1
|
Morshedi B, Esfandyari-Manesh M, Atyabi F, Ghahremani MH, Dinarvand R. Local delivery of ibrutinib by folate receptor-mediated targeting PLGA-PEG nanoparticles to glioblastoma multiform: in vitro and in vivo studies. J Drug Target 2025; 33:1026-1041. [PMID: 39960788 DOI: 10.1080/1061186x.2025.2468749] [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: 10/24/2024] [Revised: 01/19/2025] [Accepted: 02/09/2025] [Indexed: 02/25/2025]
Abstract
Glioblastoma multiforme (GBM) is a widespread and life-threatening kind of brain cancer, which has a high mortality rate. Ibrutinib, a Bruton's tyrosine kinase (BTK) inhibitor, irreversibly adheres to a conserved cysteine residue of two enzymes BTK and BMX, inhibiting their kinase activities, which leads to suppression of the growth of glioma cells. This study synthesised PLGA-PEG-folate (PPF) polymer and subsequently encapsulated ibrutinib within PPF nanoparticles (IBT-PPF-NPs). H NMR spectra confirmed the synthesis of PPF polymer. The efficiency of IBT-PPF-NPs was 97 ± 2.26% with 8.8 ± 0.2% drug loading. The particle size was 208 ± 4.8 nm. The IC50 value of free ibrutinib, IB-PPF-NPs and ibrutinib encapsulated in PLGA NPs (IB-P-NPs) was 10.2, 7.6 and 10.13 µM in C6 cell lines, whereas in U-87 MG cells was 24.4, 16 and 25.2 µM, respectively. The cellular uptake of FITC-PPF-NPs increased from 47.6% to 90.3% in C6 cells and from 55% to 97.3% in U-87 MG cells compared to FITC-P-NPs. The in vivo results indicate a significant reduction in tumour size in treatment groups in comparison to control groups, while the group that received the intratumoural injection of IB-PPF-NPs exhibited a greater reduction. The folate-targeting agent enhances the nanoparticles' effectiveness by promoting their uptake through the endocytosis pathway.
Collapse
Affiliation(s)
- Bahar Morshedi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Esfandyari-Manesh
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Atyabi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Ghahremani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Rassoul Dinarvand
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Song X, Tiek D, Lu M, Yu X, Wu R, Walker M, He Q, Sisbarro D, Hu B, Cheng SY. A Single-Cell Atlas of RNA Alternative Splicing in the Glioma-Immune Ecosystem. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.26.645511. [PMID: 40196477 PMCID: PMC11974875 DOI: 10.1101/2025.03.26.645511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Single-cell analysis has refined our understanding of cellular heterogeneity in glioma, yet RNA alternative splicing (AS)-a critical layer of transcriptome regulation-remains underexplored at single-cell resolution. Here, we present a pan-glioma single-cell AS analysis in both tumor and immune cells through integrating seven SMART-seq2 datasets of human gliomas. Our analysis reveals lineage-specific AS across glioma cellular states, with the most divergent AS landscapes between mesenchymal- and neuronal-like glioma cells, exemplified by AS in TCF12 and PTBP2. Comparison between core and peripheral glioma cells highlights AS-redox co-regulation of cytoskeleton organization. Further analysis of glioma-infiltrating immune cells reveals potential isoform-level regulation of protein glycosylation in regulatory T cells and a link between MS4A7 AS in macrophages and clinical response to anti-PD-1 therapy. This study emphasizes the role of AS in glioma cellular heterogeneity, highlighting the importance of an isoform-centric approach to better understand the complex biological processes driving tumorigenesis.
Collapse
Affiliation(s)
- Xiao Song
- The Ken & Ruth Davee Department of Neurology, The Lou and Jean Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Deanna Tiek
- The Ken & Ruth Davee Department of Neurology, The Lou and Jean Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Minghui Lu
- The Ken & Ruth Davee Department of Neurology, The Lou and Jean Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Xiaozhou Yu
- The Ken & Ruth Davee Department of Neurology, The Lou and Jean Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Runxin Wu
- The Ken & Ruth Davee Department of Neurology, The Lou and Jean Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Maya Walker
- The Ken & Ruth Davee Department of Neurology, The Lou and Jean Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Qiu He
- The Ken & Ruth Davee Department of Neurology, The Lou and Jean Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Derek Sisbarro
- The Ken & Ruth Davee Department of Neurology, The Lou and Jean Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Bo Hu
- The Ken & Ruth Davee Department of Neurology, The Lou and Jean Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Shi-Yuan Cheng
- The Ken & Ruth Davee Department of Neurology, The Lou and Jean Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
3
|
Zhao Y, Liang X, Wei R, Guo F, Qin G, Yu H, Liu J, Xia W, Gou S, Wu H, Zhou Y. BAP1 Represses Sequential Activation of IRAKs and NF-κB Signaling in Pancreatic Cancer. Int J Biol Sci 2025; 21:1949-1965. [PMID: 40083694 PMCID: PMC11900812 DOI: 10.7150/ijbs.104977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 02/05/2025] [Indexed: 03/16/2025] Open
Abstract
The deubiquitinating enzyme BRCA1 Associated Protein-1 (BAP1) has been reported to be shallowly deleted in a subset of pancreatic ductal adenocarcinomas (PDAC) and is believed to play a significant role in the development of chronic pancreatitis-driven PDAC. However, evidence suggests that BAP1 may also be involved in the progression and metastasis of PDAC, though the underlying mechanism remains unclear. Here, we demonstrate that BAP1 deletion leads to the overactivation of the nuclear factor-κB (NF-κB) signaling in PDAC, thereby promoting the proliferation, migration, and invasion of PDAC models both in vivo and in vitro. Mechanistically, BAP1 inhibits the sequential activation of interleukin-1 receptor-associated kinases (IRAKs) in an enzyme-independent manner. BAP1 binds to IRAK1 and inhibits the interaction between IRAK4 and IRAK1, as well as the IRAK4-mediated initiation of IRAK1 phosphorylation and autophosphorylation. This, in turn, prevents the dissociation of IRAK1 from the Myddosome complex and sequential activation of NF-κB. Based on this, we further identified that dual-target inhibitors of IRAK1/4 exhibited significant inhibitory effects on BAP1-deficient tumors in both in vivo and in vitro PDAC models. Our findings elucidate the mechanism by which BAP1 inhibits the NF-κB signaling and present a promising strategy for the targeted treatment of BAP1-deficient pancreatic cancer.
Collapse
Affiliation(s)
- Yuhan Zhao
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 Hubei China
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Xueyi Liang
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 Hubei China
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Ruozheng Wei
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 Hubei China
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Feng Guo
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 Hubei China
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Gengdu Qin
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 Hubei China
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Haixin Yu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Jiaying Liu
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wentao Xia
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 Hubei China
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Shanmiao Gou
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 Hubei China
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Heshui Wu
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 Hubei China
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Yingke Zhou
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 Hubei China
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| |
Collapse
|
4
|
Mao W, Xu K, Wang K, Zhang H, Ji J, Geng J, Sun S, Gu C, Bhattacharya A, Fang C, Tao T, Chen M, Wu J, Chen S, Sun C, Xu B. Single-cell RNA sequencing and spatial transcriptomics of bladder Ewing sarcoma. iScience 2024; 27:110921. [PMID: 39386756 PMCID: PMC11462044 DOI: 10.1016/j.isci.2024.110921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 07/15/2024] [Accepted: 09/06/2024] [Indexed: 10/12/2024] Open
Abstract
Bladder Ewing sarcoma/primitive neuroectodermal tumor (bladder ES/PNET) is a rare and highly malignant tumor associated with a poor prognosis, yet its underlying mechanisms remain poorly understood. Here, we employed a combination of single-cell RNA sequencing (scRNA-seq), spatial transcriptomics (ST), and functional analyses to delve into the pathogenesis of bladder ES/PNET. The investigation revealed the presence of specialized types of epithelial cells (referred to as bladder ES-Epi) and mast cells (referred to as bladder ES-Mast) within bladder ES/PNET in comparison to urothelial carcinoma. Notably, TNFRSF12A exhibited significant upregulation in bladder ES/PNET. Furthermore, mast cells possessed the ability to activate epithelial cells through the TNFSF12-TNFRSF12A ligand-receptor signaling pattern. In addition, Enavatuzumab can significantly inhibit the migratory ability of the Ewing sarcoma cell line RD-ES. This groundbreaking study provides unprecedented mechanistic insights into the progression of bladder ES/PNET and introduces a potential therapeutic avenue for treating this challenging malignancy.
Collapse
Affiliation(s)
- Weipu Mao
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing 210009, China
| | - Kangjie Xu
- Central Laboratory Department, Binhai County People’s Hospital, Yancheng 224000, China
| | - Keyi Wang
- Department of Urology, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Houliang Zhang
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing 210009, China
| | - Jie Ji
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing 210009, China
| | - Jiang Geng
- Department of Urology, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Si Sun
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing 210009, China
| | - Chaoming Gu
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing 210009, China
| | - Atrayee Bhattacharya
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Cheng Fang
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing 210009, China
| | - Tao Tao
- Department of Urology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Ming Chen
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing 210009, China
| | - Jianping Wu
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing 210009, China
| | - Shuqiu Chen
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing 210009, China
| | - Chao Sun
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing 210009, China
| | - Bin Xu
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing 210009, China
| |
Collapse
|
5
|
Lv W, Wang Y. Neural Influences on Tumor Progression Within the Central Nervous System. CNS Neurosci Ther 2024; 30:e70097. [PMID: 39469896 PMCID: PMC11519750 DOI: 10.1111/cns.70097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/21/2024] [Accepted: 10/13/2024] [Indexed: 10/30/2024] Open
Abstract
For decades, researchers have studied how brain tumors, the immune system, and drugs interact. With the advances in cancer neuroscience, which centers on defining and therapeutically targeting nervous system-cancer interactions, both within the local tumor microenvironment (TME) and on a systemic level, the subtle relationship between neurons and tumors in the central nervous system (CNS) has been deeply studied. Neurons, as the executors of brain functional activities, have been shown to significantly influence the emergence and development of brain tumors, including both primary and metastatic tumors. They engage with tumor cells via chemical or electrical synapses, directly regulating tumors or via intricate coupling networks, and also contribute to the TME through paracrine signaling, secreting proteins that exert regulatory effects. For instance, in a study involving a mouse model of glioblastoma, the authors observed a 42% increase in tumor volume when neuronal activity was stimulated, compared to controls (p < 0.01), indicating a direct correlation between neural activity and tumor growth. These thought-provoking results offer promising new strategies for brain tumor therapies, highlighting the potential of neuronal modulation to curb tumor progression. Future strategies may focus on developing drugs to inhibit or neutralize proteins and other bioactive substances secreted by neurons, break synaptic connections and interactions between infiltrating cells and tumor cells, as well as disrupt electrical coupling within glioma cell networks. By harnessing the insights gained from this research, we aspire to usher in a new era of brain tumor therapies that are both more potent and precise.
Collapse
Affiliation(s)
- Wenhao Lv
- Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouZhejiangChina
- School of PharmacyHangzhou Normal UniversityHangzhouZhejiangChina
| | - Yongjie Wang
- School of PharmacyHangzhou Normal UniversityHangzhouZhejiangChina
| |
Collapse
|
6
|
Motevasseli M, Darvishi M, Khoshnevisan A, Zeinalizadeh M, Saffar H, Bayat S, Najafi A, Abbaspour MJ, Mamivand A, Olson SB, Tabrizi M. Distinct tumor-TAM interactions in IDH-stratified glioma microenvironments unveiled by single-cell and spatial transcriptomics. Acta Neuropathol Commun 2024; 12:133. [PMID: 39148129 PMCID: PMC11328419 DOI: 10.1186/s40478-024-01837-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 07/02/2024] [Indexed: 08/17/2024] Open
Abstract
Tumor-associated macrophages (TAMs) residing in the tumor microenvironment (TME) are characterized by their pivotal roles in tumor progression, antitumor immunity, and TME remodeling. However, a thorough comparative characterization of tumor-TAM crosstalk across IDH-defined categories of glioma remains elusive, likely contributing to mixed outcomes in clinical trials. We delineated the phenotypic heterogeneity of TAMs across IDH-stratified gliomas. Notably, two TAM subsets with a mesenchymal phenotype were enriched in IDH-WT glioblastoma (GBM) and correlated with poorer patient survival and reduced response to anti-PD-1 immune checkpoint inhibitor (ICI). We proposed SLAMF9 receptor as a potential therapeutic target. Inference of gene regulatory networks identified PPARG, ELK1, and MXI1 as master transcription factors of mesenchymal BMD-TAMs. Our analyses of reciprocal tumor-TAM interactions revealed distinct crosstalk in IDH-WT tumors, including ANXA1-FPR1/3, FN1-ITGAVB1, VEGFA-NRP1, and TNFSF12-TNFRSF12A with known contribution to immunosuppression, tumor proliferation, invasion and TAM recruitment. Spatially resolved transcriptomics further elucidated the architectural organization of highlighted communications. Furthermore, we demonstrated significant upregulation of ANXA1, FN1, NRP1, and TNFRSF12A genes in IDH-WT tumors using bulk RNA-seq and RT-qPCR. Longitudinal expression analysis of candidate genes revealed no difference between primary and recurrent tumors indicating that the interactive network of malignant states with TAMs does not drastically change upon recurrence. Collectively, our study offers insights into the unique cellular composition and communication of TAMs in glioma TME, revealing novel vulnerabilities for therapeutic interventions in IDH-WT GBM.
Collapse
Affiliation(s)
- Meysam Motevasseli
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Darvishi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Khoshnevisan
- Department of Neurosurgery, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Zeinalizadeh
- Department of Neurosurgery, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Hiva Saffar
- Department of Pathology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Shiva Bayat
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Najafi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Javad Abbaspour
- Department of Neurosurgery, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Mamivand
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Susan B Olson
- Molecular and Medical Genetics, Knight Diagnostics Laboratories, Oregon Health and Science University, Portland, OR, USA
| | - Mina Tabrizi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Molecular and Medical Genetics, Knight Diagnostics Laboratories, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
7
|
Sim N, Carter JM, Deka K, Tan BKT, Sim Y, Tan SM, Li Y. TWEAK/Fn14 signalling driven super-enhancer reprogramming promotes pro-metastatic metabolic rewiring in triple-negative breast cancer. Nat Commun 2024; 15:5638. [PMID: 38965263 PMCID: PMC11224303 DOI: 10.1038/s41467-024-50071-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 06/27/2024] [Indexed: 07/06/2024] Open
Abstract
Triple Negative Breast Cancer (TNBC) is the most aggressive breast cancer subtype suffering from limited targeted treatment options. Following recent reports correlating Fibroblast growth factor-inducible 14 (Fn14) receptor overexpression in Estrogen Receptor (ER)-negative breast cancers with metastatic events, we show that Fn14 is specifically overexpressed in TNBC patients and associated with poor survival. We demonstrate that constitutive Fn14 signalling rewires the transcriptomic and epigenomic landscape of TNBC, leading to enhanced tumour growth and metastasis. We further illustrate that such mechanisms activate TNBC-specific super enhancers (SE) to drive the transcriptional activation of cancer dependency genes via chromatin looping. In particular, we uncover the SE-driven upregulation of Nicotinamide phosphoribosyltransferase (NAMPT), which promotes NAD+ and ATP metabolic reprogramming critical for filopodia formation and metastasis. Collectively, our study details the complex mechanistic link between TWEAK/Fn14 signalling and TNBC metastasis, which reveals several vulnerabilities which could be pursued for the targeted treatment of TNBC patients.
Collapse
Affiliation(s)
- Nicholas Sim
- School of Biological Sciences (SBS), Nanyang Technological University (NTU), 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Jean-Michel Carter
- School of Biological Sciences (SBS), Nanyang Technological University (NTU), 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Kamalakshi Deka
- School of Biological Sciences (SBS), Nanyang Technological University (NTU), 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Benita Kiat Tee Tan
- Division of Surgery and Surgical Oncology, Department of Breast Surgery, National Cancer Centre Singapore, 30 Hospital Blvd, Singapore, 168583, Singapore
- Division of Surgery and Surgical Oncology, Department of Breast Surgery, Singapore General Hospital, 31 Third Hospital Ave, Singapore, 168753, Singapore
- SingHealth Duke-NUS Breast Centre, Singapore, Singapore
| | - Yirong Sim
- Division of Surgery and Surgical Oncology, Department of Breast Surgery, National Cancer Centre Singapore, 30 Hospital Blvd, Singapore, 168583, Singapore
- Division of Surgery and Surgical Oncology, Department of Breast Surgery, Singapore General Hospital, 31 Third Hospital Ave, Singapore, 168753, Singapore
- SingHealth Duke-NUS Breast Centre, Singapore, Singapore
| | - Suet-Mien Tan
- School of Biological Sciences (SBS), Nanyang Technological University (NTU), 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Yinghui Li
- School of Biological Sciences (SBS), Nanyang Technological University (NTU), 60 Nanyang Drive, Singapore, 637551, Singapore.
| |
Collapse
|
8
|
Medeiros M, Guenka S, Bastos D, Oliveira KL, Brassesco MS. Amicis Omnia Sunt Communia: NF-κB Inhibition as an Alternative to Overcome Osteosarcoma Heterogeneity. Pharmaceuticals (Basel) 2024; 17:734. [PMID: 38931401 PMCID: PMC11206879 DOI: 10.3390/ph17060734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
Tumor heterogeneity poses a significant challenge in osteosarcoma (OS) treatment. In this regard, the "omics" era has constantly expanded our understanding of biomarkers and altered signaling pathways (i.e., PI3K/AKT/mTOR, WNT/β-catenin, NOTCH, SHH/GLI, among others) involved in OS pathophysiology. Despite different players and complexities, many commonalities have been described, among which the nuclear factor kappa B (NF-κB) stands out. Its altered activation is pervasive in cancer, with pleiotropic action on many disease-relevant traits. Thus, in the scope of this article, we highlight the evidence of NF-κB dysregulation in OS and its integration with other cancer-related pathways while we summarize the repertoire of compounds that have been described to interfere with its action. In silico strategies were used to demonstrate that NF-κB is closely coordinated with other commonly dysregulated signaling pathways not only by functionally interacting with several of their members but also by actively participating in the regulation of their transcription. While existing inhibitors lack selectivity or act indirectly, the therapeutic potential of targeting NF-κB is indisputable, first for its multifunctionality on most cancer hallmarks, and secondly, because, as a common downstream effector of the many dysregulated pathways influencing OS aggressiveness, it turns complex regulatory networks into a simpler picture underneath molecular heterogeneity.
Collapse
Affiliation(s)
- Mariana Medeiros
- Cell Biology Department, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirão Preto 14040-900, São Paulo, Brazil;
| | - Sophia Guenka
- Biology Department, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirão Preto 14040-900, São Paulo, Brazil; (S.G.); (D.B.)
| | - David Bastos
- Biology Department, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirão Preto 14040-900, São Paulo, Brazil; (S.G.); (D.B.)
| | - Karla Laissa Oliveira
- Regional Blood Center, University of São Paulo, Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirão Preto 14051-140, São Paulo, Brazil;
| | - María Sol Brassesco
- Biology Department, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirão Preto 14040-900, São Paulo, Brazil; (S.G.); (D.B.)
| |
Collapse
|
9
|
Jin X, Dong W, Chang K, Yan Y. Research on the signaling pathways related to the intervention of traditional Chinese medicine in Parkinson's disease:A literature review. JOURNAL OF ETHNOPHARMACOLOGY 2024; 326:117850. [PMID: 38331124 DOI: 10.1016/j.jep.2024.117850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 01/23/2024] [Accepted: 01/30/2024] [Indexed: 02/10/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Parkinson's disease (PD) is the most common progressive neurodegenerative disorder affecting more than 10 million people worldwide and is characterized by the progressive loss of Daergic (DA) neurons in the substantia nigra pars compacta. It has been reported that signaling pathways play a crucial role in the pathogenesis of PD, while the active ingredients of traditional Chinese medicine (TCM) have been found to possess a protective effect against PD. TCM has demonstrated significant potential in mitigating oxidative stress (OS), neuroinflammation, and apoptosis of DA neurons via the regulation of signaling pathways associated with PD. AIM OF THE REVIEW This study discussed and analyzed the signaling pathways involved in the occurrence and development of PD and the mechanism of active ingredients of TCM regulating PD via signaling pathways, with the aim of providing a basis for the development and clinical application of therapeutic strategies for TCM in PD. MATERIALS AND METHODS With "Parkinson's disease", "Idiopathic Parkinson's Disease", "Lewy Body Parkinson's Disease", "Parkinson's Disease, Idiopathic", "Parkinson Disease, Idiopathic", "Parkinson's disorders", "Parkinsonism syndrome", "Traditional Chinese medicine", "Chinese herbal medicine", "active ingredients", "medicinal plants" as the main keywords, PubMed, Web of Science and other online search engines were used for literature retrieval. RESULTS PD exhibits a close association with various signaling pathways, including but not limited to MAPKs, NF-κB, PI3K/Akt, Nrf2/ARE, Wnt/β-catenin, TLR/TRIF, NLRP3, Notch. The therapeutic potential of TCM lies in its ability to regulate these signaling pathways. In addition, the active ingredients of TCM have shown significant effects in improving OS, neuroinflammation, and DA neuron apoptosis in PD. CONCLUSION The active ingredients of TCM have unique advantages in regulating PD-related signaling pathways. It is suggested to combine network pharmacology and bioinformatics to study the specific targets of TCM. This not only provides a new way for the prevention and treatment of PD with the active ingredients of TCM, but also provides a scientific basis for the selection and development of TCM preparations.
Collapse
Affiliation(s)
- Xiaxia Jin
- National Key Laboratory of Quality Assurance and Sustainable Utilization of Authentic Medicinal Materials, Chinese Medicine Resource Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Wendi Dong
- Foshan Clinical Medical College, Guangzhou University of Traditional Chinese Medicine, Foshan 528000, China
| | - Kaile Chang
- Shaanxi University of Traditional Chinese Medicine, Xianyang, 712046, China
| | - Yongmei Yan
- National Key Laboratory of Quality Assurance and Sustainable Utilization of Authentic Medicinal Materials, Chinese Medicine Resource Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China; Department of Encephalopathy, Affiliated Hospital of Shaanxi University of Traditional Chinese Medicine, Xianyang 712000, China.
| |
Collapse
|
10
|
Pflug KM, Lee DW, Tripathi A, Bankaitis VA, Burgess K, Sitcheran R. Cyanine Dye Conjugation Enhances Crizotinib Localization to Intracranial Tumors, Attenuating NF-κB-Inducing Kinase Activity and Glioma Progression. Mol Pharm 2023; 20:6140-6150. [PMID: 37939020 DOI: 10.1021/acs.molpharmaceut.3c00496] [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] [Indexed: 11/10/2023]
Abstract
Glioblastoma (GBM) is a highly aggressive form of brain cancer with a poor prognosis and limited treatment options. The ALK and c-MET inhibitor Crizotinib has demonstrated preclinical therapeutic potential for newly diagnosed GBM, although its efficacy is limited by poor penetration of the blood brain barrier. Here, we identify Crizotinib as a novel inhibitor of nuclear factor-κB (NF-κB)-inducing kinase, which is a key regulator of GBM growth and proliferation. We further show that the conjugation of Crizotinib to a heptamethine cyanine dye, or a near-infrared dye (IR-Crizotinib), attenuated glioma cell proliferation and survival in vitro to a greater extent than unconjugated Crizotinib. Moreover, we observed increased IR-Crizotinib localization to orthotopic mouse xenograft GBM tumors, which resulted in impaired tumor growth in vivo. Overall, IR-Crizotinib exhibited improved intracranial chemotherapeutic delivery and tumor localization with concurrent inhibition of NIK and noncanonical NF-κB signaling, thereby reducing glioma growth in vitro, as well as in vivo, and increasing survival in a preclinical rodent model.
Collapse
Affiliation(s)
- Kathryn M Pflug
- Department of Cellular Biology and Genetics, Texas A&M University Health Science Center , College Station, Texas 77807, United States
| | - Dong W Lee
- Department of Cellular Biology and Genetics, Texas A&M University Health Science Center , College Station, Texas 77807, United States
| | - Ashutosh Tripathi
- Department of Cellular Biology and Genetics, Texas A&M University Health Science Center , College Station, Texas 77807, United States
| | - Vytas A Bankaitis
- Department of Cellular Biology and Genetics, Texas A&M University Health Science Center , College Station, Texas 77807, United States
| | - Kevin Burgess
- Department of Chemistry, Texas A&M University, Box 30012, College Station, Texas 77842, United States
| | - Raquel Sitcheran
- Department of Cellular Biology and Genetics, Texas A&M University Health Science Center , College Station, Texas 77807, United States
| |
Collapse
|
11
|
Chen K, Shi Y, Luo W, Zhang T, Bao K, Huang C. SMIM20: a new biological signal associated with the prognosis of glioblastoma. Transl Cancer Res 2023; 12:2754-2763. [PMID: 37969370 PMCID: PMC10643959 DOI: 10.21037/tcr-23-796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/13/2023] [Indexed: 11/17/2023]
Abstract
Background Glioblastoma multiforme (GBM) is the most prevalent fatal central nervous system tumor. Notably, the survival rates after surgical intervention and active radiotherapy are not optimistic. Therefore, identifying new GBM-related biomarkers is a top priority in current research. Methods Transcriptome and clinical information of patients with GBM were obtained from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) databases. According to the SMIM20 expression levels, the samples were divided into high- and low-expression groups and used for differential expression gene (DEG) analysis. Functional enrichment analyses, including Gene Ontology (GO), gene set enrichment analysis, and immune cell infiltration, were performed on SMIM20-related DEGs. Subsequently, univariate and multivariate Cox regression analyses were performed to screen the risk factors associated with the poor prognosis of SMIM20, and the clinical significance of SMIM20 in GBM was explored by constructing a prognostic nomogram. Results In total, 156 DEGs were screened, of which 131 were upregulated and 25 were downregulated. Kaplan-Meier analysis revealed that the total survival time of the SMIM20 high expression group was significantly lower than that of the SMIM20 low-expression group. Finally, the nomogram map had good predictive value for evaluating GBM prognosis of patients. Conclusions High expression of SMIM20 is associated with poor outcomes in GBM. The DEGs and pathways identified in this study reveal potential molecular mechanisms underlying the occurrence and progression of GBM. Our study identifies potential new biomarkers and therapeutic targets for the treatment of GBM.
Collapse
Affiliation(s)
- Kai Chen
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yu Shi
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wenzhang Luo
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Tianyu Zhang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Kunyang Bao
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Changren Huang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Neurosurgical Clinical Research Center of Sichuan Province, Luzhou, China
- Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Laboratory of Neurological Diseases and Brain Functions, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
12
|
Wu X, Ma Y, Wang L, Qin X. A Route for Investigating Psoriasis: From the Perspective of the Pathological Mechanisms and Therapeutic Strategies of Cancer. Int J Mol Sci 2023; 24:14390. [PMID: 37762693 PMCID: PMC10532365 DOI: 10.3390/ijms241814390] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/20/2023] [Accepted: 09/20/2023] [Indexed: 09/29/2023] Open
Abstract
Psoriasis is an incurable skin disease that develops in about two-thirds of patients before the age of 40 and requires lifelong treatment; its pathological mechanisms have not been fully elucidated. The core pathological process of psoriasis is epidermal thickening caused by the excessive proliferation of epidermal keratinocytes, which is similar to the key feature of cancer; the malignant proliferation of cancer cells causes tumor enlargement, suggesting that there is a certain degree of commonality between psoriasis and cancer. This article reviews the pathological mechanisms that are common to psoriasis and cancer, including the interaction between cell proliferation and an abnormal immune microenvironment, metabolic reprogramming, and epigenetic reprogramming. In addition, there are common therapeutic agents and drug targets between psoriasis and cancer. Thus, psoriasis and cancer share a common pathological mechanisms-drug targets-therapeutic agents framework. On this basis, it is proposed that investigating psoriasis from a cancer perspective is beneficial to enriching the research strategies related to psoriasis.
Collapse
Affiliation(s)
- Xingkang Wu
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan 030006, China; (Y.M.); (L.W.)
| | | | | | - Xuemei Qin
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan 030006, China; (Y.M.); (L.W.)
| |
Collapse
|
13
|
Duan W, Guo S, Huang HP, Tian Y, Li Z, Bi Y, Yi L, Cao M, Guo M, Li Y, Liu Y, Li C. High expression of NF-κB inducing kinase in the bulge region of hair follicle induces tumor. Immunobiology 2023; 228:152705. [PMID: 37459681 DOI: 10.1016/j.imbio.2023.152705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/19/2023] [Accepted: 07/04/2023] [Indexed: 08/29/2023]
Abstract
The bulge region, a reservoir of multipotent stem cells, is possibly responsible for tumorigenesis. NF-κB-inducing kinase (NIK) is a kinase involved in the activation of the noncanonical NF-κB pathway and exhibits positive staining in tumor cells. However, whether high expression of NIK can result in tumorigenesis has not been reported in published papers. By establishing Nik-coe (Nik-stopF/F crossed with Chat-cre) and Nik-soe (Nik-stopF/F crossed with Sox9-cre) mice, we found that overexpression of Nik in the bulge region of hair follicles induced hair follicle loss and tumorigenesis. Furthermore, RNA sequencing, proteomic and phosphopeptide analyses revealed that multiple cancer pathways are involved in tumor formation. Taken together, these findings indicate that constitutive activation of Nik in the bulge region induces tumorigenesis.
Collapse
Affiliation(s)
- Weisong Duan
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Shengmin Guo
- Hebei Senlang Biotechnology Co., Ltd., No. 136 Yellow River Avenue, Shijiazhuang High-Tech Development Zone, Hebei 050000, People's Republic of China
| | - Huai-Peng Huang
- Shijiazhuang Pingan Hospital, Shijiazhuang, Hebei 050021, People's Republic of China
| | - Yunyun Tian
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Zhongyao Li
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Yue Bi
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Le Yi
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Mengjie Cao
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Moran Guo
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Yuanyuan Li
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Yakun Liu
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Chunyan Li
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei 050000, People's Republic of China.
| |
Collapse
|
14
|
Pflug KM, Lee DW, McFadden K, Herrera L, Sitcheran R. Transcriptional induction of NF-κB-inducing kinase by E2F4/5 facilitates collective invasion of GBM cells. Sci Rep 2023; 13:13093. [PMID: 37567906 PMCID: PMC10421885 DOI: 10.1038/s41598-023-38996-9] [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: 02/23/2023] [Accepted: 07/18/2023] [Indexed: 08/13/2023] Open
Abstract
The prognosis of high-grade gliomas, such as glioblastoma multiforme (GBM), is extremely poor due to the highly invasive nature of these aggressive cancers. Previous work has demonstrated that TNF-weak like factor (TWEAK) induction of the noncanonical NF-κB pathway promotes the invasiveness of GBM cells in an NF-κB-inducing kinase (NIK)-dependent manner. While NIK activity is predominantly regulated at the posttranslational level, we show here that NIK (MAP3K14) is upregulated at the transcriptional level in invading cell populations, with the highest NIK expression observed in the most invasive cells. GBM cells with high induction of NIK gene expression demonstrate characteristics of collective invasion, facilitating invasion of neighboring cells. Furthermore, we demonstrate that the E2F transcription factors E2F4 and E2F5 directly regulate NIK transcription and are required to promote GBM cell invasion in response to TWEAK. Overall, our findings demonstrate that transcriptional induction of NIK facilitates collective cell migration and invasion, thereby promoting GBM pathogenesis.
Collapse
Affiliation(s)
- Kathryn M Pflug
- Department of Cell Biology and Genetics, School of Medicine, Texas A&M University Health Science Center, Bryan, TX, 77807, USA.
| | - Dong W Lee
- Department of Cell Biology and Genetics, School of Medicine, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
| | - Kassandra McFadden
- Department of Cell Biology and Genetics, School of Medicine, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
- 59Th Medical Wing, San Antonio Air Force Base, San Antonio, TX, 78236, USA
| | - Linda Herrera
- Department of Cell Biology and Genetics, School of Medicine, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
- Massachusetts General Hospital, 55 Fruit St., Boston, MA, 2114, USA
| | - Raquel Sitcheran
- Department of Cell Biology and Genetics, School of Medicine, Texas A&M University Health Science Center, Bryan, TX, 77807, USA.
| |
Collapse
|
15
|
Li K, Ma Y, Xia X, Huang H, Li J, Wang X, Gao Y, Zhang S, Fu T, Tong Y. Possible correlated signaling pathways with chronic urate nephropathy: A review. Medicine (Baltimore) 2023; 102:e34540. [PMID: 37565908 PMCID: PMC10419604 DOI: 10.1097/md.0000000000034540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 07/11/2023] [Indexed: 08/12/2023] Open
Abstract
Hyperuricemia nephropathy, also known as gouty nephropathy, refers to renal damage induced by hyperuricemia caused by excessive production of serum uric acid or low excretion of uric acid. the persistence of symptoms will lead to changes in renal tubular phenotype and accelerate the progress of renal fibrosis. The existence and progressive aggravation of symptoms will bring a heavy burden to patients, their families and society, affect their quality of life and reduce their well-being. With the increase of reports on hyperuricemia nephropathy, the importance of related signal pathways in the pathogenesis of hyperuricemia nephropathy is becoming more and more obvious, but most studies are limited to the upper and lower mediating relationship between 1 or 2 signal pathways. The research on the comprehensiveness of signal pathways and the breadth of crosstalk between signal pathways is limited. By synthesizing the research results of signal pathways related to hyperuricemia nephropathy in recent years, this paper will explore the specific mechanism of hyperuricemia nephropathy, and provide new ideas and methods for the treatment of hyperuricemia nephropathy based on a variety of signal pathway crosstalk and personal prospects.
Collapse
Affiliation(s)
- Kaiqing Li
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Yanchun Ma
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Xue Xia
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Huili Huang
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Jianing Li
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Xiaoxin Wang
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Yang Gao
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Shuxiang Zhang
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Tong Fu
- Brandeis University, Waltham, MA
| | - Ying Tong
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
16
|
Sim N, Li Y. NF-κB/p52 augments ETS1 binding genome-wide to promote glioma progression. Commun Biol 2023; 6:445. [PMID: 37087499 PMCID: PMC10122670 DOI: 10.1038/s42003-023-04821-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 04/06/2023] [Indexed: 04/24/2023] Open
Abstract
Gliomas are highly invasive and chemoresistant cancers, making them challenging to treat. Chronic inflammation is a key driver of glioma progression as it promotes aberrant activation of inflammatory pathways such as NF-κB signalling, which drives cancer cell invasion and angiogenesis. NF-κB factors typically dimerise with its own family members, but emerging evidence of their promiscuous interactions with other oncogenic factors has been reported to promote transcription of new target genes and function. Here, we show that non-canonical NF-κB activation directly regulates p52 at the ETS1 promoter, activating its expression. This impacts the genomic and transcriptional landscape of ETS1 in a glioma-specific manner. We further show that enhanced non-canonical NF-κB signalling promotes the co-localisation of p52 and ETS1, resulting in transcriptional activation of non-κB and/or non-ETS glioma-promoting genes. We conclude that p52-induced ETS1 overexpression in glioma cells remodels the genome-wide regulatory network of p52 and ETS1 to transcriptionally drive cancer progression.
Collapse
Affiliation(s)
- Nicholas Sim
- School of Biological Sciences (SBS), Nanyang Technological University (NTU), 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Yinghui Li
- School of Biological Sciences (SBS), Nanyang Technological University (NTU), 60 Nanyang Drive, Singapore, 637551, Singapore.
- Institute of Molecular and Cell Biology (IMCB), A*STAR, Singapore, 138673, Singapore.
| |
Collapse
|
17
|
Pflug K, Lee D, McFadden K, Herrera L, Sitcheran R. Transcriptional Induction of NF-kB-Inducing Kinase by E2F4/5 Facilitates Collective Invasion of Glioma Cells. RESEARCH SQUARE 2023:rs.3.rs-2622363. [PMID: 36945490 PMCID: PMC10029079 DOI: 10.21203/rs.3.rs-2622363/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
The prognosis of high-grade gliomas, such as glioblastoma multiforme (GBM), is extremely poor due to the highly invasive nature of these aggressive cancers. Previous work has demonstrated that TNF-weak like factor (TWEAK) induction of the noncanonical NF-κB pathway increases the invasiveness of glioma cells in an NF-κB-inducing kinase (NIK)-dependent manner. While NIK activity is predominantly regulated at the posttranslational level, we show here that NIK ( MAP3K14 ) is upregulated at the transcriptional level in invading cell populations, with the highest expression observed in the most invasive cells. Glioma cells with high induction of NIK gene expression demonstrate characteristics of collective invasion, facilitating invasion of neighboring cells. Furthermore, we demonstrate that the E2F transcription factors E2F4 and E2F5 directly regulate NIK transcription and are required to promote glioma cell invasion in response to TWEAK. Overall, our findings demonstrate that transcriptional induction of NIK facilitates collective cell migration and invasion, thereby promoting glioma pathogenesis.
Collapse
|
18
|
Bogdan M, Meca AD, Turcu-Stiolica A, Oancea CN, Kostici R, Surlin MV, Florescu C. Insights into the Relationship between Pentraxin-3 and Cancer. Int J Mol Sci 2022; 23:15302. [PMID: 36499628 PMCID: PMC9739619 DOI: 10.3390/ijms232315302] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 11/21/2022] [Accepted: 11/29/2022] [Indexed: 12/11/2022] Open
Abstract
Although cancer can be cured if detected early and treated effectively, it is still a leading cause of death worldwide. Tumor development can be limited by an appropiate immune response, but it can be promoted by chronic extensive inflammation through metabolic dysregulation and angiogenesis. In the past decade, numerous efforts have been made in order to identify novel candidates with predictive values in cancer diagnostics. In line with this, researchers have investigated the involvement of pentraxin-3 (PTX-3) in cellular proliferation and immune escape in various types of cancers, although it has not been clearly elucidated. PTX-3 is a member of the long pentraxin subfamily which plays an important role in regulating inflammation, innate immunity response, angiogenesis, and tissue remodeling. Increased synthesis of inflammatory biomarkers and activation of different cellular mechanisms can induce PTX-3 expression in various types of cells (neutrophils, monocytes, lymphocytes, myeloid dendritic cells, fibroblasts, and epithelial cells). PTX-3 has both pro- and anti-tumor functions, thus dual functions in oncogenesis. This review elucidates the potential usefulness of PTX-3 as a serum biomarker in cancer. While future investigations are needed, PTX-3 is emerging as a promising tool for cancer's diagnosis and prognosis, and also treatment monitoring.
Collapse
Affiliation(s)
- Maria Bogdan
- Department of Pharmacology, Faculty of Pharmacy, University of Medicine and Pharmacy, 200349 Craiova, Romania
| | - Andreea-Daniela Meca
- Department of Pharmacology, Faculty of Pharmacy, University of Medicine and Pharmacy, 200349 Craiova, Romania
| | - Adina Turcu-Stiolica
- Department of Pharmacoeconomics, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Carmen Nicoleta Oancea
- Department of Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Roxana Kostici
- Department of Toxicology, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Marin Valeriu Surlin
- Department of General Surgery, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Cristina Florescu
- Department of Cardiology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| |
Collapse
|
19
|
Zhang H, Wang Y, Zhao Y, Liu T, Wang Z, Zhang N, Dai Z, Wu W, Cao H, Feng S, Zhang L, Cheng Q, Liu Z. PTX3 mediates the infiltration, migration, and inflammation-resolving-polarization of macrophages in glioblastoma. CNS Neurosci Ther 2022; 28:1748-1766. [PMID: 35855654 PMCID: PMC9532932 DOI: 10.1111/cns.13913] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 06/19/2022] [Accepted: 06/20/2022] [Indexed: 11/30/2022] Open
Abstract
INTRODUCTION Pentraxin 3 (PTX3) is an essential regulator of the immune system. However, the immune-modulatory role of PTX3 in the tumor microenvironment of glioma has not been elucidated. METHODS The RNA seq samples were obtained from The Cancer Genome Atlas (TCGA) and the China Glioma Genome Atlas (CGGA) datasets. The single-cell sequencing data of glioblastoma (GBM) samples were obtained from the Single Cell Portal platform (http://singlecell.broadinstitute.org). Immunohistochemistry was used to assess PTX3 expression, HAVCR2, PD-1, PD-L1, and CD276 in glioma sections from the Xiangya cohort (n = 60). Multiplex immunofluorescence staining of PTX3, CD68, and CD163 was performed in several solid cancer types, including GBM. HMC3 was cocultured with U251 and U87, and transwell assay and flow cytometry assay were performed to explore the migration and polarization activity of HMC3. RESULTS PTX3 expression is significantly increased in GBM. PTX3 expression predicts worse survival in the Xiangya cohort. PTX3 is closely related to the expression of PD-1, PD-L1, CD276, and HAVCR2 in the tumor microenvironment. Additionally, PTX3 is involved in tumorigenic and immunogenic processes, especially the activity of macrophages based on various signaling pathways in cellular communications and critical transcription factors. Specifically, PTX3 actively mediates macrophages' infiltration, migration, and inflammation-resolving-polarization. PTX3 could also predict immunotherapy response. CONCLUSION PTX3 is critically involved in macrophage infiltration, migration, and inflammation-resolving-polarization and modulates an immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
| | - Yifan Wang
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
- Xiangya School of MedicineCentral South UniversityChangshaChina
| | - Yihan Zhao
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
- Xiangya School of MedicineCentral South UniversityChangshaChina
| | - Tao Liu
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
- Xiangya School of MedicineCentral South UniversityChangshaChina
| | - Zeyu Wang
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
| | - Nan Zhang
- College of Bioinformatics Science and TechnologyHarbin Medical UniversityHarbinChina
| | - Ziyu Dai
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
| | - Wantao Wu
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
- Department of Oncology, Xiangya HospitalCentral South UniversityChangshaChina
| | - Hui Cao
- Department of Psychiatry, The Second People's Hospital of Hunan ProvinceThe Hospital of Hunan University of Chinese MedicineChangshaChina
| | - Songshan Feng
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
| | - Liyang Zhang
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
| | - Quan Cheng
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
| |
Collapse
|
20
|
Ban D, Yu P, Xiang Z, Liu Y. TNF-like weak inducer of apoptosis / nuclear factor κB axis feedback loop promotes spinal cord injury by inducing astrocyte activation. Bioengineered 2022; 13:11503-11516. [PMID: 35506163 PMCID: PMC9275888 DOI: 10.1080/21655979.2022.2068737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Non-canonical signaling pathways have been proved to act as potent sites of astrocytes osmotic expanding or proliferation, which promotes the regeneration of axons in areas with non-neural spinal cord injury (SCI). However, the relevant signal pathway that induces autophagic cell death in astrocytes and its function relative to the TNF-like weak inducer of apoptosis/nuclear factor κB (TWEAK/NF-κB) axis remains elusive. The SCI model was established by vertically striking the spinal cord according to Allen’s model. Astrocytes and neuronal cells were prepared from spinal cells extracted from spinal cord tissues of SCI or normal C57BL/6 newborn mice. After co-culturing astrocytes and neurons, cell viability and autophagy were determined by CCK-8, transmission electron microscopy (TEM), and western blot. The expression of TWEAK, NF-κB and inflammatory cytokines was confirmed by qRT-PCR, western blot, Immunofluorescence and ELISA assay. Chromatin immunoprecipitation (CHIP) was used to evaluate the interaction between TWEAK and NF-κB. Our results demonstrated that knockdown of TWEAK and NF-κB inhibited secretion of high levels of TNF-α/IL-1β, partially counteracted by adding Rap. TWEAK/NF-κB was the positive correlation feedback loop regulating the proliferation and autophagy of astrocytes involved in SCI. Moreover, restraining the excess growth of astrocytes was beneficial to the growth of neurons. Collectively, our findings illustrated that the TWEAK/NF-κB pathway might act as a positive modulator of SCI by inducing astrocyte activation, shedding new insights for SCI treatment.
Collapse
Affiliation(s)
- Dexiang Ban
- Department of Orthopaedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Peng Yu
- Department of Orthopaedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhenyang Xiang
- Department of Orthopaedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yang Liu
- Department of Orthopaedics, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
21
|
Xiang J, Alafate W, Wu W, Wang Y, Li X, Xie W, Bai X, Li R, Wang M, Wang J. NEK2 enhances malignancies of glioblastoma via NIK/NF-κB pathway. Cell Death Dis 2022; 13:58. [PMID: 35031599 PMCID: PMC8760305 DOI: 10.1038/s41419-022-04512-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 12/16/2021] [Accepted: 01/06/2022] [Indexed: 02/08/2023]
Abstract
Glioblastoma (GBM) is one of the most lethal primary brain tumor with a poor median survival less than 15 months. Despite the development of the clinical strategies over the decades, the outcomes for GBM patients remain dismal due to the strong proliferation and invasion ability and the acquired resistance to radiotherapy and chemotherapy. Therefore, developing new biomarkers and therapeutic strategies targeting GBM is in urgent need. In this study, gene expression datasets and relevant clinical information were extracted from public cancers/glioma datasets, including TCGA, GRAVENDEEL, REMBRANDT, and GILL datasets. Differentially expressed genes were analyzed and NEK2 was picked as a candidate gene for subsequent validation. Human tissue samples and corresponding data were collected from our center and detected by immunohistochemistry analysis. Molecular biological assays and in vivo xenograft transplantation were performed to confirm the bioinformatic findings. High-throughput RNA sequencing, followed by KEGG analysis, GSEA analysis and GO analysis were conducted to identify potential signaling pathways related to NEK2 expression. Subsequent mechanism assays were used to verify the relationship between NEK2 and NF-κB signaling. Overall, we identified that NEK2 is significantly upregulated in GBM and the higher expression of NEK2 exhibited a poorer prognosis. Functionally, NEK2 knockdown attenuated cell proliferation, migration, invasion, and tumorigenesis of GBM while NEK2 overexpression promoted the GBM progression. Furthermore, High-throughput RNA sequencing and bioinformatics analysis indicated that NEK2 was positively related to the NF-κB signaling pathway in GBM. Mechanically, NEK2 activated the noncanonical NF-κB signaling pathway by phosphorylating NIK and increasing the activity and stability of NIK. In conclusion, NEK2 promoted the progression of GBM through activation of noncanonical NF-κB signaling, indicating that NEK2- NF-κB axis could be a potential drug target for GBM.
Collapse
Affiliation(s)
- Jianyang Xiang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Wahafu Alafate
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Wei Wu
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yichang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Xiaodong Li
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Wanfu Xie
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Xiaobin Bai
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Ruichun Li
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Maode Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
- Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| | - Jia Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
- Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
22
|
Chen S, Fang Y, Sun L, He R, He B, Zhang S. Long Non-Coding RNA: A Potential Strategy for the Diagnosis and Treatment of Colorectal Cancer. Front Oncol 2021; 11:762752. [PMID: 34778084 PMCID: PMC8578871 DOI: 10.3389/fonc.2021.762752] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 10/06/2021] [Indexed: 12/13/2022] Open
Abstract
Colorectal cancer (CRC), being one of the most commonly diagnosed cancers worldwide, endangers human health. Because the pathological mechanism of CRC is not fully understood, there are many challenges in the prevention, diagnosis, and treatment of this disease. Long non-coding RNAs (lncRNAs) have recently drawn great attention for their potential roles in the different stages of CRC formation, invasion, and progression, including regulation of molecular signaling pathways, apoptosis, autophagy, angiogenesis, tumor metabolism, immunological responses, cell cycle, and epithelial-mesenchymal transition (EMT). This review aims to discuss the potential mechanisms of several oncogenic lncRNAs, as well as several suppressor lncRNAs, in CRC occurrence and development to aid in the discovery of new methods for CRC diagnosis, treatment, and prognosis assessment.
Collapse
Affiliation(s)
- Shanshan Chen
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yi Fang
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China.,The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lingyu Sun
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China.,The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ruonan He
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Beihui He
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuo Zhang
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
23
|
Murphy CE, Walker AK, Weickert CS. Neuroinflammation in schizophrenia: the role of nuclear factor kappa B. Transl Psychiatry 2021; 11:528. [PMID: 34650030 PMCID: PMC8516884 DOI: 10.1038/s41398-021-01607-0] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/22/2021] [Accepted: 09/03/2021] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation, particularly in the dorsolateral prefrontal cortex, is well-established in a subset of people with schizophrenia, with significant increases in inflammatory markers including several cytokines. Yet the cause(s) of cortical inflammation in schizophrenia remains unknown. Clues as to potential microenvironmental triggers and/or intracellular deficits in immunoregulation may be gleaned from looking further upstream of effector immune molecules to transcription factors that control inflammatory gene expression. Here, we focus on the 'master immune regulator' nuclear factor kappa B (NF-κB) and review evidence in support of NF-κB dysregulation causing or contributing to neuroinflammation in patients. We discuss the utility of 'immune biotyping' as a tool to analyse immune-related transcripts and proteins in patient tissue, and the insights into cortical NF-κB in schizophrenia revealed by immune biotyping compared to studies treating patients as a single, homogenous group. Though the ubiquitous nature of NF-κB presents several hurdles for drug development, targeting this key immunoregulator with novel or repurposed therapeutics in schizophrenia is a relatively underexplored area that could aid in reducing symptoms of patients with active neuroinflammation.
Collapse
Affiliation(s)
- Caitlin E. Murphy
- grid.250407.40000 0000 8900 8842Neuroscience Research Australia, Randwick, NSW 2031 Australia
| | - Adam K. Walker
- grid.250407.40000 0000 8900 8842Neuroscience Research Australia, Randwick, NSW 2031 Australia ,grid.1005.40000 0004 4902 0432School of Psychiatry, University of New South Wales, Randwick, NSW 2031 Australia ,grid.1002.30000 0004 1936 7857Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052 Australia
| | - Cynthia Shannon Weickert
- Neuroscience Research Australia, Randwick, NSW, 2031, Australia. .,School of Psychiatry, University of New South Wales, Randwick, NSW, 2031, Australia. .,Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, NY, 13210, USA.
| |
Collapse
|
24
|
Scuderi SA, Lanza M, Casili G, Esposito F, Colarossi C, Giuffrida D, Irene P, Cuzzocrea S, Esposito E, Campolo M. TBK1 Inhibitor Exerts Antiproliferative Effect on Glioblastoma Multiforme Cells. Oncol Res 2021; 28:779-790. [PMID: 33741083 PMCID: PMC8420908 DOI: 10.3727/096504021x16161478258040] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Glioma are common malignant brain tumors, among which glioblastoma multiforme (GBM) has the worst prognosis. Different studies of GBM revealed that targeting nuclear factor B (NF-B) induced an attenuation tumor proliferation and prolonged cell survival. TBK1 {TANK [TRAF (TNF (tumor-necrosis-factor) receptor-associated factor)-associated NF-B activator]-binding kinase 1} is a serine/threonine protein kinase, and it is a member of the IB kinase (IKK) family involved in NF-B pathway activation. The aim of this study was to investigate the potential effect of BX795, an inhibitor of TBK1, in an in vitro and ex vivo model of GBM. GBM cell lines (U87 and U138) and primary GBM cells were treated with different concentrations of BX795 at different time points (24, 48, and 72h) to evaluate cell viability, autophagy, inflammation, and apoptosis. Our results demonstrated that BX795 10 M was able to reduce cell viability, showing antiproliferative effect in U87, U138, and primary GBM cells. Moreover, treatment with BX795 10 M increased the proapoptotic proteins Bax, p53, caspase 3, and caspase 9, whereas the antiapoptotic Bcl-2 expression was reduced. Additionally, our results showed a marked decrease in autophagy following BX795 treatment, reducing Atg 7, Atg 5/12, and AKT expression. The anti-inflammatory effect of BX795 was demonstrated by a significantly reduction in NIK, IKK, and TNF- expression, accompanied by a downregulation of angiogenesis. Furthermore, in primary GBM cell, BX795 10 M was able to reduce TBK1 pathway activation and SOX3 expression. In conclusion, these findings showed that TBK1 is involved in GBM proliferation, demonstrating that the inhibitor BX795, thanks to its abilities, could improve therapeutic strategies for GBM treatment.
Collapse
Affiliation(s)
- Sarah A. Scuderi
- *Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Marika Lanza
- *Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Giovanna Casili
- *Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | | | | | | | - Paterniti Irene
- *Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Salvatore Cuzzocrea
- *Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Emanuela Esposito
- *Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Michela Campolo
- *Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| |
Collapse
|
25
|
Connolly NP, Galisteo R, Xu S, Bar EE, Peng S, Tran NL, Ames HM, Kim AJ, Woodworth GF, Winkles JA. Elevated fibroblast growth factor-inducible 14 expression transforms proneural-like gliomas into more aggressive and lethal brain cancer. Glia 2021; 69:2199-2214. [PMID: 33991013 PMCID: PMC8596752 DOI: 10.1002/glia.24018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 03/28/2021] [Accepted: 04/28/2021] [Indexed: 12/20/2022]
Abstract
High-grade gliomas (HGGs) are aggressive, treatment-resistant, and often fatal human brain cancers. The TNF-like weak inducer of apoptosis (TWEAK)/fibroblast growth factor-inducible 14 (Fn14) signaling axis is involved in tissue repair after injury and constitutive signaling has been implicated in the pathogenesis of numerous solid cancers. The Fn14 gene is expressed at low levels in the normal, uninjured brain but is highly expressed in primary isocitrate dehydrogenase wild-type and recurrent HGGs. Fn14 signaling is implicated in numerous aspects of glioma biology including brain invasion and chemotherapy resistance, but whether Fn14 overexpression can directly promote tumor malignancy has not been reported. Here, we used the replication-competent avian sarcoma-leukosis virus/tumor virus A system to examine the impact of Fn14 expression on glioma development and pathobiology. We found that the sole addition of Fn14 to an established oncogenic cocktail previously shown to generate proneural-like gliomas led to the development of highly invasive and lethal brain cancer with striking biological features including extensive pseudopalisading necrosis, constitutive canonical and noncanonical NF-κB pathway signaling, and high plasminogen activator inhibitor-1 (PAI-1) expression. Analyses of HGG patient datasets revealed that high human PAI-1 gene (SERPINE1) expression correlates with shorter patient survival, and that the SERPINE1 and Fn14 (TNFRSF12A) genes are frequently co-expressed in bulk tumor tissues, in tumor subregions, and in malignant cells residing in the tumor microenvironment. These findings provide new insights into the potential importance of Fn14 in human HGG pathobiology and designate both the NF-κB signaling node and PAI-1 as potential targets for therapeutic intervention. MAIN POINTS: This work demonstrates that elevated levels of the TWEAK receptor Fn14 in tumor-initiating, neural progenitor cells leads to the transformation of proneural-like gliomas into more aggressive and lethal tumors that exhibit constitutive NF-κB pathway activation and plasminogen activator inhibitor-1 overexpression.
Collapse
Affiliation(s)
- Nina P. Connolly
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Rebeca Galisteo
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland
| | - Su Xu
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland
- Center for Advanced Imaging Research, University of Maryland School of Medicine, Baltimore, Maryland
| | - Eli E. Bar
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Sen Peng
- Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, Arizona
| | - Nhan L. Tran
- Departments of Cancer Biology and Neurosurgery, Mayo Clinic Arizona, Scottsdale, Arizona
| | - Heather M. Ames
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Anthony J. Kim
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Graeme F. Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jeffrey A. Winkles
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
26
|
Medeiros M, Candido MF, Valera ET, Brassesco MS. The multifaceted NF-kB: are there still prospects of its inhibition for clinical intervention in pediatric central nervous system tumors? Cell Mol Life Sci 2021; 78:6161-6200. [PMID: 34333711 PMCID: PMC11072991 DOI: 10.1007/s00018-021-03906-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 12/16/2022]
Abstract
Despite advances in the understanding of the molecular mechanisms underlying the basic biology and pathogenesis of pediatric central nervous system (CNS) malignancies, patients still have an extremely unfavorable prognosis. Over the years, a plethora of natural and synthetic compounds has emerged for the pharmacologic intervention of the NF-kB pathway, one of the most frequently dysregulated signaling cascades in human cancer with key roles in cell growth, survival, and therapy resistance. Here, we provide a review about the state-of-the-art concerning the dysregulation of this hub transcription factor in the most prevalent pediatric CNS tumors: glioma, medulloblastoma, and ependymoma. Moreover, we compile the available literature on the anti-proliferative effects of varied NF-kB inhibitors acting alone or in combination with other therapies in vitro, in vivo, and clinical trials. As the wealth of basic research data continues to accumulate, recognizing NF-kB as a therapeutic target may provide important insights to treat these diseases, hopefully contributing to increase cure rates and lower side effects related to therapy.
Collapse
Affiliation(s)
- Mariana Medeiros
- Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Marina Ferreira Candido
- Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Elvis Terci Valera
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - María Sol Brassesco
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, FFCLRP-USP, University of São Paulo, Av. Bandeirantes, 3900, Bairro Monte Alegre, Ribeirão Preto, São Paulo, CEP 14040-901, Brazil.
| |
Collapse
|
27
|
Cordido A, Nuñez-Gonzalez L, Martinez-Moreno JM, Lamas-Gonzalez O, Rodriguez-Osorio L, Perez-Gomez MV, Martin-Sanchez D, Outeda P, Chiaravalli M, Watnick T, Boletta A, Diaz C, Carracedo A, Sanz AB, Ortiz A, Garcia-Gonzalez MA. TWEAK Signaling Pathway Blockade Slows Cyst Growth and Disease Progression in Autosomal Dominant Polycystic Kidney Disease. J Am Soc Nephrol 2021; 32:1913-1932. [PMID: 34155062 PMCID: PMC8455272 DOI: 10.1681/asn.2020071094] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 03/06/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND In autosomal dominant polycystic kidney disease (ADPKD), cyst development and enlargement lead to ESKD. Macrophage recruitment and interstitial inflammation promote cyst growth. TWEAK is a TNF superfamily (TNFSF) cytokine that regulates inflammatory responses, cell proliferation, and cell death, and its receptor Fn14 (TNFRSF12a) is expressed in macrophage and nephron epithelia. METHODS To evaluate the role of the TWEAK signaling pathway in cystic disease, we evaluated Fn14 expression in human and in an orthologous murine model of ADPKD. We also explored the cystic response to TWEAK signaling pathway activation and inhibition by peritoneal injection. RESULTS Meta-analysis of published animal-model data of cystic disease reveals mRNA upregulation of several components of the TWEAK signaling pathway. We also observed that TWEAK and Fn14 were overexpressed in mouse ADPKD kidney cysts, and TWEAK was significantly high in urine and cystic fluid from patients with ADPKD. TWEAK administration induced cystogenesis and increased cystic growth, worsening the phenotype in a murine ADPKD model. Anti-TWEAK antibodies significantly slowed the progression of ADPKD, preserved renal function, and improved survival. Furthermore, the anti-TWEAK cystogenesis reduction is related to decreased cell proliferation-related MAPK signaling, decreased NF-κB pathway activation, a slight reduction of fibrosis and apoptosis, and an indirect decrease in macrophage recruitment. CONCLUSIONS This study identifies the TWEAK signaling pathway as a new disease mechanism involved in cystogenesis and cystic growth and may lead to a new therapeutic approach in ADPKD.
Collapse
Affiliation(s)
- Adrian Cordido
- Group of Genetics and Developmental Biology of Renal Diseases, Nephrology Laboratory (N°11), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela Clinical Hospital Complex (CHUS), Santiago de Compostela, Spain,Genomic Medicine Group, Santiago de Compostela Clinical Hospital Complex (CHUS), Santiago de Compostela, Spain,RedInRen RETIC, Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Nuñez-Gonzalez
- Group of Genetics and Developmental Biology of Renal Diseases, Nephrology Laboratory (N°11), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela Clinical Hospital Complex (CHUS), Santiago de Compostela, Spain,Genomic Medicine Group, Santiago de Compostela Clinical Hospital Complex (CHUS), Santiago de Compostela, Spain
| | - Julio M. Martinez-Moreno
- Department of Nephrology and Hypertension, Jiménez Díaz Foundation (Health Research Institute and Autonomous University of Madrid), Madrid, Spain
| | - Olaya Lamas-Gonzalez
- Group of Genetics and Developmental Biology of Renal Diseases, Nephrology Laboratory (N°11), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela Clinical Hospital Complex (CHUS), Santiago de Compostela, Spain
| | - Laura Rodriguez-Osorio
- RedInRen RETIC, Instituto de Salud Carlos III, Madrid, Spain,Department of Nephrology and Hypertension, Jiménez Díaz Foundation (Health Research Institute and Autonomous University of Madrid), Madrid, Spain
| | - Maria Vanessa Perez-Gomez
- RedInRen RETIC, Instituto de Salud Carlos III, Madrid, Spain,Department of Nephrology and Hypertension, Jiménez Díaz Foundation (Health Research Institute and Autonomous University of Madrid), Madrid, Spain
| | - Diego Martin-Sanchez
- RedInRen RETIC, Instituto de Salud Carlos III, Madrid, Spain,Department of Nephrology and Hypertension, Jiménez Díaz Foundation (Health Research Institute and Autonomous University of Madrid), Madrid, Spain
| | - Patricia Outeda
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Marco Chiaravalli
- Division of Genetics and Cell Biology, Molecular Basis of Cystic Kidney Disorders Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS)–San Raffaele Scientific Institute, Milan, Italy
| | - Terry Watnick
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | | | - Candido Diaz
- Group of Genetics and Developmental Biology of Renal Diseases, Nephrology Laboratory (N°11), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela Clinical Hospital Complex (CHUS), Santiago de Compostela, Spain,Nephrology Service, Santiago de Compostela Clinical Hospital Complex (CHUS), Santiago de Compostela, Spain
| | - Angel Carracedo
- Genomic Medicine Group, Santiago de Compostela Clinical Hospital Complex (CHUS), Santiago de Compostela, Spain,Galician Public Foundation of Genomic Medicine, Santiago de Compostela Clinical Hospital Complex (CHUS), Santiago de Compostela, Spain,Center in Network of Rare Diseases (CIBERER), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Ana B. Sanz
- RedInRen RETIC, Instituto de Salud Carlos III, Madrid, Spain,Department of Nephrology and Hypertension, Jiménez Díaz Foundation (Health Research Institute and Autonomous University of Madrid), Madrid, Spain
| | - Alberto Ortiz
- RedInRen RETIC, Instituto de Salud Carlos III, Madrid, Spain,Department of Nephrology and Hypertension, Jiménez Díaz Foundation (Health Research Institute and Autonomous University of Madrid), Madrid, Spain
| | - Miguel A. Garcia-Gonzalez
- Group of Genetics and Developmental Biology of Renal Diseases, Nephrology Laboratory (N°11), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela Clinical Hospital Complex (CHUS), Santiago de Compostela, Spain,Genomic Medicine Group, Santiago de Compostela Clinical Hospital Complex (CHUS), Santiago de Compostela, Spain,RedInRen RETIC, Instituto de Salud Carlos III, Madrid, Spain,Galician Public Foundation of Genomic Medicine, Santiago de Compostela Clinical Hospital Complex (CHUS), Santiago de Compostela, Spain
| |
Collapse
|
28
|
Mockenhaupt K, Gonsiewski A, Kordula T. RelB and Neuroinflammation. Cells 2021; 10:1609. [PMID: 34198987 PMCID: PMC8307460 DOI: 10.3390/cells10071609] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/23/2021] [Accepted: 06/23/2021] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation within the central nervous system involves multiple cell types that coordinate their responses by secreting and responding to a plethora of inflammatory mediators. These factors activate multiple signaling cascades to orchestrate initial inflammatory response and subsequent resolution. Activation of NF-κB pathways in several cell types is critical during neuroinflammation. In contrast to the well-studied role of p65 NF-κB during neuroinflammation, the mechanisms of RelB activation in specific cell types and its roles during neuroinflammatory response are less understood. In this review, we summarize the mechanisms of RelB activation in specific cell types of the CNS and the specialized effects this transcription factor exerts during neuroinflammation.
Collapse
Affiliation(s)
| | | | - Tomasz Kordula
- Department of Biochemistry and Molecular Biology, School of Medicine and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VI 23298, USA; (K.M.); (A.G.)
| |
Collapse
|
29
|
Zhang Y, Yang X, Zhu XL, Wang ZZ, Bai H, Zhang JJ, Hao CY, Duan HB. A Novel Immune-Related Prognostic Biomarker and Target Associated With Malignant Progression of Glioma. Front Oncol 2021; 11:643159. [PMID: 33937046 PMCID: PMC8085360 DOI: 10.3389/fonc.2021.643159] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 03/23/2021] [Indexed: 12/31/2022] Open
Abstract
Background Glioma is one of the most common malignancies in the central nervous system and has limited effective therapeutic options. Therefore, we sought to identify a suitable target for immunotherapy. Materials and Methods We screened prognostic genes for glioma in the CGGA database and GSE43378 dataset using survival analysis, receiver operating characteristic (ROC) curves, independent prognostic analysis, and clinical correlation analysis. The results were intersected with immune genes from the ImmPort database through Venn diagrams to obtain likely target genes. The target genes were validated as prognostically relevant immune genes for glioma using survival, ROC curve, independent prognostic, and clinical correlation analyses in samples from the CGGA database and GSE43378 dataset, respectively. We also constructed a nomogram using statistically significant glioma prognostic factors in the CGGA samples and verified their sensitivity and specificity with ROC curves. The functions, pathways, and co-expression-related genes for the glioma target genes were assessed using PPI networks, enrichment analysis, and correlation analysis. The correlation between target gene expression and immune cell infiltration in glioma and the relationship with the survival of glioma patients were investigated using the TIMER database. Finally, target gene expression in normal brain, low-grade glioma, and high-grade glioma tissues was detected using immunohistochemical staining. Results We identified TNFRSF12A as the target gene. Satisfactory results from survival, ROC curve, independent prognosis, and clinical correlation analyses in the CGGA and GSE43378 samples verified that TNFRSF12A was significantly associated with the prognosis of glioma patients. A nomogram was constructed using glioma prognostic correlates, including TNFRSF12A expression, primary-recurrent-secondary (PRS) type, grade, age, chemotherapy, IDH mutation, and 1p19q co-deletion in CGGA samples with an AUC value of 0.860, which illustrated the accuracy of the prognosis prediction. The results of the TIMER analysis validated the significant correlation of TNFRSF12A with immune cell infiltration and glioma survival. The immunohistochemical staining results verified the progressive up-regulation of TNFRSF12A expression in normal brain, low-grade glioma, and high-grade glioma tissues. Conclusion We concluded that TNFRSF12A was a viable prognostic biomarker and a potential immunotherapeutic target for glioma.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Xin Yang
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiao-Lin Zhu
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Zhuang-Zhuang Wang
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Hao Bai
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Jun-Jie Zhang
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Chun-Yan Hao
- Department of Geriatrics, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Hu-Bin Duan
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, China.,Department of Neurosurgery, Lvliang People's Hospital, Lvliang, China
| |
Collapse
|
30
|
Kamradt ML, Jung JU, Pflug KM, Lee DW, Fanniel V, Sitcheran R. NIK promotes metabolic adaptation of glioblastoma cells to bioenergetic stress. Cell Death Dis 2021; 12:271. [PMID: 33723235 PMCID: PMC7960998 DOI: 10.1038/s41419-020-03383-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 12/22/2020] [Accepted: 12/28/2020] [Indexed: 01/31/2023]
Abstract
Cancers, including glioblastoma multiforme (GBM), undergo coordinated reprogramming of metabolic pathways that control glycolysis and oxidative phosphorylation (OXPHOS) to promote tumor growth in diverse tumor microenvironments. Adaptation to limited nutrient availability in the microenvironment is associated with remodeling of mitochondrial morphology and bioenergetic capacity. We recently demonstrated that NF-κB-inducing kinase (NIK) regulates mitochondrial morphology to promote GBM cell invasion. Here, we show that NIK is recruited to the outer membrane of dividing mitochondria with the master fission regulator, Dynamin-related protein1 (DRP1). Moreover, glucose deprivation-mediated metabolic shift to OXPHOS increases fission and mitochondrial localization of both NIK and DRP1. NIK deficiency results in decreased mitochondrial respiration, ATP production, and spare respiratory capacity (SRC), a critical measure of mitochondrial fitness. Although IκB kinase α and β (IKKα/β) and NIK are required for OXPHOS in high glucose media, only NIK is required to increase SRC under glucose deprivation. Consistent with an IKK-independent role for NIK in regulating metabolism, we show that NIK phosphorylates DRP1-S616 in vitro and in vivo. Notably, a constitutively active DRP1-S616E mutant rescues oxidative metabolism, invasiveness, and tumorigenic potential in NIK-/- cells without inducing IKK. Thus, we establish that NIK is critical for bioenergetic stress responses to promote GBM cell pathogenesis independently of IKK. Our data suggest that targeting NIK may be used to exploit metabolic vulnerabilities and improve therapeutic strategies for GBM.
Collapse
Affiliation(s)
- Michael L Kamradt
- Department of Molecular & Cellular Medicine, Texas A&M University Health Science Center, College Station, TX, 77845, USA
- Medical Sciences Graduate Program, Texas A&M University Health Science Center, College Station, TX, 77845, USA
| | - Ji-Ung Jung
- Department of Molecular & Cellular Medicine, Texas A&M University Health Science Center, College Station, TX, 77845, USA
- Medical Sciences Graduate Program, Texas A&M University Health Science Center, College Station, TX, 77845, USA
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Kathryn M Pflug
- Department of Molecular & Cellular Medicine, Texas A&M University Health Science Center, College Station, TX, 77845, USA
- Interdisciplinary Graduate Program in Genetics, Texas A&M University, College Station, TX, 77845, USA
| | - Dong W Lee
- Department of Molecular & Cellular Medicine, Texas A&M University Health Science Center, College Station, TX, 77845, USA
| | - Victor Fanniel
- Department of Molecular & Cellular Medicine, Texas A&M University Health Science Center, College Station, TX, 77845, USA
- Interdisciplinary Graduate Program in Genetics, Texas A&M University, College Station, TX, 77845, USA
| | - Raquel Sitcheran
- Department of Molecular & Cellular Medicine, Texas A&M University Health Science Center, College Station, TX, 77845, USA.
- Medical Sciences Graduate Program, Texas A&M University Health Science Center, College Station, TX, 77845, USA.
- Interdisciplinary Graduate Program in Genetics, Texas A&M University, College Station, TX, 77845, USA.
| |
Collapse
|
31
|
Pflug KM, Sitcheran R. Targeting NF-κB-Inducing Kinase (NIK) in Immunity, Inflammation, and Cancer. Int J Mol Sci 2020; 21:E8470. [PMID: 33187137 PMCID: PMC7696043 DOI: 10.3390/ijms21228470] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 11/03/2020] [Accepted: 11/07/2020] [Indexed: 12/23/2022] Open
Abstract
NF-κB-inducing kinase (NIK), the essential upstream kinase, which regulates activation of the noncanonical NF-κB pathway, has important roles in regulating immunity and inflammation. In addition, NIK is vital for maintaining cellular health through its control of fundamental cellular processes, including differentiation, growth, and cell survival. As such aberrant expression or regulation of NIK is associated with several disease states. For example, loss of NIK leads to severe immune defects, while the overexpression of NIK is observed in inflammatory diseases, metabolic disorders, and the development and progression of cancer. This review discusses recent studies investigating the therapeutic potential of NIK inhibitors in various diseases.
Collapse
Affiliation(s)
- Kathryn M. Pflug
- Interdisciplinary Program in Genetics, Texas A&M University, College Station, TX 77843, USA;
- Department of Molecular & Cellular Medicine, Texas A&M University Health Science Center, Bryan, TX 77002, USA
| | - Raquel Sitcheran
- Interdisciplinary Program in Genetics, Texas A&M University, College Station, TX 77843, USA;
- Department of Molecular & Cellular Medicine, Texas A&M University Health Science Center, Bryan, TX 77002, USA
| |
Collapse
|
32
|
Liu J, Zhan Y, Wang J, Wang J, Guo J, Kong D. Long noncoding RNA LINC01578 drives colon cancer metastasis through a positive feedback loop with the NF-κB/YY1 axis. Mol Oncol 2020; 14:3211-3233. [PMID: 33040438 PMCID: PMC7718957 DOI: 10.1002/1878-0261.12819] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 08/25/2020] [Accepted: 10/07/2020] [Indexed: 02/06/2023] Open
Abstract
Metastasis accounts for poor prognosis of cancers and related deaths. Accumulating evidence has shown that long noncoding RNAs (lncRNAs) play critical roles in several types of cancer. However, which lncRNAs contribute to metastasis of colon cancer is still largely unknown. In this study, we found that lncRNA LINC01578 was correlated with metastasis and poor prognosis of colon cancer. LINC01578 was upregulated in colon cancer, associated with metastasis, advanced clinical stages, poor overall survival, disease-specific survival, and disease-free survival. Gain-of-function and loss-of-function assays revealed that LINC01578 enhanced colon cancer cell viability and mobility in vitro and colon cancer liver metastasis in vivo. Mechanistically, nuclear factor kappa B (NF-κB) and Yin Yang 1 (YY1) directly bound to the LINC01578 promoter, enhanced its activity, and activated LINC01578 expression. LINC01578 was shown to be a chromatin-bound lncRNA, which directly bound NFKBIB promoter. Furthermore, LINC01578 interacted with and recruited EZH2 to NFKBIB promoter and further repressed NFKBIB expression, thereby activating NF-κB signaling. Through activation of NF-κB, LINC01578 further upregulated YY1 expression. Through activation of the NF-κB/YY1 axis, LINC01578 in turn enhanced its own promoter activity, suggesting that LINC01578 and NF-κB/YY1 formed a positive feedback loop. Blocking NF-κB signaling abolished the oncogenic roles of LINC01578 in colon cancer. Furthermore, the expression levels of LINC01578, NFKBIB, and YY1 were correlated in clinical tissues. Collectively, this study demonstrated that LINC01578 promoted colon cancer metastasis via forming a positive feedback loop with NF-κB/YY1 and suggested that LINC01578 represents a potential prognostic biomarker and therapeutic target for colon cancer metastasis.
Collapse
Affiliation(s)
- Jia Liu
- Department of Colorectal Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yang Zhan
- Department of Colorectal Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jiefu Wang
- Department of Colorectal Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Junfeng Wang
- Department of Colorectal Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jiansheng Guo
- Department of Colorectal Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Dalu Kong
- Department of Colorectal Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
33
|
Portela M, Mitchell T, Casas-Tintó S. Cell-to-cell communication mediates glioblastoma progression in Drosophila. Biol Open 2020; 9:bio053405. [PMID: 32878880 PMCID: PMC7541342 DOI: 10.1242/bio.053405] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 08/23/2020] [Indexed: 12/16/2022] Open
Abstract
Glioblastoma (GB) is the most aggressive and lethal tumour of the central nervous system (CNS). GB cells grow rapidly and display a network of projections, ultra-long tumour microtubes (TMs), that mediate cell to cell communication. GB-TMs infiltrate throughout the brain, enwrap neurons and facilitate the depletion of the signalling molecule wingless (Wg)/WNT from the neighbouring healthy neurons. GB cells establish a positive feedback loop including Wg signalling upregulation that activates cJun N-terminal kinase (JNK) pathway and matrix metalloproteases (MMPs) production, which in turn promote further TMs infiltration, GB progression and neurodegeneration. Thus, cellular and molecular signals other than primary mutations emerge as central players of GB. Using a Drosophila model of GB, we describe the temporal organisation of the main cellular events that occur in GB, including cell-to-cell interactions, neurodegeneration and TM expansion. We define the progressive activation of JNK pathway signalling in GB mediated by the receptor Grindelwald (Grnd) and activated by the ligand Eiger (Egr)/TNFα produced by surrounding healthy brain tissue. We propose that cellular interactions of GB with the healthy brain tissue precede TM expansion and conclude that non-autonomous signals facilitate GB progression. These results contribute to deciphering the complexity and versatility of these incurable tumours.
Collapse
Affiliation(s)
- Marta Portela
- Molecular, Cellular and Developmental Neurobiology Department, Instituto Cajal-CSIC, Av. del Doctor Arce, 37, 28002 Madrid, Spain
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Sciences, La Trobe University, 3086 Melbourne, Australia
| | - Teresa Mitchell
- Molecular, Cellular and Developmental Neurobiology Department, Instituto Cajal-CSIC, Av. del Doctor Arce, 37, 28002 Madrid, Spain
| | - Sergio Casas-Tintó
- Molecular, Cellular and Developmental Neurobiology Department, Instituto Cajal-CSIC, Av. del Doctor Arce, 37, 28002 Madrid, Spain
| |
Collapse
|
34
|
Wang M, Xie Z, Xu J, Feng Z. TWEAK/Fn14 axis in respiratory diseases. Clin Chim Acta 2020; 509:139-148. [PMID: 32526219 DOI: 10.1016/j.cca.2020.06.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/31/2020] [Accepted: 06/03/2020] [Indexed: 02/08/2023]
Abstract
Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) is a well known multifunctional cytokine extensively distributed in cell types and tissues. Accumulating evidence has shown that TWEAK binding to the receptor factor-inducible 14 (Fn14) participates in diverse pathologic processes including cell proliferation and death, angiogenesis, carcinogenesis and inflammation. Interestingly, alterations of intracellular signaling cascades are correlated to the development of respiratory disease. Recently, a several lines of evidence suggests that TWEAK in lung tissues are closely associated with these signaling pathways. In this review, we explore if TWEAK could provide a novel therapeutic strategy for managing respiratory disease in general and pulmonary arterial hypertension (PAH), obstructive sleep apnea syndrome (OSAS), asthma, idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD) and non-small cell lung cancer (NSCLC), specifically.
Collapse
Affiliation(s)
- Min Wang
- Department of Otorhinolaryngology, University of South China Affiliated Nanhua Hospital, Hengyang 421002, China
| | - Zhijuan Xie
- Department of Nephrology, The First Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Jin Xu
- School of Pharmaceutical Sciences, Changsha Medical University, Changsha 410219, Hunan, China.
| | - Zhuyu Feng
- Department of Critical Care Medicine, University of South China Affiliated Nanhua Hospital, Hengyang 421002, China.
| |
Collapse
|
35
|
Hersh DS, Harder BG, Roos A, Peng S, Heath JE, Legesse T, Kim AJ, Woodworth GF, Tran NL, Winkles JA. The TNF receptor family member Fn14 is highly expressed in recurrent glioblastoma and in GBM patient-derived xenografts with acquired temozolomide resistance. Neuro Oncol 2019; 20:1321-1330. [PMID: 29897522 DOI: 10.1093/neuonc/noy063] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background Glioblastoma (GBM) is a difficult to treat brain cancer that nearly uniformly recurs, and recurrent tumors are largely therapy resistant. Our prior work has demonstrated an important role for the tumor necrosis factor-like weak inducer of apoptosis (TWEAK) receptor fibroblast growth factor-inducible 14 (Fn14) in GBM pathobiology. In this study, we investigated Fn14 expression in recurrent GBM and in the setting of temozolomide (TMZ) resistance. Methods Fn14 mRNA expression levels in nonneoplastic brain, primary (newly diagnosed) GBM, and recurrent GBM (post-chemotherapy and radiation) specimens were obtained from The Cancer Genome Atlas data portal. Immunohistochemistry was performed using nonneoplastic brain, patient-matched primary and recurrent GBM, and gliosarcoma (GSM) specimens to examine Fn14 protein levels. Western blot analysis was used to compare Fn14 expression in parental TMZ-sensitive or matched TMZ-resistant patient-derived xenografts (PDXs) established from primary or recurrent tumor samples. The migratory capacity of control and Fn14-depleted TMZ-resistant GBM cells was assessed using the transwell migration assay. Results We found that Fn14 is more highly expressed in recurrent GBM tumors than their matched primary GBM counterparts. Fn14 expression is also significantly elevated in GSM tumors. GBM PDX cells with acquired TMZ resistance have higher Fn14 levels and greater migratory capacity than their corresponding parental TMZ-sensitive cells, and the migratory difference is due, at least in part, to Fn14 expression in the TMZ-resistant cells. Conclusions This study demonstrates that the Fn14 gene is highly expressed in recurrent GBM, GSM, and TMZ-resistant GBM PDX tumors. These findings suggest that Fn14 may be a valuable therapeutic target or drug delivery portal for treatment of recurrent GBM and GSM patients.
Collapse
Affiliation(s)
- David S Hersh
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - Bryan G Harder
- Departments of Cancer Biology and Neurosurgery, Mayo Clinic Arizona, Scottsdale, Arizona
| | - Alison Roos
- Departments of Cancer Biology and Neurosurgery, Mayo Clinic Arizona, Scottsdale, Arizona
| | - Sen Peng
- Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, Arizona
| | - Jonathan E Heath
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Teklu Legesse
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Anthony J Kim
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland.,University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, Maryland
| | - Graeme F Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland.,University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, Maryland
| | - Nhan L Tran
- Departments of Cancer Biology and Neurosurgery, Mayo Clinic Arizona, Scottsdale, Arizona
| | - Jeffrey A Winkles
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, Maryland.,Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland.,Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
36
|
Ahmadi-Beni R, Najafi A, Savar SM, Mohebbi N, Khoshnevisan A. Role of dimethyl fumarate in the treatment of glioblastoma multiforme: A review article. IRANIAN JOURNAL OF NEUROLOGY 2019; 18:127-133. [PMID: 31749934 PMCID: PMC6858600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 05/16/2019] [Indexed: 10/28/2022]
Abstract
Glioblastoma multiforme (GBM), the most frequent malignant and aggressive primary brain tumor, is characterized by genetically unstable heterogeneous cells, diffused growth pattern, microvascular proliferation, and resistance to chemotherapy. Extensive investigations are being carried out to identify the molecular origin of resistance to chemo- and radio-therapy in GBM and find novel targets for therapy to improve overall survival rate. Dimethyl fumarate (DMF) has been shown to be a safe drug with limited short and long-term side effects, and fumaric acid esters (FAEs), including DMF, present both anti-oxidative and anti-inflammatory activity in different cell types and tissues. DMF has also anti-tumoral and neuroprotective effects and so it could be repurposed in the treatment of this invasive tumor in the future. Here, we have reviewed DMF pharmacokinetics and different mechanisms by which DMF could have therapeutic effects on GBM.
Collapse
Affiliation(s)
- Reza Ahmadi-Beni
- Department of Medical Genetics, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ali Najafi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Student Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Niayesh Mohebbi
- Department of Clinical Pharmacy, School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Khoshnevisan
- Department of Neurosurgery, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
37
|
Waters MR, Gupta AS, Mockenhaupt K, Brown LN, Biswas DD, Kordula T. RelB acts as a molecular switch driving chronic inflammation in glioblastoma multiforme. Oncogenesis 2019; 8:37. [PMID: 31142741 PMCID: PMC6541631 DOI: 10.1038/s41389-019-0146-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 05/03/2019] [Accepted: 05/16/2019] [Indexed: 01/31/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a primary brain tumor characterized by extensive necrosis and immunosuppressive inflammation. The mechanisms by which this inflammation develops and persists in GBM remain elusive. We identified two cytokines interleukin-1β (IL-1) and oncostatin M (OSM) that strongly negatively correlate with patient survival. We found that these cytokines activate RelB/p50 complexes by a canonical NF-κB pathway, which surprisingly drives expression of proinflammatory cytokines in GBM cells, but leads to their inhibition in non-transformed astrocytes. We discovered that one allele of the gene encoding deacetylase Sirtuin 1 (SIRT1), needed for repression of cytokine genes, is deleted in 80% of GBM tumors. Furthermore, RelB specifically interacts with a transcription factor Yin Yang 1 (YY1) in GBM cells and activates GBM-specific gene expression programs. As a result, GBM cells continuously secrete proinflammatory cytokines and factors attracting/activating glioma-associated microglia/macrophages and thus, promote a feedforward inflammatory loop.
Collapse
Affiliation(s)
- Michael R Waters
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth, University School of Medicine and the Massey Cancer Center, Richmond, VI, 23298, USA
| | - Angela S Gupta
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth, University School of Medicine and the Massey Cancer Center, Richmond, VI, 23298, USA
| | - Karli Mockenhaupt
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth, University School of Medicine and the Massey Cancer Center, Richmond, VI, 23298, USA
| | - LaShardai N Brown
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth, University School of Medicine and the Massey Cancer Center, Richmond, VI, 23298, USA
| | - Debolina D Biswas
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth, University School of Medicine and the Massey Cancer Center, Richmond, VI, 23298, USA
| | - Tomasz Kordula
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth, University School of Medicine and the Massey Cancer Center, Richmond, VI, 23298, USA.
| |
Collapse
|
38
|
Dresselhaus EC, Meffert MK. Cellular Specificity of NF-κB Function in the Nervous System. Front Immunol 2019; 10:1043. [PMID: 31143184 PMCID: PMC6520659 DOI: 10.3389/fimmu.2019.01043] [Citation(s) in RCA: 223] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 04/24/2019] [Indexed: 12/17/2022] Open
Abstract
Nuclear Factor Kappa B (NF-κB) is a ubiquitously expressed transcription factor with key functions in a wide array of biological systems. While the role of NF-κB in processes, such as host immunity and oncogenesis has been more clearly defined, an understanding of the basic functions of NF-κB in the nervous system has lagged behind. The vast cell-type heterogeneity within the central nervous system (CNS) and the interplay between cell-type specific roles of NF-κB contributes to the complexity of understanding NF-κB functions in the brain. In this review, we will focus on the emerging understanding of cell-autonomous regulation of NF-κB signaling as well as the non-cell-autonomous functional impacts of NF-κB activation in the mammalian nervous system. We will focus on recent work which is unlocking the pleiotropic roles of NF-κB in neurons and glial cells (including astrocytes and microglia). Normal physiology as well as disorders of the CNS in which NF-κB signaling has been implicated will be discussed with reference to the lens of cell-type specific responses.
Collapse
Affiliation(s)
- Erica C Dresselhaus
- Department of Biological Chemistry and Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mollie K Meffert
- Department of Biological Chemistry and Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
39
|
Zeng F, Wang K, Huang R, Liu Y, Zhang Y, Hu H. RELB: A novel prognostic marker for glioblastoma as identified by population-based analysis. Oncol Lett 2019; 18:386-394. [PMID: 31289510 PMCID: PMC6540354 DOI: 10.3892/ol.2019.10296] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 03/22/2019] [Indexed: 12/21/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and malignant type of glioma, with a poor prognosis for patients. The survival time of patients varies greatly due to the complexity of the human genome, which harbors diverse oncogenic drivers. In order to identify the specific driving factors, 325 glioma samples from the Chinese Glioma Genome Atlas (CGGA) database were analyzed in the present study. The level of RELB proto-oncogene, NF-κβ subunit (RELB) expression increased with the pathological grade progression of the gliomas, and higher expression levels were present in the mesenchymal subtype and isocitrate dehydrogenase 1 (IDH1) wild-type gliomas. This RELB expression pattern was identified in the CGGA database and observed in three large independent databases. In patients with GBM from the CGGA database, a higher RELB expression level was associated with a shorter survival time, a mesenchymal subtype and IDH1 wild-type gliomas. Kaplan-Meier survival analysis, survival nomograms and Cox analysis demonstrated an independent prognostic value for RELB expression. Moreover, biological function analysis indicated the association of RELB with the ‘immune response’, ‘cell activation’ and the ‘apoptotic process’. In addition, RELB expression levels exhibited a negative correlation with the levels of microRNA (miR)-139-5p and miR-139-3p. The present study identified the pathological and biological roles of RELB in glioma and revealed its independent prognostic effect. These results suggested that RELB may be used as a prognostic biomarker and potential therapeutic target in glioma.
Collapse
Affiliation(s)
- Fan Zeng
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, P.R. China.,Chinese Glioma Cooperative Group (CGCG), Beijing 100070, P.R. China
| | - Kuanyu Wang
- Department of Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, P.R. China.,Chinese Glioma Cooperative Group (CGCG), Beijing 100070, P.R. China
| | - Ruoyu Huang
- Department of Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, P.R. China.,Chinese Glioma Cooperative Group (CGCG), Beijing 100070, P.R. China
| | - Yanwei Liu
- Department of Radiotherapy, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, P.R. China.,Chinese Glioma Cooperative Group (CGCG), Beijing 100070, P.R. China
| | - Ying Zhang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, P.R. China.,Chinese Glioma Cooperative Group (CGCG), Beijing 100070, P.R. China
| | - Huimin Hu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, P.R. China.,Chinese Glioma Cooperative Group (CGCG), Beijing 100070, P.R. China
| |
Collapse
|
40
|
Li S, Lv M, Qiu S, Meng J, Liu W, Zuo J, Yang L. NF-κB p65 promotes ovarian cancer cell proliferation and migration via regulating mortalin. J Cell Mol Med 2019; 23:4338-4348. [PMID: 30983127 PMCID: PMC6533498 DOI: 10.1111/jcmm.14325] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 03/05/2019] [Accepted: 03/25/2019] [Indexed: 12/30/2022] Open
Abstract
Previous studies show that mortalin, a HSP70 family member, contributes to the development and progression of ovarian cancer. However, details of the transcriptional regulation of mortalin remain unknown. We aimed to determine whether NF‐κB p65 participates in the regulation of mortalin expression in ovarian cancer cells and to elucidate the underlying mechanism. Chromatin immunoprecipitation and luciferase reporter assay were used to identify mortalin gene sequences, to which NF‐κB p65 binds. Results indicated that NF‐κB p65 binds to the mortalin promoter at a site with the sequence ‘CGGGGTTTCA’. Using lentiviral pLVX‐NF‐κB‐puro and Lentivirus‐delivered NF‐κB short hairpin RNA (shRNA), we created ovarian cancer cell lines in which NF‐κB p65 was stably up‐regulated and down‐regulated. Using these cells, we found that downregulation of NF‐κB p65 inhibits the growth and migration of ovarian cancer cells. Further experimental evidence indicated that downregulation of NF‐κB p65 reduced mortalin, and upregulation of mortalin rescued the proliferation and migration of ovarian cancer cells reduced by NF‐κB p65 knockdown. In conclusion, NF‐κB p65 binds to the mortalin promoter and promotes ovarian cancer cells proliferation and migration via regulating mortalin.
Collapse
Affiliation(s)
- Shan Li
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Mengyuan Lv
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Shi Qiu
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jiaqi Meng
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Wen Liu
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ji Zuo
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ling Yang
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
41
|
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
|
42
|
Zhou X, Shan Z, Yang H, Xu J, Li W, Guo F. RelB plays an oncogenic role and conveys chemo-resistance to DLD-1 colon cancer cells. Cancer Cell Int 2018; 18:181. [PMID: 30473630 PMCID: PMC6234565 DOI: 10.1186/s12935-018-0677-x] [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: 07/16/2018] [Accepted: 11/04/2018] [Indexed: 12/24/2022] Open
Abstract
Background Nuclear transcription factor kappa B (NF-κB) subunits exhibit crucial roles in tumorigenesis and chemo-sensitivity. Recent studies suggest that RelB, the key subunit of the alternative NF-κB pathway, plays a critical role in the progression of diverse human malignancies. However, the significance of RelB in colorectal cancer (CRC) remains unclear. Here, we systematically explored the functions of the alternative NF-κB subunit RelB in colon cancer cells and its underlying mechanism. Methods Stably transfected RelB-shRNA DLD-1 cells were established using Lipofectamine 2000. NF-κB DNA-binding capability was quantified using an ELISA-based NF-κB activity assay. Cell growth was monitored by an x-Celligence system. Cell proliferation was analyzed by a CCK-8 and a Brdu proliferation assay. Response to 5-FU was assessed by an x-Celligence system. Cell apoptosis and cell cycle was detected using flow cytometry analyses. Cell migration and invasion abilities were detected by an x-Celligence system, Transwell inserts, and wound-healing assays. RelB expression and its clinical significance were analyzed using the CRC tissue microarray. The expression of NF-κB signaling subunits, AKT/mTOR signaling molecules, cell cycle related proteins, MMP2, MMP9, and Integrin β-1 were measured by Western blotting analyses. Results The RelB-silencing inhibited cell growth of DLD-1 cells. The RelB-silencing exerted the anti-proliferative by downregulation of AKT/mTOR signaling. The RelB-silencing caused G0–G1 cell cycle arrested likely due to decreasing the expression of Cyclin D1 and CDK4, concomitant with increased expression of p27Kip1. The RelB-silencing enhanced cytotoxic effect of 5-FU and induced cell accumulation in S-phase. The RelB-silencing impaired the migration and invasion potential of DLD-1 cells, which was related to downregulation of MMP2, MMP9, and Integrin β-1. Importantly, the RelB expression was correlated with depth of tumor invasion, lymph node metastasis, metastasis stage, and pTNM stage. High-RelB expression was significantly correlated with poor overall survival in CRC patients. Conclusion Our studies here provided evidence that RelB plays an oncogenic role and conveys chemo-resistance to 5-FU. RelB can be considered as an independent indicator of prognosis in CRC.
Collapse
Affiliation(s)
- Xiaojun Zhou
- 1Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006 China
| | - Zhili Shan
- 1Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006 China
| | - Hengying Yang
- 1Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006 China
| | - Jingjing Xu
- 2Center for Clinical Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, 215006 China
| | - Wenjing Li
- 3Department of Clinical Laboratory, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, 215006 China
| | - Feng Guo
- 4Department of Oncology, Nanjing Medical University Affiliated Suzhou Hospital, Baita West Road 16, Suzhou, 215001 China
| |
Collapse
|
43
|
Yamini B. NF-κB, Mesenchymal Differentiation and Glioblastoma. Cells 2018; 7:cells7090125. [PMID: 30200302 PMCID: PMC6162779 DOI: 10.3390/cells7090125] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/14/2018] [Accepted: 08/30/2018] [Indexed: 12/11/2022] Open
Abstract
Although glioblastoma (GBM) has always been recognized as a heterogeneous tumor, the advent of largescale molecular analysis has enabled robust categorization of this malignancy into several specific subgroups. Among the subtypes designated by expression profiling, mesenchymal tumors have been associated with an inflammatory microenvironment, increased angiogenesis, and resistance to therapy. Nuclear factor-κB (NF-κB) is a ubiquitous transcription factor that plays a prominent role in mediating many of the central features associated with mesenchymal differentiation. This review summarizes the mechanisms by which NF-κB proteins and their co-regulating partners induce the transcriptional network that underlies the mesenchymal phenotype. Moreover, both the intrinsic changes within mesenchymal GBM cells and the microenvironmental factors that modify the overall NF-κB response are detailed.
Collapse
Affiliation(s)
- Bakhtiar Yamini
- Section of Neurosurgery Department of Surgery, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
44
|
Henry KL, Kellner D, Bajrami B, Anderson JE, Beyna M, Bhisetti G, Cameron T, Capacci AG, Bertolotti-Ciarlet A, Feng J, Gao B, Hopkins B, Jenkins T, Li K, May-Dracka T, Murugan P, Wei R, Zeng W, Allaire N, Buckler A, Loh C, Juhasz P, Lucas B, Ennis KA, Vollman E, Cahir-McFarland E, Hett EC, Ols ML. CDK12-mediated transcriptional regulation of noncanonical NF-κB components is essential for signaling. Sci Signal 2018; 11:eaam8216. [PMID: 30065029 DOI: 10.1126/scisignal.aam8216] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Members of the family of nuclear factor κB (NF-κB) transcription factors are critical for multiple cellular processes, including regulating innate and adaptive immune responses, cell proliferation, and cell survival. Canonical NF-κB complexes are retained in the cytoplasm by the inhibitory protein IκBα, whereas noncanonical NF-κB complexes are retained by p100. Although activation of canonical NF-κB signaling through the IκBα kinase complex is well studied, few regulators of the NF-κB-inducing kinase (NIK)-dependent processing of noncanonical p100 to p52 and the subsequent nuclear translocation of p52 have been identified. We discovered a role for cyclin-dependent kinase 12 (CDK12) in transcriptionally regulating the noncanonical NF-κB pathway. High-content phenotypic screening identified the compound 919278 as a specific inhibitor of the lymphotoxin β receptor (LTβR), and tumor necrosis factor (TNF) receptor superfamily member 12A (FN14)-dependent nuclear translocation of p52, but not of the TNF-α receptor-mediated nuclear translocation of p65. Chemoproteomics identified CDK12 as the target of 919278. CDK12 inhibition by 919278, the CDK inhibitor THZ1, or siRNA-mediated knockdown resulted in similar global transcriptional changes and prevented the LTβR- and FN14-dependent expression of MAP3K14 (which encodes NIK) as well as NIK accumulation by reducing phosphorylation of the carboxyl-terminal domain of RNA polymerase II. By coupling a phenotypic screen with chemoproteomics, we identified a pathway for the activation of the noncanonical NF-κB pathway that could serve as a therapeutic target in autoimmunity and cancer.
Collapse
Affiliation(s)
- Kate L Henry
- Biogen, 225 Binney Street, Cambridge, MA 02142, USA
- Department of Pharmacology, Boston University School of Medicine, Boston, MA 02118, USA
| | | | | | - John E Anderson
- Biogen, 225 Binney Street, Cambridge, MA 02142, USA
- Department of Pharmacology, Boston University School of Medicine, Boston, MA 02118, USA
| | | | | | - Tom Cameron
- Biogen, 225 Binney Street, Cambridge, MA 02142, USA
| | | | | | - Jun Feng
- Biogen, 225 Binney Street, Cambridge, MA 02142, USA
| | - Benbo Gao
- Biogen, 225 Binney Street, Cambridge, MA 02142, USA
| | | | | | - Kejie Li
- Biogen, 225 Binney Street, Cambridge, MA 02142, USA
| | | | | | - Ru Wei
- Biogen, 225 Binney Street, Cambridge, MA 02142, USA
| | - Weike Zeng
- Biogen, 225 Binney Street, Cambridge, MA 02142, USA
| | - Norm Allaire
- Biogen, 225 Binney Street, Cambridge, MA 02142, USA
| | - Alan Buckler
- Biogen, 225 Binney Street, Cambridge, MA 02142, USA
| | | | - Peter Juhasz
- Biogen, 225 Binney Street, Cambridge, MA 02142, USA
| | - Brian Lucas
- Biogen, 225 Binney Street, Cambridge, MA 02142, USA
| | | | | | | | - Erik C Hett
- Biogen, 225 Binney Street, Cambridge, MA 02142, USA.
| | | |
Collapse
|
45
|
Chen W, Xia T, Wang D, Huang B, Zhao P, Wang J, Qu X, Li X. Human astrocytes secrete IL-6 to promote glioma migration and invasion through upregulation of cytomembrane MMP14. Oncotarget 2018; 7:62425-62438. [PMID: 27613828 PMCID: PMC5308737 DOI: 10.18632/oncotarget.11515] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 08/08/2016] [Indexed: 01/23/2023] Open
Abstract
The brain microenvironment has emerged as an important component in malignant progression of human glioma. However, astrocytes, the most abundant glial cells in the glioma microenvironment, have as yet a poorly defined role in the development of this disease, particularly with regard to invasion. Here, we co-cultured human astrocytes with human glioma cell lines, U251 and A172, in an in vitro transwell system in order to ascertain their influence on migration and invasion of gliomas. mRNA and protein expression assays were subsequently used to identify candidate proteins mediating this activity. Astrocytes significantly increased migration and invasion of both U251 and A172 cells in migration and invasion (plus matrigel) assays. Membrane type 1 matrix metalloproteinase (MMP14) originating from glioma cells was identified in qRT-PCR as the most highly up-regulated member of the MMP family of genes (~ 3 fold, p < 0.05) in this system. A cytokine array and ELISA were used to identify interleukin-6 (IL-6) as a highly increased factor in media collected from astrocytes, especially under co-culture conditions. IL-6 was also the key cytokine inducing cytomembrane MMP14 expression, the active form of MMP14, in glioma cells. Knockdown of MMP14 with siRNA led to decreased migration and invasion. Taken together, our results indicated that cytomembrane MMP14 was induced by IL-6 secreted from astrocytes, thereby enhancing the migration and invasion of glioma cells through activation of MMP2. Therefore, this IL-6 and MMP14 axis between astrocytes and glioma cells may become a potential target for treatment of glioma patients.
Collapse
Affiliation(s)
- Weiliang Chen
- Department of Otolaryngology, Qilu Hospital, Shandong University, Jinan, China.,Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, China.,Key Laboratory of Otolaryngology, Chinese Ministry of Health, Jinan, China
| | - Tongliang Xia
- Department of Otolaryngology, Qilu Hospital, Shandong University, Jinan, China.,Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, China.,Key Laboratory of Otolaryngology, Chinese Ministry of Health, Jinan, China
| | - Donghai Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, China
| | - Bin Huang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, China
| | - Peng Zhao
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, China
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, China.,Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Xun Qu
- Institute of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, China
| |
Collapse
|
46
|
Hersh DS, Peng S, Dancy JG, Galisteo R, Eschbacher JM, Castellani RJ, Heath JE, Legesse T, Kim AJ, Woodworth GF, Tran NL, Winkles JA. Differential expression of the TWEAK receptor Fn14 in IDH1 wild-type and mutant gliomas. J Neurooncol 2018; 138:241-250. [PMID: 29453678 DOI: 10.1007/s11060-018-2799-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 02/08/2018] [Indexed: 01/22/2023]
Abstract
The TNF receptor superfamily member Fn14 is overexpressed by many solid tumor types, including glioblastoma (GBM), the most common and lethal form of adult brain cancer. GBM is notable for a highly infiltrative growth pattern and several groups have reported that high Fn14 expression levels can increase tumor cell invasiveness. We reported previously that the mesenchymal and proneural GBM transcriptomic subtypes expressed the highest and lowest levels of Fn14 mRNA, respectively. Given the recent histopathological re-classification of human gliomas by the World Health Organization based on isocitrate dehydrogenase 1 (IDH1) gene mutation status, we extended this work by comparing Fn14 gene expression in IDH1 wild-type (WT) and mutant (R132H) gliomas and in cell lines engineered to overexpress the IDH1 R132H enzyme. We found that both low-grade and high-grade (i.e., GBM) IDH1 R132H gliomas exhibit low Fn14 mRNA and protein levels compared to IDH1 WT gliomas. Forced overexpression of the IDH1 R132H protein in glioma cells reduced Fn14 expression, while treatment of IDH1 R132H-overexpressing cells with the IDH1 R132H inhibitor AGI-5198 or the DNA demethylating agent 5-aza-2'-deoxycytidine increased Fn14 expression. These results support a role for Fn14 in the more aggressive and invasive phenotype associated with IDH1 WT tumors and indicate that the low levels of Fn14 gene expression noted in IDH1 R132H mutant gliomas may be due to epigenetic regulation via changes in DNA methylation.
Collapse
Affiliation(s)
- David S Hersh
- Department of Neurosurgery, University of Maryland School of Medicine, 22 S. Greene St Suite 12D, Baltimore, MD, 21201, USA
| | - Sen Peng
- Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, AZ, 85004, USA
| | - Jimena G Dancy
- Department of Neurosurgery, University of Maryland School of Medicine, 22 S. Greene St Suite 12D, Baltimore, MD, 21201, USA
| | - Rebeca Galisteo
- Department of Surgery, University of Maryland School of Medicine, 22 S. Greene St, Baltimore, MD, 21201, USA.,Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, UMB BioPark One Room 320, 800 West Baltimore St, Baltimore, MD, 21201, USA
| | - Jennifer M Eschbacher
- Department of Neuropathology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, 85013, USA
| | - Rudy J Castellani
- Department of Pathology, University of Maryland School of Medicine, 22 S. Greene St, Baltimore, MD, 21201, USA
| | - Jonathan E Heath
- Department of Pathology, University of Maryland School of Medicine, 22 S. Greene St, Baltimore, MD, 21201, USA
| | - Teklu Legesse
- Department of Pathology, University of Maryland School of Medicine, 22 S. Greene St, Baltimore, MD, 21201, USA
| | - Anthony J Kim
- Department of Neurosurgery, University of Maryland School of Medicine, 22 S. Greene St Suite 12D, Baltimore, MD, 21201, USA.,University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, 22 S. Greene St, Baltimore, MD, 21201, USA
| | - Graeme F Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine, 22 S. Greene St Suite 12D, Baltimore, MD, 21201, USA.,University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, 22 S. Greene St, Baltimore, MD, 21201, USA
| | - Nhan L Tran
- Departments of Cancer Biology and Neurosurgery, Mayo Clinic Arizona, Scottsdale, AZ, 85259, USA
| | - Jeffrey A Winkles
- Department of Surgery, University of Maryland School of Medicine, 22 S. Greene St, Baltimore, MD, 21201, USA. .,Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, UMB BioPark One Room 320, 800 West Baltimore St, Baltimore, MD, 21201, USA. .,University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, 22 S. Greene St, Baltimore, MD, 21201, USA.
| |
Collapse
|
47
|
Zhu C, Zhang L, Liu Z, Li C, Bai Y. TWEAK/Fn14 interaction induces proliferation and migration in human airway smooth muscle cells via activating the NF-κB pathway. J Cell Biochem 2018; 119:3528-3536. [PMID: 29143982 DOI: 10.1002/jcb.26525] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 11/13/2017] [Indexed: 12/11/2022]
Abstract
Asthma, an increasingly common chronic disease among children, are characterized by airway remodeling, which is partly attributed to the proliferation and migration of airway smooth muscle cell (ASMC). The purpose of the present study was to investigate potential roles and mechanisms of the tumor necrosis factor-like weak inducer of apoptosis (TWEAK)/fibroblast growth factor-inducible molecule 14 (Fn14) axis on cell proliferation and migration in HASMCs. Compared to HASMCs from non-asthmatic patients, those from asthmatic patients showed elevated expression levels of both Fn14 and TWEAK. Additionally, similar to the response triggered by platelet-derived growth factor-BB, stimulation with recombinant TWEAK strongly induced cell proliferation and migration in HASMCs. However, depletion of Fn14 remarkably abrogated the enhancement of TWEAK on the cell proliferation and migration of HASMCs. Furthermore, treatment with TWEAK led to the activation of NF-κB. This effect was eliminated by silencing Fn14, indicating that TWEAK-induced NF-κB signaling was mediated via Fn14. Moreover, the TWEAK/Fn14 interaction promoted cell proliferation and migration. These effects were blocked by NF-κB inhibitor SN50, which suggest that the TWEAK/Fn14 signaling system partially depends on NF-κB activity. Collectively, we demonstrated that the TWEAK/Fn14 axis accelerated HASMC cell proliferation and migration by activating the NF-κB pathway, thereby exacerbating airway remodeling in asthma. Altogether, these findings indicate a novel role for the TWEAK/Fn14/NF-κB pathway as a potent option for limiting airway remodeling in asthma.
Collapse
Affiliation(s)
- Cuimin Zhu
- Department of Pediatrics, Cangzhou Central Hospital, Cangzhou, Hebei, PR China
| | - Leguo Zhang
- Department of Internal Neurology, Cangzhou Central Hospital, Cangzhou, Hebei, PR China
| | - Zhiming Liu
- Department of Pediatrics, Cangzhou Central Hospital, Cangzhou, Hebei, PR China
| | - Chen Li
- Department of Pediatrics, Cangzhou Central Hospital, Cangzhou, Hebei, PR China
| | - Yajie Bai
- Department of Pediatrics, Cangzhou Central Hospital, Cangzhou, Hebei, PR China
| |
Collapse
|
48
|
Involvement of TWEAK and the NF-κB signaling pathway in lupus nephritis. Exp Ther Med 2018; 15:2611-2619. [PMID: 29456665 DOI: 10.3892/etm.2018.5711] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 08/28/2017] [Indexed: 11/05/2022] Open
Abstract
Previous findings have identified that tumor necrosis factor-related weak inducer of apoptosis (TWEAK) is associated with lupus nephritis (LN) activity status; however, the mechanism involved remains unclear. The present study aimed to investigate the roles of TWEAK and the nuclear factor (NF)-κB signaling pathway in LN. TWEAK levels in the blood and urine of patients with LN or non-LN systemic lupus erythematosus were measured by ELISA and compared with those in healthy controls. TWEAK expression and NF-κB transcriptional activity in the kidney were detected by western blotting, and Ki-67 and cluster of differentiation (CD) 68 expression were assessed using immunofluorescence. Additionally, human mesangial cells (HMCs) were cultured in vitro and divided into five groups: Normal control, TWEAK stimulus group, TWEAK + TWEAK blocking antibody, TWEAK + NF-κB inhibitor (BAY 11-7082) and TWEAK + combined (blocking antibody + BAY 11-7082). Cell cycle activity and Ki-67 expression in the HMCs were evaluated using flow cytometry, and cell induction of macrophage chemotaxis was determined by a Transwell assay. Levels of the inflammation-associated factors interleukin (IL)-6, monocyte chemotactic protein 1 (MCP-1), chemokine ligand 5 (CCL5), IL-8 and IL-10 were also detected by reverse transcription-quantitative polymerase chain reaction. It was observed that the urine levels of TWEAK in patients with LN were significantly elevated compared with those in the other groups (P<0.05). LN kidneys exhibited markedly increased cell proliferative ability, macrophage infiltration, TWEAK expression and NF-κB transcriptional activity compared with normal kidneys. Furthermore, the results indicated that treatment with recombinant TWEAK notably enhanced NF-κB transcriptional activity and significantly promoted cell proliferation and cell cycle activity (P<0.05), induced macrophage chemotaxis (P<0.05), significantly increased the expression of the chemotactic factors IL-6, IL-8, MCP-1 and CCL5 (P<0.05), and significantly reduced anti-inflammatory cytokine IL-10 mRNA expression in HMCs (P<0.05), relative to normal controls. Accordingly, blocking TWEAK function or inhibiting NF-κB activity reversed these effects. Collectively these data indicate that urine TWEAK may be considered as a novel biomarker of LN activity, and that blocking TWEAK function or NF-κB activity may effectively alleviate glomerular mesangial cell proliferation and macrophage chemotaxis.
Collapse
|
49
|
Boulamery A, Desplat-Jégo S. Regulation of Neuroinflammation: What Role for the Tumor Necrosis Factor-Like Weak Inducer of Apoptosis/Fn14 Pathway? Front Immunol 2017; 8:1534. [PMID: 29201025 PMCID: PMC5696327 DOI: 10.3389/fimmu.2017.01534] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 10/27/2017] [Indexed: 12/25/2022] Open
Abstract
Observed in many central nervous system diseases, neuroinflammation (NI) proceeds from peripheral immune cell infiltration into the parenchyma, from cytokine secretion and from oxidative stress. Astrocytes and microglia also get activated and proliferate. NI manifestations and consequences depend on its context and on the acute or chronic aspect of the disease. The tumor necrosis factor-like weak inducer of apoptosis (TWEAK)/Fn14 pathway has been involved in chronic human inflammatory pathologies such as neurodegenerative, autoimmune, or malignant diseases. New data now describe its regulatory effects in tissues or fluids from patients with neurological diseases. In this mini-review, we aim to highlight the role of TWEAK/Fn14 in modulating NI in multiple sclerosis, neuropsychiatric systemic lupus erythematosus, stroke, or glioma. TWEAK/Fn14 can modulate NI by activating canonical and non-canonical nuclear factor-κB pathways but also by stimulating mitogen-activated protein kinase signaling. These downstream activations are associated with (i) inflammatory cytokine, chemokine and adhesion molecule expression or release, involved in NI propagation, (ii) matrix-metalloproteinase 9 secretion, implicated in blood–brain barrier disruption and tissue remodeling, (iii) astrogliosis and microgliosis, and (iv) migration of tumor cells in glioma. In addition, we report several animal and human studies pointing to TWEAK as an attractive therapeutic target.
Collapse
Affiliation(s)
- Audrey Boulamery
- Aix-Marseille University, CNRS, NICN, Marseille, France.,AP-HM, Hôpital Sainte-Marguerite, Centre Antipoison et de Toxicovigilance, Marseille, France
| | - Sophie Desplat-Jégo
- Aix-Marseille University, CNRS, NICN, Marseille, France.,Service d'Immunologie, Hôpital de la Conception, Marseille, France
| |
Collapse
|
50
|
Guan F, Wang L, Hao S, Wu Z, Bai J, Kang Z, Zhou Q, Chang H, Yin H, Li D, Tian K, Ma J, Zhang G, Zhang J. Retinol dehydrogenase-10 promotes development and progression of human glioma via the TWEAK-NF-κB axis. Oncotarget 2017; 8:105262-105275. [PMID: 29285249 PMCID: PMC5739636 DOI: 10.18632/oncotarget.22166] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 09/25/2017] [Indexed: 12/03/2022] Open
Abstract
Retinol dehydrogenase-10 (RDH10) is a member of the short-chain dehydrogenase/reductase family, which plays an important role in retinoic acid (RA) synthesis. Here, we show that RDH10 is highly expressed in human gliomas, and its expression correlates with tumor grade and patient survival times. In vitro, lentivirus-mediated shRNA knockdown of RDH10 suppressed glioma cell proliferation, survival, and invasiveness and cell cycle progression. In vivo, RDH10 knockdown reduced glioma growth in nude mice. Microarray analysis revealed that RDH10 silencing reduces expression of TNFRSF12A (Fn14), TNFSF12 (TWEAK), TRAF3, IKBKB (IKK-β), and BMPR2, while it increases expression of TRAF1, NFKBIA (IκBα), NFKBIE (IκBε), and TNFAIP3. This suggests that RDH10 promotes glioma cell proliferation and survival by regulating the TWEAK-NF-κB axis, and that it could potentially serve as a novel target for human glioma treatment.
Collapse
Affiliation(s)
- Feng Guan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Liang Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shuyu Hao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhen Wu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jian Bai
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Zhuang Kang
- Department of Glioma, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Quan Zhou
- Department of Pathology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Hong Chang
- Department of Pathology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Hui Yin
- Department of Social Medicine and Health Education, School of Public Health, Peking University, Beijing, China
| | - Da Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Kaibin Tian
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Junpeng Ma
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Guijun Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Junting Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|