kingdom
| | - Paul C Evans
- *British Heart Foundation Cardiovascular Sciences Unit, Imperial College London, London, United Kingdom; Division of Vascular and Endovascular Surgery, Center for Vascular Biology Research and the Transplant Institute, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Cambridge, Massachusetts, USA; and Department of Cardiovascular Sciences and INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
28
|
Gudi R, Haycraft CJ, Bell PD, Li Z, Vasu C. Centrobin-mediated regulation of the centrosomal protein 4.1-associated protein (CPAP) level limits centriole length during elongation stage. J Biol Chem 2015; 290:6890-902. [PMID: 25616662 DOI: 10.1074/jbc.m114.603423] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Microtubule-based centrioles in the centrosome mediate accurate bipolar cell division, spindle orientation, and primary cilia formation. Cellular checkpoints ensure that the centrioles duplicate only once in every cell cycle and achieve precise dimensions, dysregulation of which results in genetic instability and neuro- and ciliopathies. The normal cellular level of centrosomal protein 4.1-associated protein (CPAP), achieved by its degradation at mitosis, is considered as one of the major mechanisms that limits centriole growth at a predetermined length. Here we show that CPAP levels and centriole elongation are regulated by centrobin. Exogenous expression of centrobin causes abnormal elongation of centrioles due to massive accumulation of CPAP in the cell. Conversely, CPAP was undetectable in centrobin-depleted cells, suggesting that it undergoes degradation in the absence of centrobin. Only the reintroduction of full-length centrobin, but not its mutant form that lacks the CPAP binding site, could restore cellular CPAP levels in centrobin-depleted cells, indicating that persistence of CPAP requires its interaction with centrobin. Interestingly, inhibition of the proteasome in centrobin-depleted cells restored the cellular and centriolar CPAP expression, suggesting its ubiquitination and proteasome-mediated degradation when centrobin is absent. Intriguingly, however, centrobin-overexpressing cells also showed proteasome-independent accumulation of ubiquitinated CPAP and abnormal, ubiquitin-positive, elongated centrioles. Overall, our results show that centrobin interacts with ubiquitinated CPAP and prevents its degradation for normal centriole elongation function. Therefore, it appears that loss of centrobin expression destabilizes CPAP and triggers its degradation to restrict the centriole length during biogenesis.
Collapse
Affiliation(s)
- Radhika Gudi
- From the Department of Microbiology and Immunology,
| | | | | | - Zihai Li
- From the Department of Microbiology and Immunology
| | - Chenthamarakshan Vasu
- From the Department of Microbiology and Immunology, Department of Surgery, Medical University of South Carolina, Charleston, South Carolina 29425
| |
Collapse
|
29
|
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common aggressive non-Hodgkin's lymphoma. Next-generation sequencing techniques have improved our understanding of the molecular pathways that may drive oncogenesis. Many novel classes of drugs are in development that may improve the treatment of DLBCL, either as single agents or in combination, that exploit their synergy to overcome resistance. We review the key novel targets and therapeutics in the treatment of DLBCL, including immunomodulatory agents and immunotherapy.
Collapse
Affiliation(s)
- Neha Mehta-Shah
- Lymphoma Service, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Anas Younes
- Lymphoma Service, Memorial Sloan-Kettering Cancer Center, New York, NY.
| |
Collapse
|
30
|
Vaden JH, Bhattacharyya BJ, Chen PC, Watson JA, Marshall AG, Phillips SE, Wilson JA, King GD, Miller RJ, Wilson SM. Ubiquitin-specific protease 14 regulates c-Jun N-terminal kinase signaling at the neuromuscular junction. Mol Neurodegener 2015; 10:3. [PMID: 25575639 PMCID: PMC4417291 DOI: 10.1186/1750-1326-10-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 12/17/2014] [Indexed: 01/13/2023] Open
Abstract
Background Ubiquitin-specific protease 14 (USP14) is one of three proteasome-associated deubiquitinating enzymes that remove ubiquitin from proteasomal substrates prior to their degradation. In vitro evidence suggests that inhibiting USP14’s catalytic activity alters the turnover of ubiquitinated proteins by the proteasome, although whether protein degradation is accelerated or delayed seems to be cell-type and substrate specific. For example, combined inhibition of USP14 and the proteasomal deubiquitinating enzyme UCH37 halts protein degradation and promotes apoptosis in multiple myeloma cells, whereas USP14 inhibition alone accelerates the degradation of aggregate-prone proteins in immortalized cell lines. These findings have prompted interest in USP14 as a therapeutic target both inside and outside of the nervous system. However, loss of USP14 in the spontaneously occurring ataxia mouse mutant leads to a dramatic neuromuscular phenotype and early perinatal lethality, suggesting that USP14 inhibition may have adverse consequences in the nervous system. We therefore expressed a catalytically inactive USP14 mutant in the mouse nervous system to determine whether USP14’s catalytic activity is required for neuromuscular junction (NMJ) structure and function. Results Mice expressing catalytically inactive USP14 in the nervous system exhibited motor deficits, altered NMJ structure, and synaptic transmission deficits that were similar to what is observed in the USP14-deficient ataxia mice. Acute pharmacological inhibition of USP14 in wild type mice also reduced NMJ synaptic transmission. However, there was no evidence of altered proteasome activity when USP14 was inhibited either genetically or pharmacologically. Instead, these manipulations increased the levels of non-proteasome targeting ubiquitin conjugates. Specifically, we observed enhanced proteasome-independent ubiquitination of mixed lineage kinase 3 (MLK3). Consistent with the direct activation of MLK3 by ubiquitination, we also observed increased activation of its downstrea targets MAP kinase kinase 4 (MKK4) and c-Jun N-terminal kinase (JNK). In vivo inhibition of JNK improved motor function and synapse structure in the USP14 catalytic mutant mice. Conclusions USP14’s catalytic activity is required for nervous system structure and function and has an ongoing role in NMJ synaptic transmission. By regulating the ubiquitination status of protein kinases, USP14 can coordinate the activity of intracellular signaling pathways that control the development and activity of the NMJ. Electronic supplementary material The online version of this article (doi:10.1186/1750-1326-10-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jada H Vaden
- Department of Neurobiology, University of Alabama at Birmingham, Civitan International Research Center, Evelyn F. McKnight Brain Institute, 1825 University Blvd, Birmingham, AL, 35294, USA.
| | - Bula J Bhattacharyya
- Department of Molecular Pharmacology and Biological Chemistry, Northwestern University, 303 East Chicago Ave, Chicago, IL, 60611, USA.
| | - Ping-Chung Chen
- Department of Structural Biology, St. Jude Children's Research Hospital, Danny Thomas Place, Memphis, TN, 38105-3678, USA.
| | - Jennifer A Watson
- Department of Neurobiology, University of Alabama at Birmingham, Civitan International Research Center, Evelyn F. McKnight Brain Institute, 1825 University Blvd, Birmingham, AL, 35294, USA.
| | - Andrea G Marshall
- Department of Physiology, Neurobiology and Behavior, University of California, Davis, California, CA, USA.
| | - Scott E Phillips
- Department of Neurobiology, University of Alabama at Birmingham, Civitan International Research Center, Evelyn F. McKnight Brain Institute, 1825 University Blvd, Birmingham, AL, 35294, USA.
| | - Julie A Wilson
- Department of Neurobiology, University of Alabama at Birmingham, Civitan International Research Center, Evelyn F. McKnight Brain Institute, 1825 University Blvd, Birmingham, AL, 35294, USA.
| | - Gwendalyn D King
- Department of Neurobiology, University of Alabama at Birmingham, Civitan International Research Center, Evelyn F. McKnight Brain Institute, 1825 University Blvd, Birmingham, AL, 35294, USA.
| | - Richard J Miller
- Department of Molecular Pharmacology and Biological Chemistry, Northwestern University, 303 East Chicago Ave, Chicago, IL, 60611, USA.
| | - Scott M Wilson
- Department of Neurobiology, University of Alabama at Birmingham, Civitan International Research Center, Evelyn F. McKnight Brain Institute, 1825 University Blvd, Birmingham, AL, 35294, USA.
| |
Collapse
|
31
|
Kumari S, Redouane Y, Lopez-Mosqueda J, Shiraishi R, Romanowska M, Lutzmayer S, Kuiper J, Martinez C, Dikic I, Pasparakis M, Ikeda F. Sharpin prevents skin inflammation by inhibiting TNFR1-induced keratinocyte apoptosis. eLife 2014; 3. [PMID: 25443631 PMCID: PMC4225491 DOI: 10.7554/elife.03422] [Citation(s) in RCA: 244] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 10/03/2014] [Indexed: 11/17/2022] Open
Abstract
Linear Ubiquitin chain Assembly Complex (LUBAC) is an E3 ligase complex that generates linear ubiquitin chains and is important for tumour necrosis factor (TNF) signaling activation. Mice lacking Sharpin, a critical subunit of LUBAC, spontaneously develop inflammatory lesions in the skin and other organs. Here we show that TNF receptor 1 (TNFR1)-associated death domain (TRADD)-dependent TNFR1 signaling in epidermal keratinocytes drives skin inflammation in Sharpin-deficient mice. Epidermis-restricted ablation of Fas-associated protein with death domain (FADD) combined with receptor-interacting protein kinase 3 (RIPK3) deficiency fully prevented skin inflammation, while single RIPK3 deficiency only delayed and partly ameliorated lesion development in Sharpin-deficient mice, showing that inflammation is primarily driven by TRADD- and FADD-dependent keratinocyte apoptosis while necroptosis plays a minor role. At the cellular level, Sharpin deficiency sensitized primary murine keratinocytes, human keratinocytes, and mouse embryonic fibroblasts to TNF-induced apoptosis. Depletion of FADD or TRADD in Sharpin-deficient HaCaT cells suppressed TNF-induced apoptosis, indicating the importance of FADD and TRADD in Sharpin-dependent anti-apoptosis signaling in keratinocytes. DOI:http://dx.doi.org/10.7554/eLife.03422.001 In response to an injury or an infection, areas of the body can become inflamed as the immune system attempts to repair the damage and/or destroy any microbes or toxins that have entered the body. At the level of individual cells inflammation can involve cells being programmed to die in one of two ways: apoptosis and necroptosis. Apoptosis is a highly controlled process during which the contents of the cell are safely destroyed in order to prevent damage to surrounding cells. Necroptosis, on the other hand, is not controlled: the cell bursts and releases its contents into the surroundings. Inflammation is activated by a protein called TNF, which is controlled by a complex that includes a protein called Sharpin. Mice that lack the Sharpin protein develop inflammation on the skin and other organs, even in the absence of injury or infection. However, it is not clear how the Sharpin protein controls TNF to prevent inflammation. Kumari et al. have found that inflammation in mice lacking Sharpin depends on TNF interacting with another protein called TRADD. The experiments also show that the inflammation is mainly driven by apoptosis, with necroptosis having only a minor role. Further experiments carried out in mammal cells showed that TRADD and another protein (called FADD) work with Sharpin to prevent apoptosis. At the molecular level, Sharpin is known to induce a special type of protein modification (called linear ubiquitination) with two partner proteins, so the next challenge is to work out exactly how Sharpin uses this process to prevent apoptosis. DOI:http://dx.doi.org/10.7554/eLife.03422.002
Collapse
Affiliation(s)
- Snehlata Kumari
- Institute for Genetics, Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | | | - Jaime Lopez-Mosqueda
- Institute of Biochemistry II, Goethe University Medical School, Frankfurt am Main, Germany
| | | | - Malgorzata Romanowska
- Institute for Genetics, Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | | | - Jan Kuiper
- Institute for Genetics, Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | | | - Ivan Dikic
- Institute of Biochemistry II, Goethe University Medical School, Frankfurt am Main, Germany
| | - Manolis Pasparakis
- Institute for Genetics, Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Fumiyo Ikeda
- Institute of Molecular Biotechnology, Vienna, Austria
| |
Collapse
|
32
|
Rickard JA, Anderton H, Etemadi N, Nachbur U, Darding M, Peltzer N, Lalaoui N, Lawlor KE, Vanyai H, Hall C, Bankovacki A, Gangoda L, Wong WWL, Corbin J, Huang C, Mocarski ES, Murphy JM, Alexander WS, Voss AK, Vaux DL, Kaiser WJ, Walczak H, Silke J. TNFR1-dependent cell death drives inflammation in Sharpin-deficient mice. eLife 2014; 3. [PMID: 25443632 PMCID: PMC4270099 DOI: 10.7554/elife.03464] [Citation(s) in RCA: 316] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 10/26/2014] [Indexed: 12/04/2022] Open
Abstract
SHARPIN regulates immune signaling and contributes to full transcriptional activity and prevention of cell death in response to TNF in vitro. The inactivating mouse Sharpin cpdm mutation causes TNF-dependent multi-organ inflammation, characterized by dermatitis, liver inflammation, splenomegaly, and loss of Peyer's patches. TNF-dependent cell death has been proposed to cause the inflammatory phenotype and consistent with this we show Tnfr1, but not Tnfr2, deficiency suppresses the phenotype (and it does so more efficiently than Il1r1 loss). TNFR1-induced apoptosis can proceed through caspase-8 and BID, but reduction in or loss of these players generally did not suppress inflammation, although Casp8 heterozygosity significantly delayed dermatitis. Ripk3 or Mlkl deficiency partially ameliorated the multi-organ phenotype, and combined Ripk3 deletion and Casp8 heterozygosity almost completely suppressed it, even restoring Peyer's patches. Unexpectedly, Sharpin, Ripk3 and Casp8 triple deficiency caused perinatal lethality. These results provide unexpected insights into the developmental importance of SHARPIN. DOI:http://dx.doi.org/10.7554/eLife.03464.001 In response to an injury or infection, areas of the body can become inflamed as the immune system attempts to repair the damage and/or destroy any microbes or toxins that have entered the body. At the level of individual cells inflammation can involve cells being programmed to die in one of two ways: apoptosis and necroptosis. Apoptosis is a highly controlled process during which the contents of the cell are safely destroyed in order to prevent damage to surrounding cells. Necroptosis, on the other hand, is not controlled: the cell bursts and releases its contents into the surroundings. Inflammation is activated by a protein called TNFR1, which is controlled by a complex that includes a protein called SHARPIN. Mice that lack the SHARPIN protein develop inflammation on the skin and internal organs, even in the absence of injury or infection. However, it is not clear how SHARPIN controls TNFR1 to prevent inflammation. Rickard et al. and, independently Kumari et al. have now studied this process in detail. Rickard et al. cross bred mice that lack SHARPIN with mice lacking other proteins involved in inflammation and cell death. The experiments show that apoptosis is the main form of cell death in skin inflammation, but necroptosis has a bigger role in the inflammation of internal organs. Mice that lack both the apoptotic and necroptotic cell-death pathways can develop relatively normally, but they die shortly after birth if they also lack SHARPIN. Experiments on these mice could help us to understand how SHARPIN works. DOI:http://dx.doi.org/10.7554/eLife.03464.002
Collapse
Affiliation(s)
- James A Rickard
- Cell Signalling and Cell Death Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Holly Anderton
- Cell Signalling and Cell Death Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Nima Etemadi
- Cell Signalling and Cell Death Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Ueli Nachbur
- Cell Signalling and Cell Death Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Maurice Darding
- Centre for Cell Death, Cancer, and Inflammation, University College London, London, United Kingdom
| | - Nieves Peltzer
- Centre for Cell Death, Cancer, and Inflammation, University College London, London, United Kingdom
| | - Najoua Lalaoui
- Cell Signalling and Cell Death Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Kate E Lawlor
- Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Hannah Vanyai
- Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Cathrine Hall
- Cell Signalling and Cell Death Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Aleks Bankovacki
- Cell Signalling and Cell Death Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Lahiru Gangoda
- Department of Biochemistry, La Trobe University, Bundoora, Australia
| | - Wendy Wei-Lynn Wong
- Department of Immunology, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Jason Corbin
- Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Chunzi Huang
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, United States
| | - Edward S Mocarski
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, United States
| | - James M Murphy
- Cell Signalling and Cell Death Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Warren S Alexander
- Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Anne K Voss
- Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - David L Vaux
- Cell Signalling and Cell Death Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - William J Kaiser
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, United States
| | - Henning Walczak
- Centre for Cell Death, Cancer, and Inflammation, University College London, London, United Kingdom
| | - John Silke
- Cell Signalling and Cell Death Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| |
Collapse
|
33
|
Peltzer N, Rieser E, Taraborrelli L, Draber P, Darding M, Pernaute B, Shimizu Y, Sarr A, Draberova H, Montinaro A, Martinez-Barbera JP, Silke J, Rodriguez TA, Walczak H. HOIP deficiency causes embryonic lethality by aberrant TNFR1-mediated endothelial cell death. Cell Rep 2014; 9:153-165. [PMID: 25284787 DOI: 10.1016/j.celrep.2014.08.066] [Citation(s) in RCA: 206] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 07/31/2014] [Accepted: 08/26/2014] [Indexed: 10/25/2022] Open
Abstract
Linear ubiquitination is crucial for innate and adaptive immunity. The linear ubiquitin chain assembly complex (LUBAC), consisting of HOIL-1, HOIP, and SHARPIN, is the only known ubiquitin ligase that generates linear ubiquitin linkages. HOIP is the catalytically active LUBAC component. Here, we show that both constitutive and Tie2-Cre-driven HOIP deletion lead to aberrant endothelial cell death, resulting in defective vascularization and embryonic lethality at midgestation. Ablation of tumor necrosis factor receptor 1 (TNFR1) prevents cell death, vascularization defects, and death at midgestation. HOIP-deficient cells are more sensitive to death induction by both tumor necrosis factor (TNF) and lymphotoxin-α (LT-α), and aberrant complex-II formation is responsible for sensitization to TNFR1-mediated cell death in the absence of HOIP. Finally, we show that HOIP's catalytic activity is necessary for preventing TNF-induced cell death. Hence, LUBAC and its linear-ubiquitin-forming activity are required for maintaining vascular integrity during embryogenesis by preventing TNFR1-mediated endothelial cell death.
Collapse
Affiliation(s)
- Nieves Peltzer
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - Eva Rieser
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - Lucia Taraborrelli
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - Peter Draber
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - Maurice Darding
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - Barbara Pernaute
- British Heart Foundation Centre for Research Excellence, National Heart and Lung Institute (NHLI), Imperial Centre for Translational and Experimental Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
| | - Yutaka Shimizu
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - Aida Sarr
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - Helena Draberova
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - Antonella Montinaro
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - Juan Pedro Martinez-Barbera
- Birth Defects Research Centre, Developmental Biology and Cancer Programme, UCL Institute of Child Health, London WC1N 1EH, UK
| | - John Silke
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia
| | - Tristan A Rodriguez
- British Heart Foundation Centre for Research Excellence, National Heart and Lung Institute (NHLI), Imperial Centre for Translational and Experimental Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
| | - Henning Walczak
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK.
| |
Collapse
|
34
|
Rickard JA, O'Donnell JA, Evans JM, Lalaoui N, Poh AR, Rogers T, Vince JE, Lawlor KE, Ninnis RL, Anderton H, Hall C, Spall SK, Phesse TJ, Abud HE, Cengia LH, Corbin J, Mifsud S, Di Rago L, Metcalf D, Ernst M, Dewson G, Roberts AW, Alexander WS, Murphy JM, Ekert PG, Masters SL, Vaux DL, Croker BA, Gerlic M, Silke J. RIPK1 regulates RIPK3-MLKL-driven systemic inflammation and emergency hematopoiesis. Cell 2014; 157:1175-88. [PMID: 24813849 DOI: 10.1016/j.cell.2014.04.019] [Citation(s) in RCA: 580] [Impact Index Per Article: 52.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 03/28/2014] [Accepted: 04/14/2014] [Indexed: 11/26/2022]
Abstract
Upon ligand binding, RIPK1 is recruited to tumor necrosis factor receptor superfamily (TNFRSF) and Toll-like receptor (TLR) complexes promoting prosurvival and inflammatory signaling. RIPK1 also directly regulates caspase-8-mediated apoptosis or, if caspase-8 activity is blocked, RIPK3-MLKL-dependent necroptosis. We show that C57BL/6 Ripk1(-/-) mice die at birth of systemic inflammation that was not transferable by the hematopoietic compartment. However, Ripk1(-/-) progenitors failed to engraft lethally irradiated hosts properly. Blocking TNF reversed this defect in emergency hematopoiesis but, surprisingly, Tnfr1 deficiency did not prevent inflammation in Ripk1(-/-) neonates. Deletion of Ripk3 or Mlkl, but not Casp8, prevented extracellular release of the necroptotic DAMP, IL-33, and reduced Myd88-dependent inflammation. Reduced inflammation in the Ripk1(-/-)Ripk3(-/-), Ripk1(-/-)Mlkl(-/-), and Ripk1(-/-)Myd88(-/-) mice prevented neonatal lethality, but only Ripk1(-/-)Ripk3(-/-)Casp8(-/-) mice survived past weaning. These results reveal a key function for RIPK1 in inhibiting necroptosis and, thereby, a role in limiting, not only promoting, inflammation.
Collapse
Affiliation(s)
- James A Rickard
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia
| | - Joanne A O'Donnell
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia
| | - Joseph M Evans
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Biochemistry, La Trobe University, Bundoora, VIC 3086, Australia
| | - Najoua Lalaoui
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia
| | - Ashleigh R Poh
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia
| | - TeWhiti Rogers
- Department of Pathology, University of Melbourne, Parkville, VIC 3050, Australia
| | - James E Vince
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia
| | - Kate E Lawlor
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia
| | - Robert L Ninnis
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia
| | - Holly Anderton
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia
| | - Cathrine Hall
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia
| | - Sukhdeep K Spall
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia
| | - Toby J Phesse
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia
| | - Helen E Abud
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
| | - Louise H Cengia
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia
| | - Jason Corbin
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia
| | - Sandra Mifsud
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia
| | - Ladina Di Rago
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia
| | - Donald Metcalf
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia
| | - Matthias Ernst
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia
| | - Grant Dewson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia
| | - Andrew W Roberts
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia; Faculty of Medicine, University of Melbourne, Parkville, VIC 3050, Australia
| | - Warren S Alexander
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia
| | - James M Murphy
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia
| | - Paul G Ekert
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia
| | - Seth L Masters
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia
| | - David L Vaux
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia
| | - Ben A Croker
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia; Division of Hematology and Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Motti Gerlic
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia; Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| | - John Silke
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia.
| |
Collapse
|
35
|
Konrath F, Witt J, Sauter T, Kulms D. Identification of new IκBα complexes by an iterative experimental and mathematical modeling approach. PLoS Comput Biol 2014; 10:e1003528. [PMID: 24675998 PMCID: PMC3967930 DOI: 10.1371/journal.pcbi.1003528] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 02/03/2014] [Indexed: 11/21/2022] Open
Abstract
The transcription factor nuclear factor kappa-B (NFκB) is a key regulator of pro-inflammatory and pro-proliferative processes. Accordingly, uncontrolled NFκB activity may contribute to the development of severe diseases when the regulatory system is impaired. Since NFκB can be triggered by a huge variety of inflammatory, pro-and anti-apoptotic stimuli, its activation underlies a complex and tightly regulated signaling network that also includes multi-layered negative feedback mechanisms. Detailed understanding of this complex signaling network is mandatory to identify sensitive parameters that may serve as targets for therapeutic interventions. While many details about canonical and non-canonical NFκB activation have been investigated, less is known about cellular IκBα pools that may tune the cellular NFκB levels. IκBα has so far exclusively been described to exist in two different forms within the cell: stably bound to NFκB or, very transiently, as unbound protein. We created a detailed mathematical model to quantitatively capture and analyze the time-resolved network behavior. By iterative refinement with numerous biological experiments, we yielded a highly identifiable model with superior predictive power which led to the hypothesis of an NFκB-lacking IκBα complex that contains stabilizing IKK subunits. We provide evidence that other but canonical pathways exist that may affect the cellular IκBα status. This additional IκBα:IKKγ complex revealed may serve as storage for the inhibitor to antagonize undesired NFκB activation under physiological and pathophysiological conditions. In unstimulated cells, the transcription factor NFκB resides in the cytosol bound to its inhibitor IκBα. Canonical activation of NFκB by numerous stimuli leads to proteasomal depletion of IκBα, thereby liberating NFκB to translocate into the nucleus to induce transcription of genes leading to proliferation, angiogenesis, metastasis, or chronic inflammation. Consequently, only transient activity needs to be warranted by immediate NFκB-dependent induction of negative regulatory mechanisms, including up-regulation of its inhibitor IκBα. Resynthesized IκBα consequently terminates NFκB activity by binding to its nuclear localization sequence. However, under physiological or pathophysiological conditions, random NFκB activation may occur, which needs to be avoided in order to guarantee proper cellular function. Using detailed dynamical modeling, we have now identified an additional IκBα containing complex to exist in un-stimulated cells which lacks NFκB but includes IKKγ (IκBα:IKKγ complex). This additional IκBα is not depleted from cells in the canonical fashion and may therefore serve as a cellular backup to avoid random NFκB activation.
Collapse
Affiliation(s)
- Fabian Konrath
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
- Institute for System Dynamics, University of Stuttgart, Stuttgart, Germany
| | - Johannes Witt
- Institute for System Dynamics, University of Stuttgart, Stuttgart, Germany
| | - Thomas Sauter
- Life Sciences Research Unit, University of Luxembourg, Luxembourg, Luxembourg
| | - Dagmar Kulms
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
- Experimental Dermatology, Department of Dermatology, TU-Dresden, Dresden, Germany
- * E-mail:
| |
Collapse
|
36
|
Transcriptomic profiling of differential responses to drought in two freshwater mussel species, the giant floater Pyganodon grandis and the pondhorn Uniomerus tetralasmus. PLoS One 2014; 9:e89481. [PMID: 24586812 PMCID: PMC3934898 DOI: 10.1371/journal.pone.0089481] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 01/22/2014] [Indexed: 11/29/2022] Open
Abstract
The southeastern US has experienced recurrent drought during recent decades. Increasing demand for water, as precipitation decreases, exacerbates stress on the aquatic biota of the Southeast: a global hotspot for freshwater mussel, crayfish, and fish diversity. Freshwater unionid mussels are ideal candidates to study linkages between ecophysiological and behavioral responses to drought. Previous work on co-occurring mussel species suggests a coupling of physiology and behavior along a gradient ranging from intolerant species such as Pyganodon grandis (giant floater) that track receding waters and rarely burrow in the substrates to tolerant species such as Uniomerus tetralasmus (pondhorn) that rarely track receding waters, but readily burrow into the drying sediments. We utilized a next-generation sequencing-based RNA-Seq approach to examine heat/desiccation-induced transcriptomic profiles of these two species in order to identify linkages between patterns of gene expression, physiology and behavior. Sequencing produced over 425 million 100 bp reads. Using the de novo assembly package Trinity, we assembled the short reads into 321,250 contigs from giant floater (average length 835 bp) and 385,735 contigs from pondhorn (average length 929 bp). BLAST-based annotation and gene expression analysis revealed 2,832 differentially expressed genes in giant floater and 2,758 differentially expressed genes in pondhorn. Trancriptomic responses included changes in molecular chaperones, oxidative stress profiles, cell cycling, energy metabolism, immunity, and cytoskeletal rearrangements. Comparative analyses between species indicated significantly higher induction of molecular chaperones and cytoskeletal elements in the intolerant P. grandis as well as important differences in genes regulating apoptosis and immunity.
Collapse
|
37
|
Bu R, Hussain AR, Al-Obaisi KAS, Ahmed M, Uddin S, Al-Kuraya KS. Bortezomib inhibits proteasomal degradation of IκBα and induces mitochondrial dependent apoptosis in activated B-cell diffuse large B-cell lymphoma. Leuk Lymphoma 2014; 55:415-424. [PMID: 23697845 DOI: 10.3109/10428194.2013.806799] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Activated B-cell type lymphoma (ABC), a subgroup of diffuse large B-cell lymphoma (DLBCL), has a worse survival after upfront chemotherapy and is characterized by constitutive activation of the anti-apoptotic nuclear factor-κB (NFκB) pathway. The implication of NFκB inhibition in ABC has not yet been fully explored as a potential therapeutic target. Therefore, a panel of ABC cell lines was used to examine the effect of bortezomib, a proteasome inhibitor which blocks degradation of IκBα and consequently inhibits NFκB activity. Our data showed that bortezomib caused a dose-dependent growth inhibition and induction of apoptosis in all cell lines studied. We next determined the status of the NFκB pathway following bortezomib treatment and found that there was accumulation of IκBα without affecting its phosphorylation status at an early time point. Electrophoretic mobility shift assay showed that bortezomib treatment inhibited constitutive nuclear NFκB in ABC cell lines. Furthermore, treatment of ABC cell lines with bortezomib for 48 h also down-regulated the expression of NFκB-regulated gene products, such as IκBα, Bcl-2, Bcl-Xl, XIAP and survivin, leading to apoptosis via the mitochondrial apoptotic pathway. Altogether, these results suggest that NFκB may be a potential target for therapeutic intervention in DLBCL using proteasomal inhibitors such as bortezomib.
Collapse
Affiliation(s)
- Rong Bu
- Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center , Riyadh , Saudi Arabia
| | | | | | | | | | | |
Collapse
|
38
|
Enesa K, Evans P. The Biology of A20-Like Molecules. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 809:33-48. [DOI: 10.1007/978-1-4939-0398-6_3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
39
|
Abstract
Inhibitor of apoptosis (IAP) proteins interface with, and regulate a large number of, cell signaling pathways. If there is a common theme to these pathways, it is that they are involved in the development of the immune system, immune responses, and unsurprisingly, given their name, cell death. Beyond that it is difficult to discover an underlying logic because sometimes IAPs are required to inhibit or prevent signaling, whereas in other cases they are required for signaling to take place. In whatever role they play, they are recruited into signaling complexes and function as ubiquitin E3 ligases, via their RING domains. This review discusses IAP regulation of signaling pathways and focuses on the mammalian IAPs, XIAP, c-IAP1, and c-IAP2, with a particular emphasis on techniques and methods that were used to uncover their roles. We also provide a perspective on targeting IAP proteins for therapeutic intervention and methods used to define the clinical relevance of IAP proteins.
Collapse
Affiliation(s)
- John Silke
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia.
| | - Domagoj Vucic
- Department of Early Discovery Biochemistry, Genentech, Inc., South San Francisco, California, USA.
| |
Collapse
|
40
|
Hinz M, Scheidereit C. The IκB kinase complex in NF-κB regulation and beyond. EMBO Rep 2013; 15:46-61. [PMID: 24375677 DOI: 10.1002/embr.201337983] [Citation(s) in RCA: 419] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The IκB kinase (IKK) complex is the signal integration hub for NF-κB activation. Composed of two serine-threonine kinases (IKKα and IKKβ) and the regulatory subunit NEMO (also known as IKKγ), the IKK complex integrates signals from all NF-κB activating stimuli to catalyze the phosphorylation of various IκB and NF-κB proteins, as well as of other substrates. Since the discovery of the IKK complex components about 15 years ago, tremendous progress has been made in the understanding of the IKK architecture and its integration into signaling networks. In addition to the control of NF-κB, IKK subunits mediate the crosstalk with other pathways, thereby extending the complexity of their biological function. This review summarizes recent advances in IKK biology and focuses on emerging aspects of IKK structure, regulation and function.
Collapse
Affiliation(s)
- Michael Hinz
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | | |
Collapse
|
41
|
Wu ZH, Shi Y. When ubiquitin meets NF-κB: a trove for anti-cancer drug development. Curr Pharm Des 2013; 19:3263-75. [PMID: 23151140 DOI: 10.2174/1381612811319180010] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 11/01/2012] [Indexed: 02/06/2023]
Abstract
During the last two decades, the studies on ubiquitination in regulating transcription factor NF-κB activation have elucidated the expanding role of ubiquitination in modulating cellular events by non-proteolytic mechanisms, as well as by proteasomal degradation. The significance of ubiquitination has also been recognized in regulating gene transcription, epigenetic modifications, kinase activation, DNA repair and subcellular translocation. This progress has been translated into novel strategies for developing anti-cancer therapeutics, exemplified by the success of the first FDA-approved proteasome inhibitor drug Bortezomib. Here we discuss the current understanding of the ubiquitin-proteasome system and how it is involved in regulating NF-κB signaling pathways in response to a variety of stimuli. We also focus on the recent progress of anti-cancer drug development targeting various steps of ubiquitination process, and the potential of these drugs in cancer treatment as related to their impact on NF-κB activation.
Collapse
Affiliation(s)
- Zhao-Hui Wu
- Department of Pathology and Laboratory Medicine, Center for Adult Cancer Research, University of Tennessee Health Science Center, 19 S. Manassas St., Memphis, TN 38163, USA.
| | | |
Collapse
|
42
|
Chatterton DE, Nguyen DN, Bering SB, Sangild PT. Anti-inflammatory mechanisms of bioactive milk proteins in the intestine of newborns. Int J Biochem Cell Biol 2013; 45:1730-47. [DOI: 10.1016/j.biocel.2013.04.028] [Citation(s) in RCA: 251] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 04/25/2013] [Accepted: 04/28/2013] [Indexed: 01/04/2023]
|
43
|
RING-type E3 ligases: master manipulators of E2 ubiquitin-conjugating enzymes and ubiquitination. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1843:47-60. [PMID: 23747565 DOI: 10.1016/j.bbamcr.2013.05.026] [Citation(s) in RCA: 447] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 05/23/2013] [Accepted: 05/29/2013] [Indexed: 01/02/2023]
Abstract
RING finger domain and RING finger-like ubiquitin ligases (E3s), such as U-box proteins, constitute the vast majority of known E3s. RING-type E3s function together with ubiquitin-conjugating enzymes (E2s) to mediate ubiquitination and are implicated in numerous cellular processes. In part because of their importance in human physiology and disease, these proteins and their cellular functions represent an intense area of study. Here we review recent advances in RING-type E3 recognition of substrates, their cellular regulation, and their varied architecture. Additionally, recent structural insights into RING-type E3 function, with a focus on important interactions with E2s and ubiquitin, are reviewed. This article is part of a Special Issue entitled: Ubiquitin-Proteasome System. Guest Editors: Thomas Sommer and Dieter H. Wolf.
Collapse
|
44
|
Two coordinated mechanisms underlie tumor necrosis factor alpha-induced immediate and delayed IκB kinase activation. Mol Cell Biol 2013; 33:1901-15. [PMID: 23459942 DOI: 10.1128/mcb.01416-12] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Tumor necrosis factor alpha (TNF-α)-induced NF-κB activation has been believed to depend on TRAF2- and cIAP1-mediated RIP1 ubiquitination. However, recent findings have challenged the notion that these proteins play essential roles in NF-κB activation. Here, by assessing the kinetics and amplitude of IκB kinase (IKK) activation, we report that TNF-α-induced immediate and robust activation of IKK requires K63-linked and linearly linked ubiquitination of RIP1 and that in the absence of RIP1 expression, TRAF2 and cIAP1 cooperatively induce delayed IKK activation by recruiting LUBAC to TNFR1. Knockdown of HOIP (a component of LUBAC) in RIP1-deficient cells completely impairs the recruitment and activation of IKK but does not affect K63-linked ubiquitination of TRAF2 and recruitment of TAK1 to TNFR1, suggesting that the K63-linked ubiquitin chain is not capable of recruiting IKK in vivo. We also demonstrate that TRAF2 and cIAP1 together, but not either one alone, directly catalyze linearly linked ubiquitination of RIP1. Importantly, in embryonic hepatocytes, TNF-α activates NF-κB through a RIP1-independent pathway. Thus, our findings clarify molecular details of this important signaling mechanism by providing evidence for the existence of two phases of IKK activation: the immediate phase, induced by TRAF2/cIAP1-mediated ubiquitination of RIP1, and the delayed phase, activated by TRAF2/cIAP1-dependent recruitment of LUBAC.
Collapse
|
45
|
Silke J, Meier P. Inhibitor of apoptosis (IAP) proteins-modulators of cell death and inflammation. Cold Spring Harb Perspect Biol 2013; 5:5/2/a008730. [PMID: 23378585 DOI: 10.1101/cshperspect.a008730] [Citation(s) in RCA: 229] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Misregulated innate immune signaling and cell death form the basis of much human disease pathogenesis. Inhibitor of apoptosis (IAP) protein family members are frequently overexpressed in cancer and contribute to tumor cell survival, chemo-resistance, disease progression, and poor prognosis. Although best known for their ability to regulate caspases, IAPs also influence ubiquitin (Ub)-dependent pathways that modulate innate immune signaling via activation of nuclear factor κB (NF-κB). Recent research into IAP biology has unearthed unexpected roles for this group of proteins. In addition, the advances in our understanding of the molecular mechanisms that IAPs use to regulate cell death and innate immune responses have provided new insights into disease states and suggested novel intervention strategies. Here we review the functions assigned to those IAP proteins that act at the intersection of cell death regulation and inflammatory signaling.
Collapse
Affiliation(s)
- John Silke
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia.
| | | |
Collapse
|
46
|
The role of the IAP E3 ubiquitin ligases in regulating pattern-recognition receptor signalling. Nat Rev Immunol 2012; 12:833-44. [PMID: 23124073 DOI: 10.1038/nri3325] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
An inflammatory response is initiated when innate immune pattern-recognition receptors (PRRs) expressed by different cell types detect constituents of invading microorganisms and endogenous intracellular molecules released by dying cells. The intracellular cascades activated by PRRs induce the expression and maturation of inflammatory molecules that coordinate the removal of the infectious agents and of the infected or damaged cells. In this Review, we discuss the findings implicating members of the inhibitor of apoptosis protein (IAP) family in the ubiquitylation-dependent regulation of PRR signalling. Understanding the role of IAPs in innate immunity may open new therapeutic perspectives for the treatment of PRR-dependent inflammatory diseases.
Collapse
|
47
|
Stravodimou A, Mazzoccoli G, Voutsadakis IA. Peroxisome proliferator-activated receptor gamma and regulations by the ubiquitin-proteasome system in pancreatic cancer. PPAR Res 2012; 2012:367450. [PMID: 23049538 PMCID: PMC3459232 DOI: 10.1155/2012/367450] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Accepted: 08/13/2012] [Indexed: 12/11/2022] Open
Abstract
Pancreatic cancer is one of the most lethal forms of human cancer. Although progress in oncology has improved outcomes in many forms of cancer, little progress has been made in pancreatic carcinoma and the prognosis of this malignancy remains grim. Several molecular abnormalities often present in pancreatic cancer have been defined and include mutations in K-ras, p53, p16, and DPC4 genes. Nuclear receptor Peroxisome Proliferator-Activated Receptor gamma (PPARγ) has a role in many carcinomas and has been found to be overexpressed in pancreatic cancer. It plays generally a tumor suppressor role antagonizing proteins promoting carcinogenesis such as NF-κB and TGFβ. Regulation of pathways involved in pancreatic carcinogenesis is effectuated by the Ubiquitin Proteasome System (UPS). This paper will examine PPARγ in pancreatic cancer, the regulation of this nuclear receptor by the UPS, and their relationship to other pathways important in pancreatic carcinogenesis.
Collapse
Affiliation(s)
- Athina Stravodimou
- Centre Pluridisciplinaire d'Oncologie, Centre Hospitalier Universitaire Vaudois, BH06, Bugnon 46, 1011 Lausanne, Switzerland
| | - Gianluigi Mazzoccoli
- Division of Internal Medicine and Chronobiology Unit, Department of Medical Sciences, IRCCS Scientific Institute and Regional General Hospital “Casa Sollievo della Sofferenza”, San Giovanni Rotondo, Italy
| | - Ioannis A. Voutsadakis
- Centre Pluridisciplinaire d'Oncologie, Centre Hospitalier Universitaire Vaudois, BH06, Bugnon 46, 1011 Lausanne, Switzerland
| |
Collapse
|
48
|
König HG, Fenner BJ, Byrne JC, Schwamborn RF, Bernas T, Jefferies CA, Prehn JHM. Fibroblast growth factor homologous factor 1 interacts with NEMO to regulate NF-κB signaling in neurons. J Cell Sci 2012; 125:6058-70. [DOI: 10.1242/jcs.111880] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Neuronal survival and plasticity critically depend on constitutive activity of the transcription factor nuclear factor-κB (NF-κB). We here describe a role for a small intracellular fibroblast growth factor homologue, the fibroblast growth factor homologous factor 1 (FHF1/FGF12) in the regulation of NF-κB activity in mature neurons. FHF's have previously been described to control neuronal excitability, and mutations in FHF isoforms give rise to a form of progressive spinocerebellar ataxia. Using a protein-array approach, we identified FHF1b as a novel interactor of the canonical NF–κB modulator IKKγ/NEMO. Co-immunoprecipitation, pull-down and GAL4-reporter experiments, as well as proximity ligation assays confirmed the interaction of FHF1 and NEMO, and demonstrated that a major site of interaction occurred within the axon initial segment. Fhf1 gene silencing strongly activated neuronal NF-κB activity and increased neurite lengths, branching patterns and spine counts in mature cortical neurons. The effects of FHF1 on neuronal NF-κB activity and morphology required the presence of NEMO. Our results imply that FHF1 negatively regulates the constitutive NF-κB activity in neurons.
Collapse
|