1
|
Daniels MA, Teixeiro E. The NF-κB signaling network in the life of T cells. Front Immunol 2025; 16:1559494. [PMID: 40370445 PMCID: PMC12075310 DOI: 10.3389/fimmu.2025.1559494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 04/07/2025] [Indexed: 05/16/2025] Open
Abstract
NF-κB is a crucial transcription factor in lymphocyte signaling. It is activated by environmental cues that drive lymphocyte differentiation to combat infections and cancer. As a key player in inflammation, NF-κB also significantly impacts autoimmunity and transplant rejection, making it an important therapeutic target. While the signaling molecules regulating this pathway are well-studied, the effect of changes in NF-κB signaling levels on T lymphocyte differentiation, fate, and function is not fully understood. Advances in computational biology and new NF-κB-inducible animal models are beginning to clarify these questions. In this review, we highlight recent findings related to T cells, focusing on how environmental cues affecting NF-κB signaling levels determine T cell fate and function.
Collapse
Affiliation(s)
- Mark A. Daniels
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, United States
- Roy Blunt NextGen Precision Health Building, University of Missouri, Columbia, MO, United States
| | - Emma Teixeiro
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, United States
- Roy Blunt NextGen Precision Health Building, University of Missouri, Columbia, MO, United States
| |
Collapse
|
2
|
Sarkar S, Kobayashi Y, Russnak T, Shen J, Nahass RG, Dougherty JP, Gélinas C. GS143, an inhibitor of E3 ligase β-TrCP, reverses HIV-1 latency without activating T cells via unconventional activation of NFκB. PLoS Pathog 2025; 21:e1013018. [PMID: 40168443 PMCID: PMC11999137 DOI: 10.1371/journal.ppat.1013018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 04/15/2025] [Accepted: 03/03/2025] [Indexed: 04/03/2025] Open
Abstract
HIV-1 persists indefinitely in individuals living with HIV-1 even after effective treatment with antiretroviral therapy (ART). Upon cessation of the therapy, latently infected memory CD4+ T cells allow for a rapid rebound of the virus. The development of latency reversing agents (LRAs) to activate latent virus promoting immune recognition and clearance of the infected cells is pivotal for the elimination of the latent arm of the infection. Success of this strategy requires the development of potent highly specific LRAs with fewer off-target effects. LRA activity displayed by proteasome inhibitors although not highly specific opens the possibility of exploiting the high degree of specificity of the ubiquitin-proteasome system to develop targeted LRAs. Here we demonstrate that a small molecule GS143, which inhibits β-TrCP, the substrate recognition subunit of the SCFβ-TrCP E3 ubiquitin protein ligases, exhibits potent LRA activity both in a primary cell model system of latency and cells from aviremic individuals with HIV-1 treated with ART. Furthermore, GS143 reactivates latent HIV-1 without activating T cells, a desirable attribute for LRAs of clinical use. We showed that GS143 acts in a complementary fashion with at least two other classes of LRAs, thereby representing novel drug combinations for targeting HIV-1 latency. Finally, our results suggest that GS143 triggers a novel signaling pathway to reactivate latent HIV-1 that leads to the unconventional activation of NFκB p65, by initiating the noncanonical signaling via NIK, followed by activation of IKK leading to phosphorylation of p65 on S536 and its nuclear translocation. Moreover, we show that β-catenin inhibitors suppress reactivation HIV-1 by GS143, suggesting that β-catenin supports NF-κB output indirectly. Overall, our results suggest that the β-TrCP E3 ligase inhibitor GS143 represents a new type of LRA.
Collapse
Affiliation(s)
- Srijata Sarkar
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, New Jersey, United States of America
| | - Yoshifumi Kobayashi
- Department of Pharmacology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, United States of America
| | - Timothy Russnak
- Graduate Program in Molecular Genetics and Microbiology, Rutgers School of Graduate Studies, Piscataway, New Jersey, United States of America
| | - Jing Shen
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, New Jersey, United States of America
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, United States of America
| | - Ronald G. Nahass
- IDCare, Hillsborough, New Jersey, United States of America
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, United States of America
| | - Joseph P. Dougherty
- Department of Pharmacology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, United States of America
| | - Céline Gélinas
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, New Jersey, United States of America
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, United States of America
| |
Collapse
|
3
|
Yang Y, Huang B, Qin Y, Wang D, Jin Y, Su L, Wang Q, Pan Y, Zhang Y, Shen Y, Hu W, Cao Z, Jin L, Zhang F. Adipocyte microRNA-802 promotes adipose tissue inflammation and insulin resistance by modulating macrophages in obesity. eLife 2024; 13:e99162. [PMID: 39589393 DOI: 10.7554/elife.99162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 11/22/2024] [Indexed: 11/27/2024] Open
Abstract
Adipose tissue inflammation is now considered to be a key process underlying metabolic diseases in obese individuals. However, it remains unclear how adipose inflammation is initiated and maintained or the mechanism by which inflammation develops. We found that microRNA-802 (Mir802) expression in adipose tissue is progressively increased with the development of dietary obesity in obese mice and humans. The increasing trend of Mir802 preceded the accumulation of macrophages. Adipose tissue-specific knockout of Mir802 lowered macrophage infiltration and ameliorated systemic insulin resistance. Conversely, the specific overexpression of Mir802 in adipose tissue aggravated adipose inflammation in mice fed a high-fat diet. Mechanistically, Mir802 activates noncanonical and canonical NF-κB pathways by targeting its negative regulator, TRAF3. Next, NF-κB orchestrated the expression of chemokines and SREBP1, leading to strong recruitment and M1-like polarization of macrophages. Our findings indicate that Mir802 endows adipose tissue with the ability to recruit and polarize macrophages, which underscores Mir802 as an innovative and attractive candidate for miRNA-based immune therapy for adipose inflammation.
Collapse
Affiliation(s)
- Yue Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Bin Huang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yimeng Qin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Danwei Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yinuo Jin
- NanJing HanKai Academy, Nanjing, China
| | - Linmin Su
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Qingxin Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yi Pan
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yanfeng Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yumeng Shen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Wenjun Hu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Zhengyu Cao
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Liang Jin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Fangfang Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
4
|
Zhao L, Li Z. LncRNA DANCR suppresses acute myocardial infarction in mice via mediating p-RXRA/TRAF2/NIK/IKK/NF-κB signaling pathway. Aging (Albany NY) 2024; 16:13356-13370. [PMID: 39546553 PMCID: PMC11719107 DOI: 10.18632/aging.206150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024]
Abstract
OBJECTIVES This study aimed to investigate the role of LncRNA differentiation antagonizing non-protein coding RNA (DANCR) in acute myocardial infarction (AMI). METHODS A mouse model of AMI was established, and the cardiac contractile function was detected. Next, cardiomyocytes treated with oxygen-glucose deprivation (OGD) were used for gain- and loss-function experiments in vitro. RIP analysis was used to verify the binding of DANCR and Retinoid X receptor alpha (RXRA), and Co-IP assay was used to measure the binding of phosphorylated RXRA to TNF receptor associated factor 2 (TRAF2). RESULTS The expression of DANCR in myocardial tissues of AMI mice were significantly downregulated. Overexpression of DANCR decreased myocardial infarct size and enhanced cardiac contractile function in AMI mice. Moreover, overexpression of DANCR promoted proliferation and inhibited apoptosis in OGD-induced cardiomyocytes. Mechanism studies demonstrated that DANCR interacted with RXRA and promoted glycogen synthase kinase 3beta (GSK3β)-mediated phosphorylation of RXRA, and phosphorylated RXRA interacted with TRAF2 protein to downregulate TRAF2 protein level. Bexarotene (Bex), an activator of RXRA, inhibited TRAF2 protein expression, while RXRA overexpression had no effect on TRAF2 protein expression. Bex treatment or silencing TRAF2 promoted proliferation and inhibited apoptosis in cardiomyocytes. In addition, silencing DANCR inhibited cardiomyocyte proliferation and induced apoptosis by activating the NIK/IKK/NF-κB pathway, while B022, an inhibitor of NIK, counteracted the effects of DANCR silencing on cardiomyocytes. CONCLUSIONS Studies demonstrated that DANCR suppressed AMI in mice by mediating p-RXRA/TRAF2/NIK/IKK/NF-κB pathway.
Collapse
Affiliation(s)
- Li Zhao
- Department of Cardiovascular, Affiliated Hospital of Yanan University, Yanan, China
| | - Zhi Li
- Department of Cardiovascular, Affiliated Hospital of Yanan University, Yanan, China
| |
Collapse
|
5
|
Varner LR, Chaya T, Maeda Y, Tsutsumi R, Zhou S, Tsujii T, Okuzaki D, Furukawa T. The deubiquitinase Otud7b suppresses cone photoreceptor degeneration in mouse models of retinal degenerative diseases. iScience 2024; 27:109380. [PMID: 38510130 PMCID: PMC10951987 DOI: 10.1016/j.isci.2024.109380] [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: 09/05/2023] [Revised: 01/15/2024] [Accepted: 02/27/2024] [Indexed: 03/22/2024] Open
Abstract
Primary and secondary cone photoreceptor death in retinal degenerative diseases, including age-related macular degeneration (AMD) and retinitis pigmentosa (RP), leads to severe visual impairment and blindness. Although the cone photoreceptor protection in retinal degenerative diseases is crucial for maintaining vision, the underlying molecular mechanisms are unclear. Here, we found that the deubiquitinase Otud7b/Cezanne is predominantly expressed in photoreceptor cells in the retina. We analyzed Otud7b-/- mice, which were subjected to light-induced damage, a dry AMD model, or were mated with an RP mouse model, and observed increased cone photoreceptor degeneration. Using RNA-sequencing and bioinformatics analysis followed by a luciferase reporter assay, we found that Otud7b downregulates NF-κB activity. Furthermore, inhibition of NF-κB attenuated cone photoreceptor degeneration in the light-exposed Otud7b-/- retina and stress-induced neuronal cell death resulting from Otud7b deficiency. Together, our findings suggest that Otud7b protects cone photoreceptors in retinal degenerative diseases by modulating NF-κB activity.
Collapse
Affiliation(s)
- Leah Rie Varner
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
| | - Taro Chaya
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
| | - Yamato Maeda
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
| | - Ryotaro Tsutsumi
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
| | - Shanshan Zhou
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
| | - Toshinori Tsujii
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Takahisa Furukawa
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
6
|
Li Y, Liu X, Fujinaga K, Gross JD, Frankel AD. Enhanced NF-κB activation via HIV-1 Tat-TRAF6 cross-talk. SCIENCE ADVANCES 2024; 10:eadi4162. [PMID: 38241362 PMCID: PMC10798561 DOI: 10.1126/sciadv.adi4162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 12/21/2023] [Indexed: 01/21/2024]
Abstract
The Tat proteins of HIV-1 and simian immunodeficiency virus (SIV) are essential for activating viral transcription. In addition, Tat stimulates nuclear factor κB (NF-κB) signaling pathways to regulate viral gene expression although its molecular mechanism is unclear. Here, we report that Tat directly activates NF-κB through the interaction with TRAF6, which is an essential upstream signaling molecule of the canonical NF-κB pathway. This interaction increases TRAF6 oligomerization and auto-ubiquitination, as well as the synthesis of K63-linked polyubiquitin chains to further activate the NF-κB pathway and HIV-1 transcription. Moreover, ectopic expression of TRAF6 significantly activates HIV-1 transcription, whereas TRAF6 knockdown inhibits transcription. Furthermore, Tat-mediated activation of NF-κB through TRAF6 is conserved among HIV-1, HIV-2, and SIV isolates. Our study uncovers yet another mechanism by which HIV-1 subverts host transcriptional pathways to enhance its own transcription.
Collapse
Affiliation(s)
- Yang Li
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
| | - Xi Liu
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158, USA
| | - Koh Fujinaga
- Department of Medicine, University of California, San Francisco, CA 94143, USA
| | - John D. Gross
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158, USA
| | - Alan D. Frankel
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
| |
Collapse
|
7
|
Mukherjee T, Kumar N, Chawla M, Philpott DJ, Basak S. The NF-κB signaling system in the immunopathogenesis of inflammatory bowel disease. Sci Signal 2024; 17:eadh1641. [PMID: 38194476 DOI: 10.1126/scisignal.adh1641] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 12/11/2023] [Indexed: 01/11/2024]
Abstract
Inflammatory bowel disease (IBD) is an idiopathic, chronic condition characterized by episodes of inflammation in the gastrointestinal tract. The nuclear factor κB (NF-κB) system describes a family of dimeric transcription factors. Canonical NF-κB signaling is stimulated by and enhances inflammation, whereas noncanonical NF-κB signaling contributes to immune organogenesis. Dysregulation of NF-κB factors drives various inflammatory pathologies, including IBD. Signals from many immune sensors activate NF-κB subunits in the intestine, which maintain an equilibrium between local microbiota and host responses. Genetic association studies of patients with IBD and preclinical mouse models confirm the importance of the NF-κB system in host defense in the gut. Other studies have investigated the roles of these factors in intestinal barrier function and in inflammatory gut pathologies associated with IBD. NF-κB signaling modulates innate and adaptive immune responses and the production of immunoregulatory proteins, anti-inflammatory cytokines, antimicrobial peptides, and other tolerogenic factors in the intestine. Furthermore, genetic studies have revealed critical cell type-specific roles for NF-κB proteins in intestinal immune homeostasis, inflammation, and restitution that contribute to the etiopathology of IBD-associated manifestations. Here, we summarize our knowledge of the roles of these NF-κB pathways, which are activated in different intestinal cell types by specific ligands, and their cross-talk, in fueling aberrant intestinal inflammation. We argue that an in-depth understanding of aberrant immune signaling mechanisms may hold the key to identifying predictive or prognostic biomarkers and developing better therapeutics against inflammatory gut pathologies.
Collapse
Affiliation(s)
- Tapas Mukherjee
- Systems Immunology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Naveen Kumar
- Systems Immunology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Meenakshi Chawla
- Systems Immunology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Soumen Basak
- Systems Immunology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| |
Collapse
|
8
|
Cormier F, Housni S, Dumont F, Villard M, Cochand-Priollet B, Mercier-Nomé F, Perlemoine K, Bertherat J, Groussin L. NF-κB signaling activation and roles in thyroid cancers: implication of MAP3K14/NIK. Oncogenesis 2023; 12:55. [PMID: 37973791 PMCID: PMC10654696 DOI: 10.1038/s41389-023-00496-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 10/11/2023] [Accepted: 10/20/2023] [Indexed: 11/19/2023] Open
Abstract
Among follicular-derived thyroid cancers (TC), those with aggressive behavior and resistance to current treatments display poor prognosis. NF-κB signaling pathways are involved in tumor progression of various cancers. Here, we finely characterize the NF-κB pathways and their involvement in TC. By using immunoblot and gel shift assays, we demonstrated that both classical and alternative NF-κB pathways are activated in ten TC-derived cell lines, leading to activated RelA/p50 and RelB/p50 NF-κB dimers. By analyzing the RNAseq data of the large papillary thyroid carcinoma (PTC) cohort from The Cancer Genome Atlas (TCGA) project, we identified a tumor progression-related NF-κB signature in BRAFV600E mutated-PTCs. That corroborated with the role of RelA and RelB in cell migration and invasion processes that we demonstrated specifically in BRAFV600E mutated-cell lines, together with their role in the control of expression of genes implicated in invasiveness (MMP1, PLAU, LCN2 and LGALS3). We also identified NF-κB-inducing kinase (NIK) as a novel actor of the constitutive activation of the NF-κB pathways in TC-derived cell lines. Finally, its implication in invasiveness and its overexpression in PTC samples make NIK a potential therapeutic target for advanced TC treatment.
Collapse
Affiliation(s)
- Françoise Cormier
- Université Paris Cité, INSERM U1016, CNRS UMR8104, Institut Cochin, F-75014, Paris, France.
| | - Selma Housni
- Université Paris Cité, INSERM U1016, CNRS UMR8104, Institut Cochin, F-75014, Paris, France
- Service de Médecine Nucléaire, Assistance Publique-Hopitaux de Paris, Hopital Pitié-Salpêtrière, F-75013, Paris, France
| | - Florent Dumont
- Université Paris Cité, INSERM U1016, CNRS UMR8104, Institut Cochin, F-75014, Paris, France
- UMS IPSIT, Université Paris-Saclay, INSERM, CNRS, F-91400, Orsay, France
| | - Mélodie Villard
- Université Paris Cité, INSERM U1016, CNRS UMR8104, Institut Cochin, F-75014, Paris, France
| | - Béatrix Cochand-Priollet
- Service de Pathologie, Assistance Publique-Hopitaux de Paris, Hopital Cochin, Université Paris Cité, F-75014, Paris, France
| | | | - Karine Perlemoine
- Université Paris Cité, INSERM U1016, CNRS UMR8104, Institut Cochin, F-75014, Paris, France
| | - Jérôme Bertherat
- Université Paris Cité, INSERM U1016, CNRS UMR8104, Institut Cochin, F-75014, Paris, France
- Service d'Endocrinologie, Cochin AP-HP Centre, F-75014, Paris, France
| | - Lionel Groussin
- Université Paris Cité, INSERM U1016, CNRS UMR8104, Institut Cochin, F-75014, Paris, France
- Service d'Endocrinologie, Cochin AP-HP Centre, F-75014, Paris, France
| |
Collapse
|
9
|
Song EZ, Wang X, Philipson BI, Zhang Q, Thokala R, Zhang L, Assenmacher CA, Binder ZA, Ming GL, O’Rourke DM, Song H, Milone MC. The IAP antagonist birinapant enhances chimeric antigen receptor T cell therapy for glioblastoma by overcoming antigen heterogeneity. Mol Ther Oncolytics 2022; 27:288-304. [PMID: 36458202 PMCID: PMC9707011 DOI: 10.1016/j.omto.2022.11.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 11/14/2022] [Indexed: 11/17/2022] Open
Abstract
Antigen heterogeneity that results in tumor antigenic escape is one of the major obstacles to successful chimeric antigen receptor (CAR) T cell therapies in solid tumors including glioblastoma multiforme (GBM). To address this issue and improve the efficacy of CAR T cell therapy for GBM, we developed an approach that combines CAR T cells with inhibitor of apoptosis protein (IAP) antagonists, a new class of small molecules that mediate the degradation of IAPs, to treat GBM. Here, we demonstrated that the IAP antagonist birinapant could sensitize GBM cell lines and patient-derived primary GBM organoids to apoptosis induced by CAR T cell-derived cytokines, such as tumor necrosis factor. Therefore, birinapant could enhance CAR T cell-mediated bystander death of antigen-negative GBM cells, thus preventing tumor antigenic escape in antigen-heterogeneous tumor models in vitro and in vivo. In addition, birinapant could promote the activation of NF-κB signaling pathways in antigen-stimulated CAR T cells, and with a birinapant-resistant tumor model we showed that birinapant had no deleterious effect on CAR T cell functions in vitro and in vivo. Overall, we demonstrated the potential of combining the IAP antagonist birinapant with CAR T cells as a novel and feasible approach to overcoming tumor antigen heterogeneity and enhancing CAR T cell therapy for GBM.
Collapse
Affiliation(s)
- Edward Z. Song
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
| | - Xin Wang
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Benjamin I. Philipson
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
| | - Qian Zhang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
| | - Radhika Thokala
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
| | - Logan Zhang
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Charles-Antoine Assenmacher
- Comparative Pathology Core, Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zev A. Binder
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Guo-li Ming
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Donald M. O’Rourke
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hongjun Song
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael C. Milone
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
| |
Collapse
|
10
|
Stavrou EF, Chatzopoulou F, Antonatos C, Pappa P, Makridou E, Oikonomou K, Kapsoritakis A, Potamianos PS, Karmiris K, Tzathas C, Chatzidimitriou D, Vizirianakis IS, Vasilopoulos Y. Pharmacogenetic analysis of canonical versus noncanonical pathway of NF-kB in Crohn's disease patients under anti-tumor necrosis factor-α treatment. Pharmacogenet Genomics 2022; 32:235-241. [PMID: 35852914 DOI: 10.1097/fpc.0000000000000471] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
OBJECTIVES This study explores the potential of gene polymorphisms in the canonical and noncanonical NF-kB signaling pathway as a prediction biomarker of anti-tumor necrosis factor (TNF)α response in Crohn's patients. MATERIALS AND METHODS A total of 109 Greek patients with Crohn's disease (CD) were recruited, and the genotype of TLR2 rs3804099, LTA rs909253, TLR4 rs5030728, and MAP3K14/NIK rs7222094 single nucleotide polymorphisms was investigated for association with response to anti-TNFα therapy. Patient's response to therapy was based on the Crohn's Disease Activity Index, depicting the maximum response within 24 months after initiation of treatment. RESULTS Seventy-three patients (66.7%) were classified as responders while 36 as nonresponders (33.3%). Comparing allelic frequencies between responders and nonresponders, the presence of TLR2 rs3804099 T allele was associated with nonresponse (P = 0.003), even after stratification by anti-TNFα drugs (infliximab: P = 0.032, adalimumab: P = 0.026). No other association was identified for the rest of the polymorphisms under study. Haplotype analysis further enhanced the association of rs3804099 T allele with loss of response, even though the results were NS (P = 0.073). CONCLUSION Our results suggest that polymorphisms in the canonical NF-kB pathway genes could potentially act as a predictive biomarker of anti-TNFα response in CD.
Collapse
Affiliation(s)
- Eleana F Stavrou
- Laboratory of Genetics, Department of Biology, University of Patras, Patras
| | - Fani Chatzopoulou
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki
- Labnet Laboratories, Department of Molecular Biology and Genetics
- Laboratory of Microbiology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki
| | | | - Panagiota Pappa
- Laboratory of Genetics, Department of Biology, University of Patras, Patras
| | - Eutychia Makridou
- Laboratory of Genetics, Department of Biology, University of Patras, Patras
| | | | | | | | - Konstantinos Karmiris
- Gastroenterology Department, "Venizeleio Pananeio" General Hospital of Heraklion, Crete
| | - Charalambos Tzathas
- Gastroenterology Department, "Tzaneio" General Hospital of Piraeus, Piraeus, Greece
| | - Dimitris Chatzidimitriou
- Laboratory of Microbiology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki
| | - Ioannis S Vizirianakis
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki
- Department of Life and Health Sciences, University of Nicosia, Nicosia, Cyprus
| | | |
Collapse
|
11
|
Haselager MV, Eldering E. The Therapeutic Potential of Targeting NIK in B Cell Malignancies. Front Immunol 2022; 13:930986. [PMID: 35911754 PMCID: PMC9326486 DOI: 10.3389/fimmu.2022.930986] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/08/2022] [Indexed: 11/24/2022] Open
Abstract
NF-κB-inducing kinase (NIK) is a key player in non-canonical NF-κB signaling, involved in several fundamental cellular processes, and is crucial for B cell function and development. In response to certain signals and ligands, such as CD40, BAFF and lymphotoxin-β activation, NIK protein stabilization and subsequent NF-κB activation is achieved. Overexpression or overactivation of NIK is associated with several malignancies, including activating mutations in multiple myeloma (MM) and gain-of-function in MALT lymphoma as a result of post-translational modifications. Consequently, drug discovery studies are devoted to pharmacologic modulation of NIK and development of specific novel small molecule inhibitors. However, disease-specific in vitro and in vivo studies investigating NIK inhibition are as of yet lacking, and clinical trials with NIK inhibitors remain to be initiated. In order to bridge the gap between bench and bedside, this review first briefly summarizes our current knowledge on NIK activation, functional activity and stability. Secondly, we compare current inhibitors targeting NIK based on efficacy and specificity, and provide a future perspective on the therapeutic potential of NIK inhibition in B cell malignancies.
Collapse
Affiliation(s)
- Marco V. Haselager
- Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam, Netherlands
- Lymphoma and Myeloma Center Amsterdam, Lymphoma and Myeloma Center Amsterdam, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Immunology, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, Netherlands
| | - Eric Eldering
- Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam, Netherlands
- Lymphoma and Myeloma Center Amsterdam, Lymphoma and Myeloma Center Amsterdam, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Immunology, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, Netherlands
- *Correspondence: Eric Eldering,
| |
Collapse
|
12
|
Han X, Ren J, Lohner H, Yakoumatos L, Liang R, Wang H. SGK1 negatively regulates inflammatory immune responses and protects against alveolar bone loss through modulation of TRAF3 activity. J Biol Chem 2022; 298:102036. [PMID: 35588785 PMCID: PMC9190018 DOI: 10.1016/j.jbc.2022.102036] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/09/2022] [Accepted: 05/10/2022] [Indexed: 12/05/2022] Open
Abstract
Serum- and glucocorticoid-regulated kinase 1 (SGK1) is a serine/threonine kinase that plays important roles in the cellular stress response. While SGK1 has been reported to restrain inflammatory immune responses, the molecular mechanisms involved remain elusive, especially in oral bacteria-induced inflammatory milieu. Here, we found that SGK1 curtails Porphyromonas gingivalis-induced inflammatory responses through maintaining levels of tumor necrosis factor receptor-associated factor (TRAF) 3, thereby suppressing NF-κB signaling. Specifically, SGK1 inhibition significantly enhances production of proinflammatory cytokines, including tumor necrosis factor α, interleukin (IL)-6, IL-1β, and IL-8 in P. gingivalis-stimulated innate immune cells. The results were confirmed with siRNA and LysM-Cre-mediated SGK1 KO mice. Moreover, SGK1 deletion robustly increased NF-κB activity and c-Jun expression but failed to alter the activation of mitogen-activated protein kinase signaling pathways. Further mechanistic data revealed that SGK1 deletion elevates TRAF2 phosphorylation, leading to TRAF3 degradation in a proteasome-dependent manner. Importantly, siRNA-mediated traf3 silencing or c-Jun overexpression mimics the effect of SGK1 inhibition on P. gingivalis-induced inflammatory cytokines and NF-κB activation. In addition, using a P. gingivalis infection-induced periodontal bone loss model, we found that SGK1 inhibition modulates TRAF3 and c-Jun expression, aggravates inflammatory responses in gingival tissues, and exacerbates alveolar bone loss. Altogether, we demonstrated for the first time that SGK1 acts as a rheostat to limit P. gingivalis-induced inflammatory immune responses and mapped out a novel SGK1-TRAF2/3-c-Jun-NF-κB signaling axis. These findings provide novel insights into the anti-inflammatory molecular mechanisms of SGK1 and suggest novel interventional targets to inflammatory diseases relevant beyond the oral cavity.
Collapse
Affiliation(s)
- Xiao Han
- Department of Oral and Craniofacial Molecular Biology, VCU Philips Institute for Oral Health Research, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Junling Ren
- Department of Oral and Craniofacial Molecular Biology, VCU Philips Institute for Oral Health Research, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Hannah Lohner
- Department of Oral and Craniofacial Molecular Biology, VCU Philips Institute for Oral Health Research, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Lan Yakoumatos
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, Kentucky, USA
| | - Ruqiang Liang
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, California, USA
| | - Huizhi Wang
- Department of Oral and Craniofacial Molecular Biology, VCU Philips Institute for Oral Health Research, Virginia Commonwealth University, Richmond, Virginia, USA.
| |
Collapse
|
13
|
Xiao P, Takiishi T, Violato NM, Licata G, Dotta F, Sebastiani G, Marselli L, Singh SP, Sze M, Van Loo G, Dejardin E, Gurzov EN, Cardozo AK. NF-κB-inducing kinase (NIK) is activated in pancreatic β-cells but does not contribute to the development of diabetes. Cell Death Dis 2022; 13:476. [PMID: 35589698 PMCID: PMC9120028 DOI: 10.1038/s41419-022-04931-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/04/2022] [Accepted: 05/09/2022] [Indexed: 12/14/2022]
Abstract
The transcription factor nuclear factor-κB (NF-κB) has a key role in the pathogenesis of diabetes and its complications. Although activation of the canonical NF-κB pathway in β-cells is generally deleterious, little is known about the role of the non-canonical NF-κB signalling and its main regulator, the NF-κB-inducing kinase (NIK), on pancreatic β-cell survival and function. Previous studies based on models of NIK overexpression in pancreatic islet cells showed that NIK induced either spontaneous β-cell death due to islet inflammation or glucose intolerance during diet-induced obesity (DIO) in mice. Therefore, NIK has been proposed as a potential target for diabetes therapy. However, no clear studies showed whether inhibition of NIK improves diabetes development. Here we show that genetic silencing of NIK in pancreatic β-cells neither modifies diabetes incidence nor inflammatory responses in a mouse model of immune-mediated diabetes. Moreover, NIK silencing in DIO mice did not influence body weight gain, nor glucose metabolism. In vitro studies corroborated the in vivo findings in terms of β-cell survival, function, and downstream gene regulation. Taken together, our data suggest that NIK activation is dispensable for the development of diabetes.
Collapse
Affiliation(s)
- Peng Xiao
- Inflammation and Cell Death Signalling group, Laboratoire de Gastroentérologie Expérimental et Endotools, Université libre de Bruxelles, Brussels, Belgium
| | - Tatiana Takiishi
- Inflammation and Cell Death Signalling group, Laboratoire de Gastroentérologie Expérimental et Endotools, Université libre de Bruxelles, Brussels, Belgium
| | - Natalia Moretti Violato
- Inflammation and Cell Death Signalling group, Laboratoire de Gastroentérologie Expérimental et Endotools, Université libre de Bruxelles, Brussels, Belgium
| | - Giada Licata
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
- Fondazione Umberto Di Mario, c/o Toscana Life Sciences, Siena, Italy
| | - Francesco Dotta
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
- Fondazione Umberto Di Mario, c/o Toscana Life Sciences, Siena, Italy
- Tuscany Centre for Precision Medicine (CReMeP), Siena, Italy
| | - Guido Sebastiani
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
- Fondazione Umberto Di Mario, c/o Toscana Life Sciences, Siena, Italy
| | - Lorella Marselli
- Department of Clinical and Experimental Medicine, Islet Laboratory, University of Pisa, Pisa, Italy
| | - Sumeet Pal Singh
- Institute for Interdisciplinary Research in Human and Molecular Biology, Medical Faculty, Université libre de Bruxelles, Brussels, Belgium
| | - Mozes Sze
- Center for Inflammation Research, VIB, B-9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, B-9052, Ghent, Belgium
| | - Geert Van Loo
- Center for Inflammation Research, VIB, B-9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, B-9052, Ghent, Belgium
| | - Emmanuel Dejardin
- Laboratory of Molecular Immunology and Signal Transduction, GIGA-Insitute, ULiege, Liège, Belgium
| | - Esteban Nicolas Gurzov
- Signal Transduction and Metabolism Laboratory, Laboratoire de Gastroentérologie Expérimental et Endotools, Université libre de Bruxelles, Brussels, Belgium
| | - Alessandra Kupper Cardozo
- Inflammation and Cell Death Signalling group, Laboratoire de Gastroentérologie Expérimental et Endotools, Université libre de Bruxelles, Brussels, Belgium.
| |
Collapse
|
14
|
Lusk JB, Chua EHZ, Kaur P, Sung ICH, Lim WK, Lam VYM, Harmston N, Tolwinski NS. A non-canonical Raf function is required for dorsal-ventral patterning during Drosophila embryogenesis. Sci Rep 2022; 12:7684. [PMID: 35538124 PMCID: PMC9090920 DOI: 10.1038/s41598-022-11699-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 04/28/2022] [Indexed: 11/08/2022] Open
Abstract
Proper embryonic development requires directional axes to pattern cells into embryonic structures. In Drosophila, spatially discrete expression of transcription factors determines the anterior to posterior organization of the early embryo, while the Toll and TGFβ signalling pathways determine the early dorsal to ventral pattern. Embryonic MAPK/ERK signaling contributes to both anterior to posterior patterning in the terminal regions and to dorsal to ventral patterning during oogenesis and embryonic stages. Here we describe a novel loss of function mutation in the Raf kinase gene, which leads to loss of ventral cell fates as seen through the loss of the ventral furrow, the absence of Dorsal/NFκB nuclear localization, the absence of mesoderm determinants Twist and Snail, and the expansion of TGFβ. Gene expression analysis showed cells adopting ectodermal fates much like loss of Toll signaling. Our results combine novel mutants, live imaging, optogenetics and transcriptomics to establish a novel role for Raf, that appears to be independent of the MAPK cascade, in embryonic patterning.
Collapse
Affiliation(s)
- Jay B Lusk
- Division of Science, Yale-NUS College, Singapore, 138527, Singapore
| | | | - Prameet Kaur
- Division of Science, Yale-NUS College, Singapore, 138527, Singapore
| | | | - Wen Kin Lim
- Division of Science, Yale-NUS College, Singapore, 138527, Singapore
| | | | - Nathan Harmston
- Division of Science, Yale-NUS College, Singapore, 138527, Singapore
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Nicholas S Tolwinski
- Division of Science, Yale-NUS College, Singapore, 138527, Singapore.
- Yale-NUS College Research Labs @ E6, E6, 5 Engineering Drive 1, #04-02, Singapore, 117608, Singapore.
| |
Collapse
|
15
|
Li B, Li H, Dai L, Liu C, Wang L, Li Q, Gu C. NIK-SIX1 signalling axis regulates high glucose-induced endothelial cell dysfunction and inflammation. Autoimmunity 2022; 55:86-94. [PMID: 34894925 DOI: 10.1080/08916934.2021.2015579] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/05/2021] [Accepted: 12/04/2021] [Indexed: 12/12/2022]
Abstract
Endothelial dysfunction and inflammation are the main manifestations of diabetes-associated atherosclerosis. This paper studied the roles of NF-κB-inducing kinase (NIK) and sine oculis homeobox homolog 1 (SIX1) in regulating high glucose-induced endothelial dysfunction and inflammation. The expression of NIK and SIX1 in human umbilical vein endothelial cells (HUVECs) was silenced by transfection with the specific shRNAs. HUVECs exposed to high glucose were considered as a cell model of endothelial dysfunction. Expression of NIK and SIX1 following transfection was measured by qRT-PCR and western blotting analysis. The proliferation, migration, and inflammation of HUVECs were evaluated by EdU staining, scratch test, ELISA, and western blotting. High glucose (30 mM) significantly decreased the proliferation and migration of HUVECs. High glucose-induced the expression of adhesion molecules VCAM-1 and ICAM-1. Moreover, high glucose increased the release of IL-1β, IL-6, TNF-α, and MCP-1. Transfection of cells with NIK shRNA significantly reversed the toxic effects of high glucose on HUVECs. Of contrast, SIX1 shRNA accelerated the effects of high glucose on HUVECs. NIK shRNA inhibited the accumulation of RelA, RelB, and p52. Meanwhile, NIK shRNA led to SIX1 downregulation which further induced the activation of the NF-κB pathway. NIK-SIX1 signalling axis was suggested to be critical in the regulation of high glucose-induced endothelial dysfunction and inflammation. SIX1 may function as an immunological gatekeeper to control the excessive inflammation mediated by NIK in diabetes-associated atherosclerosis.
Collapse
Affiliation(s)
- Bo Li
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Haiming Li
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Longsheng Dai
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Changcheng Liu
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Liangshan Wang
- Department of Cardiac Surgery Intensive Care Unit, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Qin Li
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Chengxiong Gu
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
16
|
Nobari ST, Nojadeh JN, Talebi M. B-cell maturation antigen targeting strategies in multiple myeloma treatment, advantages and disadvantages. J Transl Med 2022; 20:82. [PMID: 35144648 PMCID: PMC8832753 DOI: 10.1186/s12967-022-03285-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 01/29/2022] [Indexed: 01/02/2023] Open
Abstract
B cell maturation antigen (BCMA), a transmembrane glycoprotein member of the tumor necrosis factor receptor superfamily 17 (TNFRSF17), highly expressed on the plasma cells of Multiple myeloma (MM) patients, as well as the normal population. BCMA is used as a biomarker for MM. Two members of the TNF superfamily proteins, including B-cell activating factor (BAFF) and A proliferation-inducing ligand (APRIL), are closely related to BCMA and play an important role in plasma cell survival and progression of MM. Despite the maximum specificity of the monoclonal antibody technologies, introducing the tumor-specific antigen(s) is not applicable for all malignancies, such as MM that there plenty of relatively specific antigens such as GPCR5D, MUC1, SLAMF7 and etc., but higher expression of BCMA on these cells in comparison with normal ones can be regarded as a relatively exclusive marker. Currently, different monoclonal antibody (mAb) technologies applied in anti-MM therapies such as daratuzumab, SAR650984, GSK2857916, and CAR-T cell therapies are some of these tools that are reviewed in the present manuscript. By the way, the structure, function, and signaling of the BCMA and related molecule(s) role in normal plasma cells and MM development, evaluated as well as the potential side effects of its targeting by different CAR-T cells generations. In conclusion, BCMA can be regarded as an ideal molecule to be targeted in immunotherapeutic methods, regarding lower potential systemic and local side effects.
Collapse
Affiliation(s)
- Shirin Teymouri Nobari
- Department of Medical Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Jafar Nouri Nojadeh
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Talebi
- Department of Applied Cells Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
17
|
Figurová D, Tokárová K, Greifová H, Knížatová N, Kolesárová A, Lukáč N. Inflammation, It's Regulation and Antiphlogistic Effect of the Cyanogenic Glycoside Amygdalin. Molecules 2021; 26:5972. [PMID: 34641516 PMCID: PMC8512454 DOI: 10.3390/molecules26195972] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/27/2021] [Accepted: 09/29/2021] [Indexed: 12/14/2022] Open
Abstract
The inflammatory reaction accompanies in part or in full any disease process in the vascularized metazoan. This complicated reaction is controlled by regulatory mechanisms, some of which produce unpleasant symptomatic manifestations of inflammation. Therefore, there has been an effort to develop selective drugs aimed at removing pain, fever, or swelling. Gradually, however, serious adverse side effects of such inhibitors became apparent. Scientific research has therefore continued to explore new possibilities, including naturally available substances. Amygdalin is a cyanogenic glycoside present, e.g., in bitter almonds. This glycoside has already sparked many discussions among scientists, especially about its anticancer potential and related toxic cyanides. However, toxicity at different doses made it generally unacceptable. Although amygdalin given at the correct oral dose may not lead to poisoning, it has not yet been accurately quantified, as its action is often affected by different intestinal microbial consortia. Its pharmacological activities have been studied, but its effects on the body's inflammatory response are lacking. This review discusses the chemical structure, toxicity, and current knowledge of the molecular mechanism of amygdalin activity on immune functions, including the anti-inflammatory effect, but also discusses inflammation as such, its mediators with diverse functions, which are usually targeted by drugs.
Collapse
Affiliation(s)
| | - Katarína Tokárová
- Department of Animal Physiology, Faculty of Biotechnology and Food Science, Slovak University of Agriculture in Nitra, Trieda Andreja Hlinku 2, 949 76 Nitra, Slovakia; (D.F.); (H.G.); (N.K.); (A.K.); (N.L.)
| | | | | | | | | |
Collapse
|
18
|
Chawla M, Mukherjee T, Deka A, Chatterjee B, Sarkar UA, Singh AK, Kedia S, Lum J, Dhillon MK, Banoth B, Biswas SK, Ahuja V, Basak S. An epithelial Nfkb2 pathway exacerbates intestinal inflammation by supplementing latent RelA dimers to the canonical NF-κB module. Proc Natl Acad Sci U S A 2021; 118:e2024828118. [PMID: 34155144 PMCID: PMC8237674 DOI: 10.1073/pnas.2024828118] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aberrant inflammation, such as that associated with inflammatory bowel disease (IBD), is fueled by the inordinate activity of RelA/NF-κB factors. As such, the canonical NF-κB module mediates controlled nuclear activation of RelA dimers from the latent cytoplasmic complexes. What provokes pathological RelA activity in the colitogenic gut remains unclear. The noncanonical NF-κB pathway typically promotes immune organogenesis involving Nfkb2 gene products. Because NF-κB pathways are intertwined, we asked whether noncanonical signaling aggravated inflammatory RelA activity. Our investigation revealed frequent engagement of the noncanonical pathway in human IBD. In a mouse model of experimental colitis, we established that Nfkb2-mediated regulations escalated the RelA-driven proinflammatory gene response in intestinal epithelial cells, exacerbating the infiltration of inflammatory cells and colon pathologies. Our mechanistic studies clarified that cell-autonomous Nfkb2 signaling supplemented latent NF-κB dimers, leading to a hyperactive canonical RelA response in the inflamed colon. In sum, the regulation of latent NF-κB dimers appears to link noncanonical Nfkb2 signaling to RelA-driven inflammatory pathologies and may provide for therapeutic targets.
Collapse
Affiliation(s)
- Meenakshi Chawla
- Systems Immunology Laboratory, National Institute of Immunology, New Delhi 110067, India
| | - Tapas Mukherjee
- Systems Immunology Laboratory, National Institute of Immunology, New Delhi 110067, India
| | - Alvina Deka
- Systems Immunology Laboratory, National Institute of Immunology, New Delhi 110067, India
| | - Budhaditya Chatterjee
- Systems Immunology Laboratory, National Institute of Immunology, New Delhi 110067, India
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Uday Aditya Sarkar
- Systems Immunology Laboratory, National Institute of Immunology, New Delhi 110067, India
| | - Amit K Singh
- Department of Gastroenterology, All India Institute of Medical Science, New Delhi 110029, India
| | - Saurabh Kedia
- Department of Gastroenterology, All India Institute of Medical Science, New Delhi 110029, India
| | - Josephine Lum
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore 138632
| | - Manprit Kaur Dhillon
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore 138632
| | - Balaji Banoth
- Systems Immunology Laboratory, National Institute of Immunology, New Delhi 110067, India
| | - Subhra K Biswas
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore 138632
| | - Vineet Ahuja
- Department of Gastroenterology, All India Institute of Medical Science, New Delhi 110029, India
| | - Soumen Basak
- Systems Immunology Laboratory, National Institute of Immunology, New Delhi 110067, India;
| |
Collapse
|
19
|
Mohallem R, Aryal UK. Regulators of TNFα mediated insulin resistance elucidated by quantitative proteomics. Sci Rep 2020; 10:20878. [PMID: 33257747 PMCID: PMC7705713 DOI: 10.1038/s41598-020-77914-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 11/03/2020] [Indexed: 12/21/2022] Open
Abstract
Obesity is a growing epidemic worldwide and is a major risk factor for several chronic diseases, including diabetes, kidney disease, heart disease, and cancer. Obesity often leads to type 2 diabetes mellitus, via the increased production of proinflammatory cytokines such as tumor necrosis factor-α (TNFα). Our study combines different proteomic techniques to investigate the changes in the global proteome, secretome and phosphoproteome of adipocytes under chronic inflammation condition, as well as fundamental cross-talks between different cellular pathways regulated by chronic TNFα exposure. Our results show that many key regulator proteins of the canonical and non-canonical NF-κB pathways, such as Nfkb2, and its downstream effectors, including Csf-1 and Lgals3bp, directly involved in leukocyte migration and invasion, were significantly upregulated at the intra and extracellular proteomes suggesting the progression of inflammation. Our data provides evidence of several key proteins that play a role in the development of insulin resistance.
Collapse
Affiliation(s)
- Rodrigo Mohallem
- Department of Comparative Pathobiology, Purdue University, West Lafayette, USA
- Purdue Proteomics Facility, Bindley Bioscience Center, Purdue University, West Lafayette, USA
| | - Uma K Aryal
- Department of Comparative Pathobiology, Purdue University, West Lafayette, USA.
- Purdue Proteomics Facility, Bindley Bioscience Center, Purdue University, West Lafayette, USA.
| |
Collapse
|
20
|
Fomicheva M, Macara IG. Genome-wide CRISPR screen identifies noncanonical NF-κB signaling as a regulator of density-dependent proliferation. eLife 2020; 9:63603. [PMID: 33185187 PMCID: PMC7685705 DOI: 10.7554/elife.63603] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 11/06/2020] [Indexed: 12/17/2022] Open
Abstract
Epithelial cells possess intrinsic mechanisms to maintain an appropriate cell density for normal tissue morphogenesis and homeostasis. Defects in such mechanisms likely contribute to hyperplasia and cancer initiation. To identify genes that regulate the density-dependent proliferation of murine mammary epithelial cells, we developed a fluorescence-activated cell sorting assay based on fluorescence ubiquitination cell cycle indicator, which marks different stages of the cell cycle with distinct fluorophores. Using this powerful assay, we performed a genome-wide CRISPR/Cas9 knockout screen, selecting for cells that proliferate normally at low density but continue to divide at high density. Unexpectedly, one top hit was Traf3, a negative regulator of NF-κB signaling that has never previously been linked to density-dependent proliferation. We demonstrate that loss of Traf3 specifically activates noncanonical NF-κB signaling. This in turn triggers an innate immune response and drives cell division independently of known density-dependent proliferation mechanisms, including YAP/TAZ signaling and cyclin-dependent kinase inhibitors, by blocking entry into quiescence.
Collapse
Affiliation(s)
- Maria Fomicheva
- Department of Cell and Developmental Biology Vanderbilt University School of Medicine Nashville, Nashville, United States
| | - Ian G Macara
- Department of Cell and Developmental Biology Vanderbilt University School of Medicine Nashville, Nashville, United States
| |
Collapse
|
21
|
Morgan EL, Chen Z, Van Waes C. Regulation of NFκB Signalling by Ubiquitination: A Potential Therapeutic Target in Head and Neck Squamous Cell Carcinoma? Cancers (Basel) 2020; 12:E2877. [PMID: 33036368 PMCID: PMC7601648 DOI: 10.3390/cancers12102877] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/22/2020] [Accepted: 09/29/2020] [Indexed: 02/08/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide, with over 600,000 cases per year. The primary causes for HNSCC include smoking and alcohol consumption, with an increasing number of cases attributed to infection with Human Papillomavirus (HPV). The treatment options for HNSCC currently include surgery, radiotherapy, and/or platinum-based chemotherapeutics. Cetuximab (targeting EGFR) and Pembrolizumab (targeting PD-1) have been approved for advanced stage, recurrent, and/or metastatic HNSCC. Despite these advances, whilst HPV+ HNSCC has a 3-year overall survival (OS) rate of around 80%, the 3-year OS for HPV- HNSCC is still around 55%. Aberrant signal activation of transcription factor NFκB plays an important role in the pathogenesis and therapeutic resistance of HNSCC. As an important mediator of inflammatory signalling and the immune response to pathogens, the NFκB pathway is tightly regulated to prevent chronic inflammation, a key driver of tumorigenesis. Here, we discuss how NFκB signalling is regulated by the ubiquitin pathway and how this pathway is deregulated in HNSCC. Finally, we discuss the current strategies available to target the ubiquitin pathway and how this may offer a potential therapeutic benefit in HNSCC.
Collapse
Affiliation(s)
- Ethan L. Morgan
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute of Deafness and Other Communication Disorders, NIH, Bethesda, MD 20892, USA;
| | - Zhong Chen
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute of Deafness and Other Communication Disorders, NIH, Bethesda, MD 20892, USA;
| | | |
Collapse
|
22
|
Rayego-Mateos S, Morgado-Pascual JL, Valdivielso JM, Sanz AB, Bosch-Panadero E, Rodrigues-Díez RR, Egido J, Ortiz A, González-Parra E, Ruiz-Ortega M. TRAF3 Modulation: Novel Mechanism for the Anti-inflammatory Effects of the Vitamin D Receptor Agonist Paricalcitol in Renal Disease. J Am Soc Nephrol 2020; 31:2026-2042. [PMID: 32631974 DOI: 10.1681/asn.2019111206] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 04/07/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND CKD leads to vitamin D deficiency. Treatment with vitamin D receptor agonists (VDRAs) may have nephroprotective and anti-inflammatory actions, but their mechanisms of action are poorly understood. METHODS Modulation of the noncanonical NF-κB2 pathway and its component TNF receptor-associated factor 3 (TRAF3) by the VDRA paricalcitol was studied in PBMCs from patients with ESKD, cytokine-stimulated cells, and preclinical kidney injury models. RESULTS In PBMCs isolated from patients with ESKD, TRAF3 protein levels were lower than in healthy controls. This finding was associated with evidence of noncanonical NF-κB2 activation and a proinflammatory state. However, PBMCs from patients with ESKD treated with paricalcitol did not exhibit these features. Experiments in cultured cells confirmed the link between TRAF3 and NF-κB2/inflammation. Decreased TRAF3 ubiquitination in K48-linked chains and cIAP1-TRAF3 interaction mediated the mechanisms of paricalcitol action.TRAF3 overexpression by CRISPR/Cas9 technology mimicked VDRA's effects. In a preclinical model of kidney injury, paricalcitol inhibited renal NF-κB2 activation and decreased renal inflammation. In VDR knockout mice with renal injury, paricalcitol prevented TRAF3 downregulation and NF-κB2-dependent gene upregulation, suggesting a VDR-independent anti-inflammatory effect of paricalcitol. CONCLUSIONS These data suggest the anti-inflammatory actions of paricalcitol depend on TRAF3 modulation and subsequent inhibition of the noncanonical NF-κB2 pathway, identifying a novel mechanism for VDRA's effects. Circulating TRAF3 levels could be a biomarker of renal damage associated with the inflammatory state.
Collapse
Affiliation(s)
- Sandra Rayego-Mateos
- Molecular and Cellular Biology in Renal and Vascular Pathology, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz,Universidad autonoma de madrid, Madrid, Spain.,Vascular and Renal Translational Research Group. Institut de Receca Biomedica de Lleida (IRBLleida), Lleida, Spain
| | - Jose Luis Morgado-Pascual
- Molecular and Cellular Biology in Renal and Vascular Pathology, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz,Universidad autonoma de madrid, Madrid, Spain.,REDinREN (Red de Investigación Renal), Madrid, Spain
| | - José Manuel Valdivielso
- Vascular and Renal Translational Research Group. Institut de Receca Biomedica de Lleida (IRBLleida), Lleida, Spain.,REDinREN (Red de Investigación Renal), Madrid, Spain
| | - Ana Belén Sanz
- REDinREN (Red de Investigación Renal), Madrid, Spain.,Laboratory of Nephrology and Hypertension, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Enrique Bosch-Panadero
- Laboratory of Nephrology and Hypertension, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Raúl R Rodrigues-Díez
- Molecular and Cellular Biology in Renal and Vascular Pathology, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz,Universidad autonoma de madrid, Madrid, Spain
| | - Jesús Egido
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz.Universidad Autónoma. 28040 Madrid, Spain; Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM). 28029 Madrid, Spain
| | - Alberto Ortiz
- REDinREN (Red de Investigación Renal), Madrid, Spain.,Laboratory of Nephrology and Hypertension, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Emilio González-Parra
- Laboratory of Nephrology and Hypertension, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Marta Ruiz-Ortega
- Molecular and Cellular Biology in Renal and Vascular Pathology, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz,Universidad autonoma de madrid, Madrid, Spain .,REDinREN (Red de Investigación Renal), Madrid, Spain
| |
Collapse
|
23
|
Chen B, Li C, Yao J, Shi L, Liu W, Wang F, Huo S, Zhang Y, Lu Y, Ashraf U, Ye J, Liu X. Zebrafish NIK Mediates IFN Induction by Regulating Activation of IRF3 and NF-κB. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:1881-1891. [PMID: 32066597 DOI: 10.4049/jimmunol.1900561] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 01/09/2020] [Indexed: 01/19/2023]
Abstract
Type I IFN mediates the innate immune system to provide defense against viral infections. NF-κB-inducing kinase (NIK) potentiates the basal activation of endogenous STING, which facilitates the recruitment of TBK1 with the ectopically expressed IRF3 to induce IFN production. Moreover, NIK phosphorylates IKKα and confers its ability to phosphorylate p100 (also known as NF-κB2) in mammals. Our study demonstrated that NIK plays a critical role in IFN production in teleost fish. It was found that NIK interacts with IKKα in the cytoplasm and that IKKα phosphorylates the NIK at the residue Thr432, which is different from the mammals. Overexpression of NIK caused the activation of IRF3 and NF-κB, which in turn led to the production of IFN and IFN-stimulated genes (ISGs). Furthermore, the ectopic expression of NIK was observed to be associated with a reduced replication of the fish virus, whereas silencing of endogenous NIK had an opposite effect in vitro. Furthermore, NIK knockdown significantly reduced the expression of IFN and key ISGs in zebrafish larvae after spring viremia of carp virus infection. Additionally, the replication of spring viremia of carp virus was enhanced in NIK knockdown zebrafish larvae, leading to a lower survival rate. In summary, our findings revealed a previously undescribed function of NIK in activating IFN and ISGs as a host antiviral response. These findings may facilitate the establishment of antiviral therapy to combat fish viruses.
Collapse
Affiliation(s)
- Bo Chen
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, Hubei, China
- Hubei Engineering Technology Research Center for Aquatic Animal Diseases Control and Prevention, Wuhan 430070, Hubei, China
| | - Chen Li
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, Hubei, China
- Hubei Engineering Technology Research Center for Aquatic Animal Diseases Control and Prevention, Wuhan 430070, Hubei, China
| | - Jian Yao
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, Hubei, China
- Hubei Engineering Technology Research Center for Aquatic Animal Diseases Control and Prevention, Wuhan 430070, Hubei, China
| | - Lin Shi
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, Hubei, China
- Hubei Engineering Technology Research Center for Aquatic Animal Diseases Control and Prevention, Wuhan 430070, Hubei, China
| | - Wanmeng Liu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, Hubei, China
- Hubei Engineering Technology Research Center for Aquatic Animal Diseases Control and Prevention, Wuhan 430070, Hubei, China
| | - Fang Wang
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, Hubei, China
- Hubei Engineering Technology Research Center for Aquatic Animal Diseases Control and Prevention, Wuhan 430070, Hubei, China
| | - Shitian Huo
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, Hubei, China
- Hubei Engineering Technology Research Center for Aquatic Animal Diseases Control and Prevention, Wuhan 430070, Hubei, China
| | - Yongan Zhang
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, Hubei, China
- Hubei Engineering Technology Research Center for Aquatic Animal Diseases Control and Prevention, Wuhan 430070, Hubei, China
| | - Yuanan Lu
- Department of Public Health Sciences, University of Hawaii at Manoa, Honolulu, Hawaii 96822; and
| | - Usama Ashraf
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Jing Ye
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Xueqin Liu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, Hubei, China;
- Hubei Engineering Technology Research Center for Aquatic Animal Diseases Control and Prevention, Wuhan 430070, Hubei, China
| |
Collapse
|
24
|
Zheng B, Zhou J, Wang H. Host microRNAs and exosomes that modulate influenza virus infection. Virus Res 2020; 279:197885. [PMID: 31981772 DOI: 10.1016/j.virusres.2020.197885] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/18/2020] [Accepted: 01/21/2020] [Indexed: 02/07/2023]
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that post-transcriptionally regulate over half of human protein-coding genes and play a vital role in cellular development, proliferation, metabolism, and homeostasis. Exosomes are rounded or cup-like extracellular vesicles that carry proteins, mRNAs, miRNAs, and lipids for release and exchange messages between cells involved in various cellular processes. Influenza virus is a substantial public health challenge. The expression of host miRNAs is altered in response to stimulation by influenza virus. These dysregulated miRNAs directly or indirectly target viral genes to regulate viral replication and stimulate or suppress innate immune responses and cell apoptosis during viral infection. Exosomes released by infected cells are associated with the transfer of antigens and key molecules that activate and modulate immune function. Dysregulation of miRNAs and secretion of exosomes are associated with pathogenicity and immune regulation during influenza infection. This review provides a comprehensive summary of the information available regarding host miRNAs and exosomes that are involved in the modulation of influenza virus infection and will facilitate the development of preventative or therapeutic strategies against influenza virus.
Collapse
Affiliation(s)
- Baojia Zheng
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Jinan University, Guangzhou, 510632, China
| | - Junmei Zhou
- Key Laboratory of Tropical Diseases Control, Ministry of Education, and Deparment of Medical Microbiology, Zhongshan Medical College, Sun Yat-Sen University, Guangzhou, 510080, China.
| | - Hui Wang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
25
|
Wu H, Liu H, Zhao X, Zheng Y, Liu B, Zhang L, Gao C. IKIP Negatively Regulates NF-κB Activation and Inflammation through Inhibition of IKKα/β Phosphorylation. THE JOURNAL OF IMMUNOLOGY 2019; 204:418-427. [PMID: 31826938 DOI: 10.4049/jimmunol.1900626] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 11/07/2019] [Indexed: 01/27/2023]
Abstract
Stringent regulation of the transcription factor NF-κB signaling is essential for the activation of host immune responses and maintaining homeostasis, yet the molecular mechanisms involved in its tight regulation are not completely understood. In this study, we report that IKK-interacting protein (IKIP) negatively regulates NF-κB activation. IKIP interacted with IKKα/β to block its association with NEMO, thereby inhibiting the phosphorylation of IKKα/β and the activation of NF-κB. Upon LPS, TNF-α, and IL-1β stimulation, IKIP-deficient macrophages exhibited more and prolonged IKKα/β phosphorylation, IκB, and p65 phosphorylation and production of NF-κB-responsive genes. Moreover, IKIP-deficient mice were more susceptible to LPS-induced septic shock and dextran sodium sulfate-induced colitis. Our study identifies a previously unrecognized role for IKIP in the negative regulation of NF-κB activation by inhibition of IKKα/β phosphorylation through the disruption of IKK complex formation.
Collapse
Affiliation(s)
- Haifeng Wu
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, China; and
| | - Hansen Liu
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, China; and
| | - Xueying Zhao
- Department of Transfusion, The Second Hospital of Shandong University, Jinan 250000, China
| | - Yi Zheng
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, China; and
| | - Bingyu Liu
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, China; and
| | - Lei Zhang
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, China; and
| | - Chengjiang Gao
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, China; and
| |
Collapse
|
26
|
Dhar A, Chawla M, Chattopadhyay S, Oswal N, Umar D, Gupta S, Bal V, Rath S, George A, Arimbasseri GA, Basak S. Role of NF-kappaB2-p100 in regulatory T cell homeostasis and activation. Sci Rep 2019; 9:13867. [PMID: 31554891 PMCID: PMC6761191 DOI: 10.1038/s41598-019-50454-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 09/10/2019] [Indexed: 12/12/2022] Open
Abstract
The immunological roles of the nuclear factor-kappaB (NF-κB) pathway are mediated via the canonical components in immune responses and via non-canonical components in immune organogenesis and homeostasis, although the two components are capable of crosstalk. Regulatory CD4 T cells (Tregs) are homeostatically functional and represent an interesting potential meeting point of these two NF-κB components. We show that mice deficient in the non-canonical NF-κB component gene Nfkb2 (p100) had normal thymic development and suppressive function of Tregs. However, they had enhanced frequencies of peripheral 'effector-phenotype' Tregs (eTregs). In bi-parental chimeras of wild-type (WT) and Nfkb2-/- mice, the Nfkb2-/- genotype was over-represented in Tregs, with a further increase in the relative prominence of eTregs. Consistent with distinct properties of eTregs, the Nfkb2-/- genotype was more prominent in Tregs in extra-lymphoid tissues such as liver in the bi-parental chimeras. The Nfkb2-/- Tregs also displayed greater survival, activation and proliferation in vivo. These Nfkb2-/- Tregs showed higher nuclear NF-κB activity mainly comprising of RelB-containing dimers, in contrast to the prominence of cRel- and RelA-containing dimers in WT Tregs. Since p100 is an inhibitor of RelB activation as well as a participant as cleaved p52 in RelB nuclear activity, we tested bi-parental chimeras of WT and Relb-/- mice, and found normal frequencies of Relb-/- Tregs and eTregs in these chimeric mice. Our findings confirm and extend recent data, and indicate that p100 normally restrains RelB-mediated Treg activation, and in the absence of p100, p50-RelB dimers can contribute to Treg activation.
Collapse
Affiliation(s)
- Atika Dhar
- National Institute of Immunology, New Delhi, India
| | | | | | - Neelam Oswal
- National Institute of Immunology, New Delhi, India
| | - Danish Umar
- National Institute of Immunology, New Delhi, India
| | - Suman Gupta
- National Institute of Immunology, New Delhi, India
| | - Vineeta Bal
- National Institute of Immunology, New Delhi, India
| | | | - Anna George
- National Institute of Immunology, New Delhi, India
| | | | - Soumen Basak
- National Institute of Immunology, New Delhi, India
| |
Collapse
|
27
|
Gamboa-Cedeño AM, Castillo M, Xiao W, Waldmann TA, Ranuncolo SM. Alternative and canonical NF-kB pathways DNA-binding hierarchies networks define Hodgkin lymphoma and Non-Hodgkin diffuse large B Cell lymphoma respectively. J Cancer Res Clin Oncol 2019; 145:1437-1448. [PMID: 30941572 PMCID: PMC8317045 DOI: 10.1007/s00432-019-02909-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 03/25/2019] [Indexed: 12/22/2022]
Abstract
PURPOSE Despite considerable evidence that supports the NF-kB role in the immune system and lymphomagenesis, it is unclear whether specific NF-kB dimers control a particular set of genes that account for their biological functions. Our previous work showed that Hodgkin Lymphoma (HL) is unique, among germinal center (GC)-derived lymphomas, with respect to its dependency on Rel-B to survive. In contrast, diffuse large B-Cell lymphoma (DLBCL) including both Activated B-Cell-Like and Germinal Center B-Cell-Like, requires cREL and Rel-A to survive and it is not affected by Rel-B depletion. These findings highlighted the activity of specific NF-kB subunits in different GC-derived lymphomas. METHODS Sequenced chromatin immunoprecipitated DNA fragments (ChIP-Seq) analysis revealed an extensive NF-kB DNA-binding network in DLBCL and HL. The ChIP-Seq data was merged with microarray analysis following the Rel-A, Rel-B or cRel knockdown to determine effectively regulated genes. RESULTS Downstream target analysis showed enrichment for cell cycle control, among other signatures. Rel-B and cRel controlled different genes within the same signature in HL and DLBCL, respectively. BCL2 was exclusively controlled by Rel-B in HL. Both mRNA and protein levels decreased following Rel-B depletion meanwhile there was no change upon cRel knock-down. BCL2 exogenous expression partially rescued the death induced by decreased Rel-B in HL cells. CONCLUSION The Rel-B hierarchical network defined HL and the cRel hierarchical network characterized DLBCL. Each Rel member performs specific functions in distinct GC-derived lymphomas. This result should be considered for the development of targeted therapies that are aimed to selectively inhibit individual NF-kB dimers.
Collapse
Affiliation(s)
- Angélica María Gamboa-Cedeño
- Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB), CONICET-Instituto Universitario del Hospital Italiano-Hospital Italiano de Buenos Aires, Potosí 4240 C.P., C1183AEG, Buenos Aires, Argentina
| | - Mariángeles Castillo
- Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB), CONICET-Instituto Universitario del Hospital Italiano-Hospital Italiano de Buenos Aires, Potosí 4240 C.P., C1183AEG, Buenos Aires, Argentina
| | - Wenming Xiao
- Center for Information Technology, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Thomas A Waldmann
- Lymphoid Malignancies Branch, Center for Cancer Research (CCR), NCI-NIH, Bethesda, MD, USA
| | - Stella Maris Ranuncolo
- Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB), CONICET-Instituto Universitario del Hospital Italiano-Hospital Italiano de Buenos Aires, Potosí 4240 C.P., C1183AEG, Buenos Aires, Argentina.
- Departamento de Histología y Biología Celular, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.
- National Council of Scientific and Technological Research (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
28
|
Kucharzewska P, Maracle CX, Jeucken KCM, van Hamburg JP, Israelsson E, Furber M, Tas SW, Olsson HK. NIK-IKK complex interaction controls NF-κB-dependent inflammatory activation of endothelium in response to LTβR ligation. J Cell Sci 2019; 132:jcs225615. [PMID: 30837284 DOI: 10.1242/jcs.225615] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 02/14/2019] [Indexed: 12/26/2022] Open
Abstract
NF-κB-inducing kinase (NIK; also known as MAP3K14) is a central regulator of non-canonical NF-κB signaling in response to stimulation of TNF receptor superfamily members, such as the lymphotoxin-β receptor (LTβR), and is implicated in pathological angiogenesis associated with chronic inflammation and cancer. Here, we identify a previously unrecognized role of the LTβR-NIK axis during inflammatory activation of human endothelial cells (ECs). Engagement of LTβR-triggered canonical and non-canonical NF-κB signaling promoted expression of inflammatory mediators and adhesion molecules, and increased immune cell adhesion to ECs. Sustained LTβR-induced inflammatory activation of ECs was NIK dependent, but independent of p100, indicating that the non-canonical arm of NF-κB is not involved. Instead, prolonged activation of canonical NF-κB signaling, through the interaction of NIK with IκB kinase α and β (also known as CHUK and IKBKB, respectively), was required for the inflammatory response. Endothelial inflammatory activation induced by synovial fluid from rheumatoid arthritis patients was significantly reduced by NIK knockdown, suggesting that NIK-mediated alternative activation of canonical NF-κB signaling is a key driver of pathological inflammatory activation of ECs. Targeting NIK could thus provide a novel approach for treating chronic inflammatory diseases.
Collapse
Affiliation(s)
- Paulina Kucharzewska
- Respiratory, Inflammation and Autoimmunity IMED Biotech Unit, AstraZeneca, Gothenburg, SE-431 83 Mölndal, Sweden
| | - Chrissta X Maracle
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology and Rheumatology and Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Kim C M Jeucken
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology and Rheumatology and Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Jan Piet van Hamburg
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology and Rheumatology and Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Elisabeth Israelsson
- Respiratory, Inflammation and Autoimmunity IMED Biotech Unit, AstraZeneca, Gothenburg, SE-431 83 Mölndal, Sweden
| | - Mark Furber
- Respiratory, Inflammation and Autoimmunity IMED Biotech Unit, AstraZeneca, Gothenburg, SE-431 83 Mölndal, Sweden
| | - Sander W Tas
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology and Rheumatology and Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Henric K Olsson
- Respiratory, Inflammation and Autoimmunity IMED Biotech Unit, AstraZeneca, Gothenburg, SE-431 83 Mölndal, Sweden
| |
Collapse
|
29
|
Nighot M, Rawat M, Al-Sadi R, Castillo EF, Nighot P, Ma TY. Lipopolysaccharide-Induced Increase in Intestinal Permeability Is Mediated by TAK-1 Activation of IKK and MLCK/MYLK Gene. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:797-812. [PMID: 30711488 PMCID: PMC6446229 DOI: 10.1016/j.ajpath.2018.12.016] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 12/11/2018] [Accepted: 12/20/2018] [Indexed: 01/13/2023]
Abstract
Lipopolysaccharides (LPSs) are a major component of Gram-negative bacterial cell wall and play an important role in promoting intestinal inflammatory responses. Recent studies have shown that physiologically relevant concentrations of LPS (0 to 2000 pg/mL) cause an increase in intestinal epithelial tight junction (TJ) permeability without causing cell death. However, the intracellular pathways and the mechanisms that mediate LPS-induced increase in intestinal TJ permeability remain unclear. The aim was to delineate the intracellular pathways that mediate the LPS-induced increase in intestinal permeability using in vitro and in vivo intestinal epithelial models. LPS-induced increase in intestinal epithelial TJ permeability was preceded by an activation of transforming growth factor-β-activating kinase-1 (TAK-1) and canonical NF-κB (p50/p65) pathways. The siRNA silencing of TAK-1 inhibited the activation of NF-κB p50/p65. The siRNA silencing of TAK-1 and p65/p50 subunit inhibited the LPS-induced increase in intestinal TJ permeability and the increase in myosin light chain kinase (MLCK) expression, confirming the regulatory role of TAK-1 and NF-κB p65/p50 in up-regulating MLCK expression and the subsequent increase in TJ permeability. The data also showed that toll-like receptor (TLR)-4/myeloid differentiation primary response (MyD)88 pathway was crucial upstream regulator of TAK-1 and NF-κB p50/p65 activation. In conclusion, activation of TAK-1 by the TLR-4/MyD88 signal transduction pathway and MLCK by NF-κB p65/p50 regulates the LPS-induced increase in intestinal epithelial TJ permeability.
Collapse
Affiliation(s)
- Meghali Nighot
- Department of Medicine, Pennsylvania State University, School of Medicine, Hershey, Pennsylvania
| | - Manmeet Rawat
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico
| | - Rana Al-Sadi
- Department of Medicine, Pennsylvania State University, School of Medicine, Hershey, Pennsylvania
| | - Eliseo F Castillo
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico
| | - Prashant Nighot
- Department of Medicine, Pennsylvania State University, School of Medicine, Hershey, Pennsylvania
| | - Thomas Y Ma
- Department of Medicine, Pennsylvania State University, School of Medicine, Hershey, Pennsylvania; Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico.
| |
Collapse
|
30
|
The impact of NF-κB signaling on pathogenesis and current treatment strategies in multiple myeloma. Blood Rev 2019; 34:56-66. [DOI: 10.1016/j.blre.2018.11.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 11/14/2018] [Accepted: 11/22/2018] [Indexed: 12/13/2022]
|
31
|
Suarez G, Romero-Gallo J, Piazuelo MB, Sierra JC, Delgado AG, Washington MK, Shah SC, Wilson KT, Peek RM. Nod1 Imprints Inflammatory and Carcinogenic Responses toward the Gastric Pathogen Helicobacter pylori. Cancer Res 2019; 79:1600-1611. [PMID: 30696658 DOI: 10.1158/0008-5472.can-18-2651] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 12/21/2018] [Accepted: 01/24/2019] [Indexed: 12/21/2022]
Abstract
Helicobacter pylori (H. pylori) is the strongest known risk for gastric cancer. The H. pylori cag type IV secretion system is an oncogenic locus that translocates peptidoglycan into host cells, where it is recognized by NOD1, an innate immune receptor. Beyond this, the role of NOD1 in H. pylori-induced cancer remains undefined. To address this knowledge gap, we infected two genetic models of Nod1 deficiency with the H. pylori cag + strain PMSS1: C57BL/6 mice, which rarely develop cancer, and INS-GAS FVB/N mice, which commonly develop cancer. Infected C57BL/6 Nod1-/- and INS-GAS Nod1-/- mice acutely developed more severe gastritis, and INS-GAS Nod1-/- mice developed gastric dysplasia more frequently compared with Nod1+/+ mice. Because Nod1 genotype status did not alter microbial phenotypes of in vivo-adapted H. pylori, we investigated host immunologic responses. H. pylori infection of Nod1-/- mice led to significantly increased gastric mucosal levels of Th1, Th17, and Th2 cytokines compared with Nod1 wild-type (WT) mice. To define the role of specific innate immune cells, we quantified cytokine secretion from H. pylori-infected primary gastric organoids generated from WT or Nod1-/- mice that were cocultured with or without WT or Nod1-/- macrophages. Infection increased cytokine production from gastric epithelial cells and macrophages and elevations were significantly increased with Nod1 deficiency. Furthermore, H. pylori infection altered the polarization status of Nod1-/- macrophages compared with Nod1+/+ macrophages. Collectively, these studies demonstrate that loss of Nod1 augments inflammatory and injury responses to H. pylori. Nod1 may exert its restrictive role by altering macrophage polarization, leading to immune evasion and microbial persistence. SIGNIFICANCE: These findings suggest that manipulation of NOD1 may represent a novel strategy to prevent or treat pathologic outcomes induced by H. pylori infection.
Collapse
Affiliation(s)
- Giovanni Suarez
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Judith Romero-Gallo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Maria B Piazuelo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Johanna C Sierra
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Alberto G Delgado
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - M Kay Washington
- Department of Pathology, Microbiology, and Immunology; Vanderbilt University Medical Center, Nashville, Tennessee
| | - Shailja C Shah
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Keith T Wilson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Pathology, Microbiology, and Immunology; Vanderbilt University Medical Center, Nashville, Tennessee
| | - Richard M Peek
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.
- Department of Pathology, Microbiology, and Immunology; Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
32
|
Maubach G, Feige MH, Lim MCC, Naumann M. NF-kappaB-inducing kinase in cancer. Biochim Biophys Acta Rev Cancer 2019; 1871:40-49. [PMID: 30419317 DOI: 10.1016/j.bbcan.2018.10.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 10/31/2018] [Accepted: 10/31/2018] [Indexed: 02/07/2023]
Abstract
Dysregulation of the alternative NF-κB signaling has severe developmental consequences that can ultimately lead to oncogenesis. Pivotal for the activation of the alternative NF-κB pathway is the stabilization of the NF-κB-inducing kinase (NIK). The aim of this review is to focus on the emerging role of NIK in cancer. The documented subversion of NIK in cancers highlights NIK as a possible therapeutic target. Recent studies show that the alterations of NIK or the components of its regulatory complex are manifold including regulation on the transcript level, copy number changes, mutations as well as protein modifications. High NIK activity is associated with different human malignancies and has adverse effects on tumor patient survival. We discuss here research focusing on deciphering the contribution of NIK towards cancer development and progression. We also report that it is possible to engineer inhibitors with high specificity for NIK and describe developments in this area.
Collapse
Affiliation(s)
- Gunter Maubach
- Institute of Experimental Internal Medicine, Otto von Guericke University, 39120 Magdeburg, Germany
| | - Michael H Feige
- Institute of Experimental Internal Medicine, Otto von Guericke University, 39120 Magdeburg, Germany
| | - Michelle C C Lim
- Institute of Experimental Internal Medicine, Otto von Guericke University, 39120 Magdeburg, Germany
| | - Michael Naumann
- Institute of Experimental Internal Medicine, Otto von Guericke University, 39120 Magdeburg, Germany.
| |
Collapse
|
33
|
NF-κB Signaling in Targeting Tumor Cells by Oncolytic Viruses-Therapeutic Perspectives. Cancers (Basel) 2018; 10:cancers10110426. [PMID: 30413032 PMCID: PMC6265863 DOI: 10.3390/cancers10110426] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 11/04/2018] [Accepted: 11/06/2018] [Indexed: 12/14/2022] Open
Abstract
In recent years, oncolytic virotherapy became a promising therapeutic approach, leading to the introduction of a novel generation of anticancer drugs. However, despite evoking an antitumor response, introducing an oncolytic virus (OV) to the patient is still inefficient to overcome both tumor protective mechanisms and the limitation of viral replication by the host. In cancer treatment, nuclear factor (NF)-κB has been extensively studied among important therapeutic targets. The pleiotropic nature of NF-κB transcription factor includes its involvement in immunity and tumorigenesis. Therefore, in many types of cancer, aberrant activation of NF-κB can be observed. At the same time, the activity of NF-κB can be modified by OVs, which trigger an immune response and modulate NF-κB signaling. Due to the limitation of a monotherapy exploiting OVs only, the antitumor effect can be enhanced by combining OV with NF-κB-modulating drugs. This review describes the influence of OVs on NF-κB activation in tumor cells showing NF-κB signaling as an important aspect, which should be taken into consideration when targeting tumor cells by OVs.
Collapse
|
34
|
Lacher SM, Thurm C, Distler U, Mohebiany AN, Israel N, Kitic M, Ebering A, Tang Y, Klein M, Wabnitz GH, Wanke F, Samstag Y, Bopp T, Kurschus FC, Simeoni L, Tenzer S, Waisman A. NF-κB inducing kinase (NIK) is an essential post-transcriptional regulator of T-cell activation affecting F-actin dynamics and TCR signaling. J Autoimmun 2018; 94:110-121. [PMID: 30061013 DOI: 10.1016/j.jaut.2018.07.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 07/20/2018] [Accepted: 07/25/2018] [Indexed: 11/16/2022]
Abstract
NF-κB inducing kinase (NIK) is the key protein of the non-canonical NF-κB pathway and is important for the development of lymph nodes and other secondary immune organs. We elucidated the specific role of NIK in T cells using T-cell specific NIK-deficient (NIKΔT) mice. Despite showing normal development of lymphoid organs, NIKΔT mice were resistant to induction of CNS autoimmunity. T cells from NIKΔT mice were deficient in late priming, failed to up-regulate T-bet and to transmigrate into the CNS. Proteomic analysis of activated NIK-/- T cells showed de-regulated expression of proteins involved in the formation of the immunological synapse: in particular, proteins involved in cytoskeleton dynamics. In line with this we found that NIK-deficient T cells were hampered in phosphorylation of Zap70, LAT, AKT, ERK1/2 and PLCγ upon TCR engagement. Hence, our data disclose a hitherto unknown function of NIK in T-cell priming and differentiation.
Collapse
MESH Headings
- Actins/genetics
- Actins/immunology
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/immunology
- Animals
- Central Nervous System/immunology
- Central Nervous System/pathology
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Gene Expression Profiling
- Gene Expression Regulation
- Lymph Nodes/immunology
- Lymph Nodes/pathology
- Lymphocyte Activation
- Membrane Proteins/genetics
- Membrane Proteins/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mitogen-Activated Protein Kinase 1/genetics
- Mitogen-Activated Protein Kinase 1/immunology
- Mitogen-Activated Protein Kinase 3/genetics
- Mitogen-Activated Protein Kinase 3/immunology
- Myelin-Oligodendrocyte Glycoprotein/administration & dosage
- Peptide Fragments/administration & dosage
- Phospholipase C gamma/genetics
- Phospholipase C gamma/immunology
- Phosphoproteins/genetics
- Phosphoproteins/immunology
- Primary Cell Culture
- Protein Serine-Threonine Kinases/deficiency
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/immunology
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Signal Transduction
- Spleen/immunology
- Spleen/pathology
- T-Box Domain Proteins/genetics
- T-Box Domain Proteins/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/pathology
- ZAP-70 Protein-Tyrosine Kinase/genetics
- ZAP-70 Protein-Tyrosine Kinase/immunology
- NF-kappaB-Inducing Kinase
Collapse
Affiliation(s)
- Sonja M Lacher
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Christoph Thurm
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology, and Inflammation, Otto von Guericke University, Magdeburg, Germany
| | - Ute Distler
- Institute for Immunology, University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
| | - Alma N Mohebiany
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Nicole Israel
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Maja Kitic
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Anna Ebering
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Yilang Tang
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Matthias Klein
- Institute for Immunology, University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
| | - Guido H Wabnitz
- Institute of Immunology, Section Molecular Immunology, Ruprecht-Karls-University, Heidelberg, Germany
| | - Florian Wanke
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Yvonne Samstag
- Institute of Immunology, Section Molecular Immunology, Ruprecht-Karls-University, Heidelberg, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
| | - Florian C Kurschus
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Luca Simeoni
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology, and Inflammation, Otto von Guericke University, Magdeburg, Germany
| | - Stefan Tenzer
- Institute for Immunology, University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.
| |
Collapse
|
35
|
Boyce BF, Li J, Xing L, Yao Z. Bone Remodeling and the Role of TRAF3 in Osteoclastic Bone Resorption. Front Immunol 2018; 9:2263. [PMID: 30323820 PMCID: PMC6172306 DOI: 10.3389/fimmu.2018.02263] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 09/11/2018] [Indexed: 02/05/2023] Open
Abstract
Skeletal health is maintained by bone remodeling, a process in which microscopic sites of effete or damaged bone are degraded on bone surfaces by osteoclasts and subsequently replaced by new bone, which is laid down by osteoblasts. This normal process can be disturbed in a variety of pathologic processes, including localized or generalized inflammation, metabolic and endocrine disorders, primary and metastatic cancers, and during aging as a result of low-grade chronic inflammation. Osteoclast formation and activity are promoted by factors, including cytokines, hormones, growth factors, and free radicals, and require expression of macrophage-colony stimulating factor (M-CSF) and receptor activator of NF-κB ligand (RANKL) by accessory cells in the bone marrow, including osteoblastic and immune cells. Expression of TNF receptor-associated factor 6 (TRAF6) is required in osteoclast precursors to mediate RANKL-induced activation of NF-κB, which is also necessary for osteoclast formation and activity. TRAF3, in contrast is not required for osteoclast formation, but it limits RANKL-induced osteoclast formation by promoting proteasomal degradation of NF-κB-inducing kinase in a complex with TRAF2 and cellular inhibitor of apoptosis proteins (cIAP). TRAF3 also limits osteoclast formation induced by TNF, which mediates inflammation and joint destruction in inflammatory diseases, including rheumatoid arthritis. Chloroquine and hydroxychloroquine, anti-inflammatory drugs used to treat rheumatoid arthritis, prevent TRAF3 degradation in osteoclast precursors and inhibit osteoclast formation in vitro. Chloroquine also inhibits bone destruction induced by ovariectomy and parathyroid hormone in mice in vivo. Mice genetically engineered to have TRAF3 deleted in osteoclast precursors and macrophages develop early onset osteoporosis, inflammation in multiple tissues, infections, and tumors, indicating that TRAF3 suppresses inflammation and tumors in myeloid cells. Mice with TRAF3 conditionally deleted in mesenchymal cells also develop early onset osteoporosis due to a combination of increased osteoclast formation and reduced osteoblast formation. TRAF3 protein levels decrease in bone and bone marrow during aging in mice and humans. Development of drugs to prevent TRAF3 degradation in immune and bone cells could be a novel therapeutic approach to prevent or reduce bone loss and the incidence of several common diseases associated with aging.
Collapse
Affiliation(s)
- Brendan F. Boyce
- Department of Pathology and Laboratory Medicine, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| | | | | | | |
Collapse
|
36
|
Bishop GA, Stunz LL, Hostager BS. TRAF3 as a Multifaceted Regulator of B Lymphocyte Survival and Activation. Front Immunol 2018; 9:2161. [PMID: 30319624 PMCID: PMC6165887 DOI: 10.3389/fimmu.2018.02161] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 09/03/2018] [Indexed: 12/20/2022] Open
Abstract
The adaptor protein TNF receptor-associated factor 3 (TRAF3) serves as a powerful negative regulator in multiple aspects of B cell biology. Early in vitro studies in transformed cell lines suggested the potential of TRAF3 to inhibit signaling by its first identified binding receptor, CD40. However, because the canonical TRAF3 binding site on many receptors also mediates binding of other TRAFs, and whole-mouse TRAF3 deficiency is neonatally lethal, an accurate understanding of TRAF3's specific functions was delayed until conditional TRAF3-deficient mice were produced. Studies of B cell-specific TRAF3-deficient mice, complemented by investigations in normal and malignant mouse and human B cells, reveal that TRAF3 has powerful regulatory roles that are unique to this TRAF, as well as functions context-specific to the B cell. This review summarizes the current state of knowledge of these roles and functions. These include inhibition of signaling by plasma membrane receptors, negative regulation of intracellular receptors, and restraint of cytoplasmic NF- κB pathways. TRAF3 is also now known to function as a resident nuclear protein, and to impact B cell metabolism. Through these and additional mechanisms TRAF3 exerts powerful restraint upon B cell survival and activation. It is thus perhaps not surprising that TRAF3 has been revealed as an important tumor suppressor in B cells. The many and varied functions of TRAF3 in B cells, and new directions to pursue in future studies, are summarized and discussed here.
Collapse
Affiliation(s)
- Gail A. Bishop
- Department of Microbiology & Immunology, University of Iowa, Iowa City, IA, United States
- Department of Internal Medicine, University of Iowa, Iowa City, IA, United States
- Iowa City VA Health Care System, Iowa City, Iowa City, IA, United States
| | - Laura L. Stunz
- Department of Microbiology & Immunology, University of Iowa, Iowa City, IA, United States
| | - Bruce S. Hostager
- Department of Microbiology & Immunology, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
37
|
NF-κB pathways in the development and progression of colorectal cancer. Transl Res 2018; 197:43-56. [PMID: 29550444 DOI: 10.1016/j.trsl.2018.02.002] [Citation(s) in RCA: 184] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 02/13/2018] [Accepted: 02/14/2018] [Indexed: 12/12/2022]
Abstract
Nuclear factor-κB (NF-κB) has been widely implicated in the development and progression of cancer. In colorectal cancer (CRC), NF-κB has a key role in cancer-related processes such as cell proliferation, apoptosis, angiogenesis, and metastasis. The role of NF-κB in CRC is complex, owed to the cross talk with other signaling pathways. Although there is sufficient evidence gained from cell lines and animal models that NF-κB is involved in cancer-related processes, because of a lack of studies in human tissue, the clinical evidence of its importance is limited in patients with CRC. This review summarizes evidence relating to how NF-κB is involved in the development and progression of CRC and comments on future work to be carried out.
Collapse
|
38
|
Roy P, Sarkar UA, Basak S. The NF-κB Activating Pathways in Multiple Myeloma. Biomedicines 2018; 6:biomedicines6020059. [PMID: 29772694 PMCID: PMC6027071 DOI: 10.3390/biomedicines6020059] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/14/2018] [Accepted: 05/14/2018] [Indexed: 12/29/2022] Open
Abstract
Multiple myeloma(MM), an incurable plasma cell cancer, represents the second most prevalent hematological malignancy. Deregulated activity of the nuclear factor kappaB (NF-κB) family of transcription factors has been implicated in the pathogenesis of multiple myeloma. Tumor microenvironment-derived cytokines and cancer-associated genetic mutations signal through the canonical as well as the non-canonical arms to activate the NF-κB system in myeloma cells. In fact, frequent engagement of both the NF-κB pathways constitutes a distinguishing characteristic of myeloma. In turn, NF-κB signaling promotes proliferation, survival and drug-resistance of myeloma cells. In this review article, we catalog NF-κB activating genetic mutations and microenvironmental cues associated with multiple myeloma. We then describe how the individual canonical and non-canonical pathways transduce signals and contribute towards NF-κB -driven gene-expressions in healthy and malignant cells. Furthermore, we discuss signaling crosstalk between concomitantly triggered NF-κB pathways, and its plausible implication for anomalous NF-κB activation and NF-κB driven pro-survival gene-expressions in multiple myeloma. Finally, we propose that mechanistic understanding of NF-κB deregulations may provide for improved therapeutic and prognostic tools in multiple myeloma.
Collapse
Affiliation(s)
- Payel Roy
- Systems Immunology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India.
| | - Uday Aditya Sarkar
- Systems Immunology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India.
| | - Soumen Basak
- Systems Immunology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India.
| |
Collapse
|
39
|
Prossomariti A, Sokol H, Ricciardiello L. Nucleotide-Binding Domain Leucine-Rich Repeat Containing Proteins and Intestinal Microbiota: Pivotal Players in Colitis and Colitis-Associated Cancer Development. Front Immunol 2018; 9:1039. [PMID: 29868004 PMCID: PMC5960679 DOI: 10.3389/fimmu.2018.01039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 04/26/2018] [Indexed: 12/14/2022] Open
Abstract
The nucleotide-binding domain leucine-rich repeat containing (NLR) proteins play a fundamental role in innate immunity and intestinal tissue repair. A dysbiotic intestinal microbiota, developed as a consequence of alterations in NLR proteins, has recently emerged as a crucial hit for the development of ulcerative colitis (UC) and colitis-associated cancer (CAC). The concept of the existence of functional axes interconnecting bacteria with NLR proteins in a causal role in intestinal inflammation and CAC aroused a great interest for the potential development of preventive and therapeutic strategies against UC and CAC. However, the most recent scientific evidence, which highlights many confounding factors in studies based on microbiota characterization, underlines the need for an in-depth reconsideration of the data obtained until now. The purpose of this review is to discuss the recent findings concerning the cross talk between the NLR signaling and the intestinal microbiota in UC and CAC development, and to highlight the open issues that should be explored and addressed in future studies.
Collapse
Affiliation(s)
- Anna Prossomariti
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy.,Center for Applied Biomedical Research (CRBA), S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Harry Sokol
- Sorbonne Université, École normale supérieure, PSL Research University, CNRS, INSERM, AP-HP, Hôpital Saint-Antoine, Laboratoire de biomolécules, LBM, Paris, France.,INRA, UMR1319 Micalis & AgroParisTech, Jouy en Josas, France
| | - Luigi Ricciardiello
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
40
|
Zhao P, Wong KI, Sun X, Reilly SM, Uhm M, Liao Z, Skorobogatko Y, Saltiel AR. TBK1 at the Crossroads of Inflammation and Energy Homeostasis in Adipose Tissue. Cell 2018; 172:731-743.e12. [PMID: 29425491 PMCID: PMC5808582 DOI: 10.1016/j.cell.2018.01.007] [Citation(s) in RCA: 207] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 11/17/2017] [Accepted: 01/04/2018] [Indexed: 12/22/2022]
Abstract
The noncanonical IKK family member TANK-binding kinase 1 (TBK1) is activated by pro-inflammatory cytokines, but its role in controlling metabolism remains unclear. Here, we report that the kinase uniquely controls energy metabolism. Tbk1 expression is increased in adipocytes of HFD-fed mice. Adipocyte-specific TBK1 knockout (ATKO) attenuates HFD-induced obesity by increasing energy expenditure; further studies show that TBK1 directly inhibits AMPK to repress respiration and increase energy storage. Conversely, activation of AMPK under catabolic conditions can increase TBK1 activity through phosphorylation, mediated by AMPK's downstream target ULK1. Surprisingly, ATKO also exaggerates adipose tissue inflammation and insulin resistance. TBK1 suppresses inflammation by phosphorylating and inducing the degradation of the IKK kinase NIK, thus attenuating NF-κB activity. Moreover, TBK1 mediates the negative impact of AMPK activity on NF-κB activation. These data implicate a unique role for TBK1 in mediating bidirectional crosstalk between energy sensing and inflammatory signaling pathways in both over- and undernutrition.
Collapse
Affiliation(s)
- Peng Zhao
- Division of Metabolism and Endocrinology, Department of Medicine, University of California-San Diego, La Jolla, CA 92093, USA; Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kai In Wong
- Division of Metabolism and Endocrinology, Department of Medicine, University of California-San Diego, La Jolla, CA 92093, USA
| | - Xiaoli Sun
- Division of Metabolism and Endocrinology, Department of Medicine, University of California-San Diego, La Jolla, CA 92093, USA
| | - Shannon M Reilly
- Division of Metabolism and Endocrinology, Department of Medicine, University of California-San Diego, La Jolla, CA 92093, USA; Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Maeran Uhm
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Zhongji Liao
- Division of Metabolism and Endocrinology, Department of Medicine, University of California-San Diego, La Jolla, CA 92093, USA
| | - Yuliya Skorobogatko
- Division of Metabolism and Endocrinology, Department of Medicine, University of California-San Diego, La Jolla, CA 92093, USA; Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Alan R Saltiel
- Division of Metabolism and Endocrinology, Department of Medicine, University of California-San Diego, La Jolla, CA 92093, USA; Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
41
|
Rothschild DE, McDaniel DK, Ringel-Scaia VM, Allen IC. Modulating inflammation through the negative regulation of NF-κB signaling. J Leukoc Biol 2018; 103:10.1002/JLB.3MIR0817-346RRR. [PMID: 29389019 PMCID: PMC6135699 DOI: 10.1002/jlb.3mir0817-346rrr] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 12/21/2017] [Accepted: 12/21/2017] [Indexed: 12/16/2022] Open
Abstract
Immune system activation is essential to thwart the invasion of pathogens and respond appropriately to tissue damage. However, uncontrolled inflammation can result in extensive collateral damage underlying a diverse range of auto-inflammatory, hyper-inflammatory, and neoplastic diseases. The NF-κB signaling pathway lies at the heart of the immune system and functions as a master regulator of gene transcription. Thus, this signaling cascade is heavily targeted by mechanisms designed to attenuate overzealous inflammation and promote resolution. Mechanisms associated with the negative regulation of NF-κB signaling are currently under intense investigation and have yet to be fully elucidated. Here, we provide an overview of mechanisms that negatively regulate NF-κB signaling through either attenuation of signal transduction, inhibition of posttranscriptional signaling, or interference with posttranslational modifications of key pathway components. While the regulators discussed for each group are far from comprehensive, they exemplify common mechanistic approaches that inhibit this critical biochemical signaling cascade. Despite their diversity, a commonality among these regulators is their selection of specific targets at key inflection points in the pathway, such as TNF-receptor-associated factor family members or essential kinases. A better understanding of these negative regulatory mechanisms will be essential to gain greater insight related to the maintenance of immune system homeostasis and inflammation resolution. These processes are vital elements of disease pathology and have important implications for targeted therapeutic strategies.
Collapse
Affiliation(s)
- Daniel E. Rothschild
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg VA 24061
| | - Dylan K. McDaniel
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg VA 24061
| | - Veronica M. Ringel-Scaia
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA 24061
| | - Irving C. Allen
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg VA 24061
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA 24061
- Department of Basic Science Education, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016
| |
Collapse
|
42
|
Schweighoffer E, Tybulewicz VL. Signalling for B cell survival. Curr Opin Cell Biol 2017; 51:8-14. [PMID: 29149682 DOI: 10.1016/j.ceb.2017.10.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 10/10/2017] [Indexed: 12/21/2022]
Abstract
The number of mature B cells is carefully controlled by signalling from receptors that support B cell survival. The best studied of these are the B cell antigen receptor (BCR) and BAFFR. Recent work has shown that signalling from these receptors is closely linked, involves the CD19 co-receptor, and leads to activation of canonical and non-canonical NF-κB pathways, ERK1, ERK2 and ERK5 MAP kinases, and PI-3 kinases. Importantly, studies show that investigation of the importance of signalling molecules in cell survival requires the use of inducible gene deletions within mature B cells. This overcomes the limitations of many earlier studies using constitutive gene deletions which were unable to distinguish between requirements for a protein in development versus survival.
Collapse
Affiliation(s)
| | - Victor Lj Tybulewicz
- The Francis Crick Institute, London NW1 1AT, UK; Imperial College, London W12 0NN, UK.
| |
Collapse
|
43
|
The non-canonical NF-κB pathway in immunity and inflammation. NATURE REVIEWS. IMMUNOLOGY 2017. [PMID: 28580957 DOI: 10.1038/nri.2017.52)] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
The nuclear factor-κB (NF-κB) family of transcription factors is activated by canonical and non-canonical signalling pathways, which differ in both signalling components and biological functions. Recent studies have revealed important roles for the non-canonical NF-κB pathway in regulating different aspects of immune functions. Defects in non-canonical NF-κB signalling are associated with severe immune deficiencies, whereas dysregulated activation of this pathway contributes to the pathogenesis of various autoimmune and inflammatory diseases. Here we review the signalling mechanisms and the biological function of the non-canonical NF-κB pathway. We also discuss recent progress in elucidating the molecular mechanisms regulating non-canonical NF-κB pathway activation, which may provide new opportunities for therapeutic strategies.
Collapse
|
44
|
Giampazolias E, Zunino B, Dhayade S, Bock F, Cloix C, Cao K, Roca A, Lopez J, Ichim G, Proïcs E, Rubio-Patiño C, Fort L, Yatim N, Woodham E, Orozco S, Taraborrelli L, Peltzer N, Lecis D, Machesky L, Walczak H, Albert ML, Milling S, Oberst A, Ricci JE, Ryan KM, Blyth K, Tait SW. Mitochondrial permeabilization engages NF-κB-dependent anti-tumour activity under caspase deficiency. Nat Cell Biol 2017; 19:1116-1129. [PMID: 28846096 PMCID: PMC5624512 DOI: 10.1038/ncb3596] [Citation(s) in RCA: 199] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 07/20/2017] [Indexed: 12/14/2022]
Abstract
Apoptosis represents a key anti-cancer therapeutic effector mechanism. During apoptosis, mitochondrial outer membrane permeabilization (MOMP) typically kills cells even in the absence of caspase activity. Caspase activity can also have a variety of unwanted consequences that include DNA damage. We therefore investigated whether MOMP-induced caspase-independent cell death (CICD) might be a better way to kill cancer cells. We find that cells undergoing CICD display potent pro-inflammatory effects relative to apoptosis. Underlying this, MOMP was found to stimulate NF-κB activity through the downregulation of inhibitor of apoptosis proteins. Strikingly, engagement of CICD displays potent anti-tumorigenic effects, often promoting complete tumour regression in a manner dependent on intact immunity. Our data demonstrate that by activating NF-κB, MOMP can exert additional signalling functions besides triggering cell death. Moreover, they support a rationale for engaging caspase-independent cell death in cell-killing anti-cancer therapies.
Collapse
Affiliation(s)
- Evangelos Giampazolias
- Cancer Research UK Beatson Institute
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1BD, U.K
| | | | | | - Florian Bock
- Cancer Research UK Beatson Institute
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1BD, U.K
| | - Catherine Cloix
- Cancer Research UK Beatson Institute
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1BD, U.K
| | - Kai Cao
- Cancer Research UK Beatson Institute
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1BD, U.K
| | - Alba Roca
- Cancer Research UK Beatson Institute
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1BD, U.K
| | - Jonathan Lopez
- Cancer Research UK Beatson Institute
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1BD, U.K
| | - Gabriel Ichim
- Cancer Research UK Beatson Institute
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1BD, U.K
| | | | | | - Loic Fort
- Cancer Research UK Beatson Institute
| | - Nader Yatim
- Laboratory of Dendritic Cell Biology, Department of Immunology, Institut Pasteur, 25 Rue du Docteur Roux, 75015 Paris, France
| | | | - Susana Orozco
- Molecular & Cellular Biology Program and Dept. of Immunology, University of Washington, 750 Republican St., Seattle, WA 981, U.S.A
| | - Lucia Taraborrelli
- Centre for Cell Death, Cancer and Inflammation, UCL Cancer Institute, UCL, London WC1E 6BT, UK
| | - Nieves Peltzer
- Centre for Cell Death, Cancer and Inflammation, UCL Cancer Institute, UCL, London WC1E 6BT, UK
| | - Daniele Lecis
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy
| | | | - Henning Walczak
- Centre for Cell Death, Cancer and Inflammation, UCL Cancer Institute, UCL, London WC1E 6BT, UK
| | - Matthew L. Albert
- Laboratory of Dendritic Cell Biology, Department of Immunology, Institut Pasteur, 25 Rue du Docteur Roux, 75015 Paris, France
- Department of Cancer Immunology, Genentech Inc., 1 DNA way, South San Francisco, California 94080, USA
| | - Simon Milling
- Centre for Immunobiology, Institute for Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, U.K
| | - Andrew Oberst
- Molecular & Cellular Biology Program and Dept. of Immunology, University of Washington, 750 Republican St., Seattle, WA 981, U.S.A
| | | | | | | | - Stephen W.G. Tait
- Cancer Research UK Beatson Institute
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1BD, U.K
| |
Collapse
|
45
|
Interference of Apoptosis by Hepatitis B Virus. Viruses 2017; 9:v9080230. [PMID: 28820498 PMCID: PMC5580487 DOI: 10.3390/v9080230] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/07/2017] [Accepted: 08/10/2017] [Indexed: 12/16/2022] Open
Abstract
Hepatitis B virus (HBV) causes liver diseases that have been a consistent problem for human health, leading to more than one million deaths every year worldwide. A large proportion of hepatocellular carcinoma (HCC) cases across the world are closely associated with chronic HBV infection. Apoptosis is a programmed cell death and is frequently altered in cancer development. HBV infection interferes with the apoptosis signaling to promote HCC progression and viral proliferation. The HBV-mediated alteration of apoptosis is achieved via interference with cellular signaling pathways and regulation of epigenetics. HBV X protein (HBX) plays a major role in the interference of apoptosis. There are conflicting reports on the HBV interference of apoptosis with the majority showing inhibition of and the rest reporting induction of apoptosis. In this review, we described recent studies on the mechanisms of the HBV interference with the apoptosis signaling during the virus infection and provided perspective.
Collapse
|
46
|
Abstract
The nuclear factor-κB (NF-κB) family of transcription factors is activated by canonical and non-canonical signalling pathways, which differ in both signalling components and biological functions. Recent studies have revealed important roles for the non-canonical NF-κB pathway in regulating different aspects of immune functions. Defects in non-canonical NF-κB signalling are associated with severe immune deficiencies, whereas dysregulated activation of this pathway contributes to the pathogenesis of various autoimmune and inflammatory diseases. Here we review the signalling mechanisms and the biological function of the non-canonical NF-κB pathway. We also discuss recent progress in elucidating the molecular mechanisms regulating non-canonical NF-κB pathway activation, which may provide new opportunities for therapeutic strategies.
Collapse
Affiliation(s)
- Shao-Cong Sun
- Department of Immunology, The University of Texas MD Anderson Cancer Center, MD Anderson Cancer Center UT Heath Graduate School of Biomedical Sciences, 7455 Fannin Street, Box 902, Houston, Texas 77030, USA
| |
Collapse
|
47
|
Zemankova N, Chlebova K, Matiasovic J, Prodelalova J, Gebauer J, Faldyna M. Bovine lactoferrin free of lipopolysaccharide can induce a proinflammatory response of macrophages. BMC Vet Res 2016; 12:251. [PMID: 27829421 PMCID: PMC5103330 DOI: 10.1186/s12917-016-0878-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 11/02/2016] [Indexed: 12/12/2022] Open
Abstract
Background Lactoferrin (LF) is an 80 kDa glycoprotein which is known for its effects against bacteria, viruses and other pathogens. It also has a high potential in nutrition therapy and welfare of people and a variety of animals, including piglets. The ability to bind lipopolysaccharide (LPS) is one of the described anti-inflammatory mechanisms of LF. Previous studies suggested that cells can be stimulated even by LPS-free LF. Therefore, the aim of our study was to bring additional information about this possibility. Porcine monocyte derived macrophages (MDMF) and human embryonic kidney (HEK) cells were stimulated with unpurified LF in complex with LPS and with purified LF without bound LPS. Results Both cell types were stimulated with unpurified as well as purified LF. On the other hand, neither HEK0 cells not expressing any TLR nor HEK4a cells transfected with TLR4 produced any pro-inflammatory cytokine transcripts after stimulation with purified LF. This suggests that purified LF without LPS stimulates cells via another receptor than TLR4. An alternative, TLR4-independent, pathway was further confirmed by analyses of the NF-kappa-B-inducing kinase (NIK) activation. Western blot analyses showed NIK which activates different NFκB subunits compared to LF-LPS signaling via TLR4. Though, this confirmed an alternative pathway which is used by the purified LF free of LPS. This stimulation of MDMF led to low, but significant amounts of pro-inflammatory cytokines, which can be considered as a positive stimulation of the immune system. Conclusion Our results suggest that LF’s ability is not only to bind LPS, but LF itself may be a stimulant of pro-inflammatory pathways.
Collapse
Affiliation(s)
- Nada Zemankova
- Veterinary Research Institute, Hudcova 296/70, Brno, 621 00, Czech Republic.,Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Katarina Chlebova
- Veterinary Research Institute, Hudcova 296/70, Brno, 621 00, Czech Republic.,Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Jan Matiasovic
- Veterinary Research Institute, Hudcova 296/70, Brno, 621 00, Czech Republic
| | - Jana Prodelalova
- Veterinary Research Institute, Hudcova 296/70, Brno, 621 00, Czech Republic
| | - Jan Gebauer
- Veterinary Research Institute, Hudcova 296/70, Brno, 621 00, Czech Republic.,Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Martin Faldyna
- Veterinary Research Institute, Hudcova 296/70, Brno, 621 00, Czech Republic.
| |
Collapse
|
48
|
Transcription factors of the alternative NF-κB pathway are required for germinal center B-cell development. Proc Natl Acad Sci U S A 2016; 113:9063-8. [PMID: 27457956 DOI: 10.1073/pnas.1602728113] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The NF-κB signaling cascade relays external signals essential for B-cell growth and survival. This cascade is frequently hijacked by cancers that arise from the malignant transformation of germinal center (GC) B cells, underscoring the importance of deciphering the function of NF-κB in these cells. The NF-κB signaling cascade is comprised of two branches, the canonical and alternative NF-κB pathways, mediated by distinct transcription factors. The expression and function of the transcription factors of the alternative pathway, RELB and NF-κB2, in late B-cell development is incompletely understood. Using conditional deletion of relb and nfkb2 in GC B cells, we here report that ablation of both RELB and NF-κB2, but not of the single transcription factors, resulted in the collapse of established GCs. RELB/NF-κB2 deficiency in GC B cells was associated with impaired cell-cycle entry and reduced expression of the cell-surface receptor inducible T-cell costimulator ligand that promotes optimal interactions between B and T cells. Analysis of human tonsillar tissue revealed that plasma cells and their precursors in the GC expressed high levels of NF-κB2 relative to surrounding lymphocytes. Accordingly, deletion of nfkb2 in murine GC B cells resulted in a dramatic reduction of antigen-specific antibody-secreting cells, whereas deletion of relb had no effect. These results demonstrate that the transcription factors of the alternative NF-κB pathway control distinct stages of late B-cell development, which may have implications for B-cell malignancies that aberrantly activate this pathway.
Collapse
|
49
|
Abstract
The signaling adapter protein tumor necrosis factor receptor (TNFR)-associated factor 3 (TRAF3) is both modified by and contributes to several types of ubiquitination events. TRAF3 plays a variety of context-dependent regulatory roles in all types of immune cells. In B lymphocytes, TRAF3 contributes to regulation of signaling by members of both the TNFR superfamily and innate immune receptors. TRAF3 also plays a unique cell type-specific and critical role in the restraint of B-cell homeostatic survival, a role with important implications for both B-cell differentiation and the pathogenesis of B-cell malignancies. This review focuses upon the relationship between ubiquitin and TRAF3, and how this contributes to multiple functions of TRAF3 in the regulation of signal transduction, transcriptional activation, and effector functions of B lymphocytes.
Collapse
Affiliation(s)
- Wai W Lin
- The Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA
| | - Bruce S Hostager
- Department of Pediatrics, University of Iowa, Iowa City, IA, USA
| | - Gail A Bishop
- The Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA.,Department of Microbiology, University of Iowa, Iowa City, IA, USA.,Department of Internal Medicine, University of Iowa, Iowa City, IA, USA.,VA Medical Center, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
50
|
Macrophage TCF-4 co-activates p65 to potentiate chronic inflammation and insulin resistance in mice. Clin Sci (Lond) 2016; 130:1257-68. [PMID: 27129186 DOI: 10.1042/cs20160192] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 04/26/2016] [Indexed: 12/29/2022]
Abstract
Transcription factor 4 (TCF-4) was recently identified as a candidate gene for the cause of type 2 diabetes, although the mechanisms have not been fully elucidated. In the present study, we demonstrated that the TCF-4 transgene in macrophages aggravated high-fat diet (HFD)-induced insulin resistance and chronic inflammation, characterized by the elevation of proinflammatory cytokines in the blood, liver and white adipose tissue, as well as a proinflammatory profile of immune cells in visceral fats in mice. Mechanistically, TCF-4 functioned as a co-activator of p65 to amplify the saturated free fatty acid (FFA)-stimulated promoter activity, mRNA transcription and secretion of proinflammatory cytokines in primary macrophages. Blockage of p65 with a specific interfering RNA or inhibitor could prevent TCF-4-enhanced expression of proinflammatory cytokines in FFA/lipopolysaccharide-treated primary macrophages. The p65 inhibitor could abolish macrophage TCF-4 transgene-aggravated systemic inflammation, glucose intolerance and insulin resistance in HFD-treated mice. In addition, we demonstrated that the mRNA expression of TCF-4 in the peripheral blood monocytes from humans was positively correlated to the levels of interleukin (IL)-1β, tumour necrosis factor α, IL-6 and fasting plasma glucose. In summary, we identified TCF-4 as a co-activator of p65 in the potentiation of proinflammatory cytokine production in macrophages and aggravation of HFD-induced chronic inflammation and insulin resistance in mice.
Collapse
|