1
|
Mozaffari S, Moen A, Ng CY, Nicolaes GA, Wichapong K. Structural bioinformatics for rational drug design. Res Pract Thromb Haemost 2025; 9:102691. [PMID: 40027444 PMCID: PMC11869865 DOI: 10.1016/j.rpth.2025.102691] [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: 10/24/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 03/05/2025] Open
Abstract
A State of the Art lecture titled "structural bioinformatics technologies for rational drug design: from in silico to in vivo" was presented at the International Society on Thrombosis and Haemostasis (ISTH) Congress in 2024. Drug discovery remains a resource-intensive and complex endeavor, which usually takes over a decade and costs billions to bring a new therapeutic agent to market. However, the landscape of drug discovery has been transformed by the recent advancements in bioinformatics and cheminformatics. Key techniques, including structure- and ligand-based virtual screening, molecular dynamics simulations, and artificial intelligence-driven models are allowing researchers to explore vast chemical spaces, investigate molecular interactions, predict binding affinity, and optimize drug candidates with unprecedented accuracy and efficiency. These computational methods complement experimental techniques by accelerating the identification of viable drug candidates and refining lead compounds. Artificial intelligence models, alongside traditional physics-based simulations, now play an important role in predicting key properties such as binding affinity and toxicity, contributing to more informed decision-making, particularly early in the drug discovery process. Despite these advancements, challenges remain in terms of accuracy, interpretability, and the needed computational power. This review explores the state of the art in computational drug discovery, examining the latest methods and technologies, their transformative impact on the drug development pipeline, and the future directions needed to overcome remaining limitations. Finally, we summarize relevant data and highlight cases where various computational approaches were successfully applied to develop novel inhibitors, as presented during the ISTH 2024 Congress.
Collapse
Affiliation(s)
- Soroush Mozaffari
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Agnethe Moen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Che Yee Ng
- Hillmark B.V., Maastricht, the Netherlands
| | - Gerry A.F. Nicolaes
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
- Hillmark B.V., Maastricht, the Netherlands
| | - Kanin Wichapong
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
- Hillmark B.V., Maastricht, the Netherlands
| |
Collapse
|
2
|
Raizada S, Obukhov AG, Bharti S, Wadhonkar K, Baig MS. Pharmacological targeting of adaptor proteins in chronic inflammation. Inflamm Res 2024; 73:1645-1656. [PMID: 39052063 DOI: 10.1007/s00011-024-01921-5] [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: 02/13/2024] [Revised: 05/28/2024] [Accepted: 07/11/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Inflammation, a biological response of the immune system, can be triggered by various factors such as pathogens, damaged cells, and toxic compounds. These factors can lead to chronic inflammatory responses, potentially causing tissue damage or disease. Both infectious and non-infectious agents, as well as cell damage, activate inflammatory cells and trigger common inflammatory signalling pathways, including NF-κB, MAPK, and JAK-STAT pathways. These pathways are activated through adaptor proteins, which possess distinct protein binding domains that connect corresponding interacting molecules to facilitate downstream signalling. Adaptor molecules have gained widespread attention in recent years due to their key role in chronic inflammatory diseases. METHODS In this review, we explore potential pharmacological agents that can be used to target adaptor molecules in chronic inflammatory responses. A comprehensive analysis of published studies was performed to obtain information on pharmacological agents. CONCLUSION This review highlights the therapeutic strategies involving small molecule inhibitors, antisense oligonucleotide therapy, and traditional medicinal compounds that have been found to inhibit the inflammatory response and pro-inflammatory cytokine production. These strategies primarily block the protein-protein interactions in the inflammatory signaling cascade. Nevertheless, extensive preclinical studies and risk assessment methodologies are necessary to ensure their safety.
Collapse
Affiliation(s)
- Shubhi Raizada
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, 453552, MP, India
| | - Alexander G Obukhov
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Shreya Bharti
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, 453552, MP, India
| | - Khandu Wadhonkar
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, 453552, MP, India
| | - Mirza S Baig
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, 453552, MP, India.
| |
Collapse
|
3
|
Wang PF, Jiang F, Zeng QM, Yin WF, Hu YZ, Li Q, Hu ZL. Mitochondrial and metabolic dysfunction of peripheral immune cells in multiple sclerosis. J Neuroinflammation 2024; 21:28. [PMID: 38243312 PMCID: PMC10799425 DOI: 10.1186/s12974-024-03016-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 01/08/2024] [Indexed: 01/21/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disorder characterized by the infiltration of inflammatory cells and demyelination of nerves. Mitochondrial dysfunction has been implicated in the pathogenesis of MS, as studies have shown abnormalities in mitochondrial activities, metabolism, mitochondrial DNA (mtDNA) levels, and mitochondrial morphology in immune cells of individuals with MS. The presence of mitochondrial dysfunctions in immune cells contributes to immunological dysregulation and neurodegeneration in MS. This review provided a comprehensive overview of mitochondrial dysfunction in immune cells associated with MS, focusing on the potential consequences of mitochondrial metabolic reprogramming on immune function. Current challenges and future directions in the field of immune-metabolic MS and its potential as a therapeutic target were also discussed.
Collapse
Affiliation(s)
- Peng-Fei Wang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, 139 Ren-Min Central Road, Changsha City, 410011, Hunan, China
| | - Fei Jiang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha City, 410011, Hunan, China
| | - Qiu-Ming Zeng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha City, 410011, Hunan, China
| | - Wei-Fan Yin
- Department of Neurology, The Second Xiangya Hospital, Central South University, 139 Ren-Min Central Road, Changsha City, 410011, Hunan, China
| | - Yue-Zi Hu
- Clinical Laboratory, The Second Hospital of Hunan University of Chinese Medicine, 233 Cai' e North Road, Changsha City, 410005, Hunan, China
| | - Qiao Li
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, 139 Ren-Min Central Road, Changsha City, 410011, Hunan, China
| | - Zhao-Lan Hu
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, 139 Ren-Min Central Road, Changsha City, 410011, Hunan, China.
| |
Collapse
|
4
|
Fu XX, Qu H, Wang J, Cai HY, Jiang H, Chen HH, Han S. Novel nano-carriers with N-formylmethionyl-leucyl-phenylalanine-modified liposomes improve effects of C16-angiopoietin 1 in acute animal model of multiple sclerosis. Drug Deliv 2023; 30:2241664. [PMID: 37545034 PMCID: PMC10987045 DOI: 10.1080/10717544.2023.2241664] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 07/17/2023] [Indexed: 08/08/2023] Open
Abstract
Gradual loss of neuronal structure and function due to impaired blood-brain barrier (BBB) and neuroinflammation are important factors in multiple sclerosis (MS) progression. Our previous studies demonstrated that the C16 peptide and angiopoietin 1 (Ang-1) compound (C + A) could modulate inflammation and vascular protection in many models of MS. In this study, nanotechnology and a novel nanovector of the leukocyte chemotactic peptide N-formyl-methionyl-leucyl-phenylalanine (fMLP) were used to examine the effects of C + A on MS. The acute experimental autoimmune encephalomyelitis (EAE) model of MS was established in Lewis rats. The C + A compounds were conjugated to control nano-carriers and fMLP-nano-carriers and administered to animals by intravenous injection. The neuropathological changes in the brain cortex and spinal cord were examined using multiple approaches. The stimulation of vascular injection sites was examined using rabbits. The results showed that all C + A compounds (C + A alone, nano-carrier C + A, and fMLP-nano-carrier C + A) reduced neuronal inflammation, axonal demyelination, gliosis, neuronal apoptosis, vascular leakage, and BBB impairment induced by EAE. In addition, the C + A compounds had minimal side effects on liver and kidney functions. Furthermore, the fMLP-nano-carrier C + A compound had better effects compared to C + A alone and the nano-carrier C + A. This study indicated that the fMLP-nano-carrier C + A could attenuate inflammation-related pathological changes in EAE and may be a potential therapeutic strategy for the treatment of MS and EAE.
Collapse
Affiliation(s)
- Xiao-Xiao Fu
- Institute of Anatomy and Cell Biology, Medical College, Zhejiang University, Hangzhou, PR China
- Institute of Human Anatomy, Histology and Embryology, Basic Medical College, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Han Qu
- Institute of Anatomy and Cell Biology, Medical College, Zhejiang University, Hangzhou, PR China
| | - Jing Wang
- Department of Neurology, Sir Run Run Shaw Hospital, Medical College, Zhejiang University, Hangzhou, PR China
| | - Hua-Ying Cai
- Department of Neurology, Sir Run Run Shaw Hospital, Medical College, Zhejiang University, Hangzhou, PR China
| | - Hong Jiang
- Department of Electrophysiology, Sir Run Run Shaw Hospital, Medical College, Zhejiang University, Hangzhou, PR China
| | - Hao-Hao Chen
- Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, Jinhua, PR China
| | - Shu Han
- Institute of Anatomy and Cell Biology, Medical College, Zhejiang University, Hangzhou, PR China
| |
Collapse
|
5
|
Marengo M, Migliori M, Merlotti G, Fabbrini P, Panichi V, Cantaluppi V. Role of the CD40-CD40 Ligand Pathway in Cardiovascular Events, Neurological Alterations, and Other Clinical Complications of Chronic Hemodialysis Patients: Protective Role of Adsorptive Membranes. Blood Purif 2023; 52 Suppl 1:27-42. [PMID: 37331328 PMCID: PMC10568606 DOI: 10.1159/000530808] [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: 02/21/2023] [Accepted: 04/16/2023] [Indexed: 06/20/2023]
Abstract
Despite the recent advances in dialysis technology, mortality rate of chronic uremic patients still remains excessively high: of note, in comparison to age- and sex-matched healthy controls, this frail population shows a higher incidence of infections, cancer, cognitive decline, and, in particular, major adverse cardiovascular events (MACE) that represent nowadays the first cause of mortality. Several traditional and nontraditional factors contribute to this increased risk for MACE and accelerated cellular senescence: among these, inflammation has been shown to play a key role. The costimulatory pathway CD40-CD40 Ligand (CD40L) is harmfully activated during inflammation and uremia-associated clinical complications: in particular, the soluble form of CD40L (sCD40L) can bind to the CD40 receptor triggering a cascade of detrimental pathways in immune and nonimmune cells. In this narrative review, we summarize the current concepts of the biological role of the CD40-CD40L pathway in uremia-associated organ dysfunction, focusing on the above-described main causes of mortality. Moreover, we discuss the interaction of the CD40-CD40L pathway with extracellular vesicles, microparticles recently identified as new uremic toxins. The biological effects of sCD40L in MACE, cognitive decline, infections, and cancer will be also briefly commented. Last, based on recent studies and ongoing clinical trials, we herein describe the modulatory activity of adsorptive dialysis membranes in polymethylmethacrylate on CD40-CD40L detrimental activation.
Collapse
Affiliation(s)
| | | | - Guido Merlotti
- Department of Translational Medicine (DIMET), Nephrology and Kidney Transplantation Unit, "Maggiore della Carità" University Hospital, University of Piemonte Orientale (UPO), Novara, Italy,
| | - Paolo Fabbrini
- Nephrology and Dialysis Unit, ASST Nord Milano, "Bassini" Hospital, Cinisello Balsamo, Italy
| | - Vincenzo Panichi
- Nephrology, Dialysis and Kidney Transplantation Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Vincenzo Cantaluppi
- Department of Translational Medicine (DIMET), Nephrology and Kidney Transplantation Unit, "Maggiore della Carità" University Hospital, University of Piemonte Orientale (UPO), Novara, Italy
| |
Collapse
|
6
|
Liu Y, Chen S, Liu S, Wallace KL, Zille M, Zhang J, Wang J, Jiang C. T-cell receptor signaling modulated by the co-receptors: Potential targets for stroke treatment. Pharmacol Res 2023; 192:106797. [PMID: 37211238 DOI: 10.1016/j.phrs.2023.106797] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/02/2023] [Accepted: 05/16/2023] [Indexed: 05/23/2023]
Abstract
Stroke is a severe and life-threatening disease, necessitating more research on new treatment strategies. Infiltrated T lymphocytes, an essential adaptive immune cell with extensive effector function, are crucially involved in post-stroke inflammation. Immediately after the initiation of the innate immune response triggered by microglia/macrophages, the adaptive immune response associated with T lymphocytes also participates in the complex pathophysiology of stroke and partially informs the outcome of stroke. Preclinical and clinical studies have revealed the conflicting roles of T cells in post-stroke inflammation and as potential therapeutic targets. Therefore, exploring the mechanisms that underlie the adaptive immune response associated with T lymphocytes in stroke is essential. The T-cell receptor (TCR) and its downstream signaling regulate T lymphocyte differentiation and activation. This review comprehensively summarizes the various molecules that regulate TCR signaling and the T-cell response. It covers both the co-stimulatory and co-inhibitory molecules and their roles in stroke. Because immunoregulatory therapies targeting TCR and its mediators have achieved great success in some proliferative diseases, this article also summarizes the advances in therapeutic strategies related to TCR signaling in lymphocytes after stroke, which can facilitate translation. DATA AVAILABILITY: No data was used for the research described in the article.
Collapse
Affiliation(s)
- Yuanyuan Liu
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China
| | - Shuai Chen
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China
| | - Simon Liu
- Medical Genomics Unit, National Human Genome Research Institute, Bethesda, MD, 20814, USA
| | - Kevin L Wallace
- College of Mathematical and Natural Sciences, University of Maryland, College Park, MD, 20742, USA
| | - Marietta Zille
- Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of Vienna, A-1090 Vienna, Austria
| | - Jiewen Zhang
- Department of Neurology, People's Hospital of Zhengzhou University, 450000, Zhengzhou, P. R. China.
| | - Jian Wang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China; Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, 450001, Zhengzhou, P. R. China.
| | - Chao Jiang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China.
| |
Collapse
|
7
|
Huang Y, Wu J, Zhan C, Liu R, Zhou Z, Huang X, Tian Y, Lin Z, Song Z. TRAF-STOP alleviates osteoclastogenesis in periodontitis. Front Pharmacol 2023; 14:1119847. [PMID: 37261283 PMCID: PMC10229065 DOI: 10.3389/fphar.2023.1119847] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 05/02/2023] [Indexed: 06/02/2023] Open
Abstract
The enhanced osteoclastogenesis contributes to alveolar bone resorption in periodontitis, which increases the risk of tooth loss. To reduce bone destruction, the inhibition of osteoclast development is proposed as a feasible treatment. CD40L-CD40-TRAF6 signal transduction plays a crucial role in inflammation, but how it regulates osteoclast activity in periodontitis has not been elucidated. In this study, we showed the potential role of CD40L-CD40-TRAF6 signaling in periodontitis. CD40L obviously promoted osteoclast formation and bone resorption capacity in vitro. Mechanistically, we found that osteoclastogenesis was enhanced by the overexpression of NFATc1 and NF-κB activation. Importantly, osteoclast activity was effectively suppressed by TRAF-STOP, a small molecular inhibitor of TRAF6. Furthermore, local injection of TRAF-STOP-loaded injectable PLGA-PEG-PLGA hydrogel could alleviate ligation-induced periodontitis in vivo. Taken together, TRAF-STOP shows promising clinical efficacy in periodontitis through alleviating osteoclastogenesis.
Collapse
Affiliation(s)
- Yaxian Huang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, Guangzhou, China
| | - Jinyan Wu
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, Guangzhou, China
| | - Chi Zhan
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, Guangzhou, China
| | - Rong Liu
- School of Medicine, South China University of Technology, Guangzhou 510006, Guangzhou, China
| | - Zhaocai Zhou
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, Guangzhou, China
| | - Xin Huang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, Guangzhou, China
| | - Yaguang Tian
- Department of Stomatology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou 570311, Hainan, China
| | - Zhengmei Lin
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, Guangzhou, China
| | - Zhi Song
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, Guangzhou, China
| |
Collapse
|
8
|
Lu Y, Xu M, Dorrier CE, Zhang R, Mayer CT, Wagner D, McGavern DB, Hodes RJ. CD40 Drives Central Nervous System Autoimmune Disease by Inducing Complementary Effector Programs via B Cells and Dendritic Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:2083-2092. [PMID: 36426970 PMCID: PMC10065987 DOI: 10.4049/jimmunol.2200439] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 09/19/2022] [Indexed: 01/04/2023]
Abstract
Costimulatory CD40 plays an essential role in autoimmune diseases, including experimental autoimmune encephalomyelitis (EAE), a murine model of human multiple sclerosis (MS). However, how CD40 drives autoimmune disease pathogenesis is not well defined. Here, we used a conditional knockout approach to determine how CD40 orchestrates a CNS autoimmune disease induced by recombinant human myelin oligodendrocyte glycoprotein (rhMOG). We found that deletion of CD40 in either dendritic cells (DCs) or B cells profoundly reduced EAE disease pathogenesis. Mechanistically, CD40 expression on DCs was required for priming pathogenic Th cells in peripheral draining lymph nodes and promoting their appearance in the CNS. By contrast, B cell CD40 was essential for class-switched MOG-specific Ab production, which played a crucial role in disease pathogenesis. In fact, passive transfer of MOG-immune serum or IgG into mice lacking CD40 on B cells but not DCs reconstituted autoimmune disease, which was associated with inundation of the spinal cord parenchyma by Ig and complement. These data demonstrate that CD40 supports distinct effector programs in B cells and DCs that converge to drive a CNS autoimmune disease and identify targets for intervention.
Collapse
Affiliation(s)
- Ying Lu
- Experimental Immunology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Max Xu
- Experimental Immunology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Cayce E. Dorrier
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ray Zhang
- Experimental Immunology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christian T. Mayer
- Experimental Immunology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - David Wagner
- Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045 USA
| | - Dorian B. McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Richard J. Hodes
- Experimental Immunology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
9
|
Strohm L, Ubbens H, Münzel T, Daiber A, Daub S. Role of CD40(L)-TRAF signaling in inflammation and resolution-a double-edged sword. Front Pharmacol 2022; 13:995061. [PMID: 36267276 PMCID: PMC9577411 DOI: 10.3389/fphar.2022.995061] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 09/12/2022] [Indexed: 11/26/2022] Open
Abstract
Cardiovascular diseases (CVD) and cardiovascular risk factors are the leading cause of death in the world today. According to the Global Burden of Disease Study, hypertension together with ischemic heart and cerebrovascular diseases is responsible for approximately 40% of all deaths worldwide. The major pathomechanism underlying almost all CVD is atherosclerosis, an inflammatory disorder of the vascular system. Recent large-scale clinical trials demonstrated that inflammation itself is an independent cardiovascular risk factor. Specific anti-inflammatory therapy could decrease cardiovascular mortality in patients with atherosclerosis (increased markers of inflammation). Inflammation, however, can also be beneficial by conferring so-called resolution, a process that contributes to clearing damaged tissue from cell debris upon cell death and thereby represents an essential step for recovery from, e.g., ischemia/reperfusion damage. Based on these considerations, the present review highlights features of the detrimental inflammatory reactions as well as of the beneficial process of immune cell-triggered resolution. In this context, we discuss the polarization of macrophages to either M1 or M2 phenotype and critically assess the role of the CD40L-CD40-TRAF signaling cascade in atherosclerosis and its potential link to resolution. As CD40L can bind to different cellular receptors, it can initiate a broad range of inflammatory processes that may be detrimental or beneficial. Likewise, the signaling of CD40L downstream of CD40 is mainly determined by activation of TRAF1-6 pathways that again can be detrimental or beneficial. Accordingly, CD40(L)-based therapies may be Janus-faced and require sophisticated fine-tuning in order to promote cardioprotection.
Collapse
Affiliation(s)
- Lea Strohm
- Department of Cardiology, Cardiology I—Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Henning Ubbens
- Department of Cardiology, Cardiology I—Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Thomas Münzel
- Department of Cardiology, Cardiology I—Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology, Cardiology I—Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Steffen Daub
- Department of Cardiology, Cardiology I—Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
10
|
Li T, Li Y, Li JW, Qin YH, Zhai H, Feng B, Li H, Zhang NN, Yang CS. Expression of TRAF6 in peripheral blood B cells of patients with myasthenia gravis. BMC Neurol 2022; 22:302. [PMID: 35978310 PMCID: PMC9382794 DOI: 10.1186/s12883-022-02833-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
Background Tumor necrosis factor receptor-associated factor 6 (TRAF6) can regulate the activation of inflammatory signaling pathways by acting as an E3 ubiquitin ligase, which enhances B cell activation. This study aimed to evaluate the expression of TRAF6 in the peripheral blood B cells of myasthenia gravis (MG) patients and analyze the relationships between TRAF6 expression and clinical characteristics. Method In our study, the expression level of TRAF6 in peripheral blood B cells of 89 patients was measured by flow cytometry compared with that of healthy subjects. The effects of disease severity, MG classification and immunotherapy on TRAF6 expression level were also analyzed. Results In our study, TRAF6 expression was elevated in CD19+ B cells and CD19+CD27+ memory B cells in generalized MG (GMG) patients compared with ocular MG (OMG) patients (p = 0.03 and p = 0.03, respectively). There was a significant positive correlation between the TRAF6 expression level and disease severity in both OMG patients and GMG patients (CD19+ B cells: OMG: p < 0.001, r = 0.89; GMG: p = 0.001, r = 0.59; CD29+CD27+ B cells: OMG: p = 0.001, r = 0.80; GMG: p = 0.048, r = 0.38). TRAF6 expression was significantly elevated in CD19+ B cells and CD19+CD27+ memory B cells in GMG with acute aggravation compared with GMG in MMS (p = 0.009 and p = 0.028, respectively). In the eleven MG patients who were followed, TRAF6 expression in B cells and memory B cells was significantly decreased after treatment (p = 0.03 and p < 0.01, respectively). Conclusion TRAF6 is potentially a useful biomarker of inflammation in patients with MG, and might be used to evaluate the effectiveness of treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12883-022-02833-9.
Collapse
Affiliation(s)
- Ting Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China.
| | - Yue Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Jia-Wen Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Ying-Hui Qin
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Hui Zhai
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Bin Feng
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - He Li
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
| | - Ning-Nannan Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, School of Medical Imaging, Tianjin Medical University General Hospital, Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Chun-Sheng Yang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China.
| |
Collapse
|
11
|
Rabatscher PA, Trendelenburg M. Anti-C1q autoantibodies from systemic lupus erythematosus patients enhance CD40-CD154-mediated inflammation in peripheral blood mononuclear cells in vitro. Clin Transl Immunology 2022; 11:e1408. [PMID: 35928801 PMCID: PMC9345742 DOI: 10.1002/cti2.1408] [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] [Received: 03/06/2022] [Revised: 05/09/2022] [Accepted: 07/14/2022] [Indexed: 01/03/2023] Open
Abstract
Objectives Systemic lupus erythematosus (SLE) is a clinically heterogeneous autoimmune disease with complex pathogenic mechanisms. Complement C1q has been shown to play a major role in SLE, and autoantibodies against C1q (anti‐C1q) are strongly associated with SLE disease activity and severe lupus nephritis suggesting a pathogenic role for anti‐C1q. Whereas C1q alone has anti‐inflammatory effects on human monocytes and macrophages, C1q/anti‐C1q complexes favor a pro‐inflammatory phenotype. This study aimed to elucidate the inflammatory effects of anti‐C1q on peripheral blood mononuclear cells (PBMCs). Methods Isolated monocytes, isolated T cells and bulk PBMCs of healthy donors with or without concomitant T cell activation were exposed to C1q or complexes of C1q and SLE patient‐derived anti‐C1q (C1q/anti‐C1q). Functional consequences of C1q/anti‐C1q on cells were assessed by determining cytokine secretion, monocyte surface marker expression, T cell activation and proliferation. Results Exposure of isolated T cells to C1q or C1q/anti‐C1q did not affect their activation and proliferation. However, unspecific T cell activation in PBMCs in the presence of C1q/anti‐C1q resulted in increased TNF, IFN‐γ and IL‐10 secretion compared with C1q alone. Co‐culture and inhibition experiments showed that the inflammatory effect of C1q/anti‐C1q on PBMCs was due to a direct CD40–CD154 interaction between activated T cells and C1q/anti‐C1q‐primed monocytes. The CD40‐mediated inflammatory reaction of monocytes involves TRAF6 and JAK3‐STAT5 signalling. Conclusion In conclusion, C1q/anti‐C1q have a pro‐inflammatory effect on monocytes that depends on T cell activation and CD40–CD154 signalling. This signalling pathway could serve as a therapeutic target for anti‐C1q‐mediated inflammation.
Collapse
Affiliation(s)
| | - Marten Trendelenburg
- Laboratory of Clinical Immunology, Department of Biomedicine University of Basel Basel Switzerland.,Division of Internal Medicine University Hospital Basel Basel Switzerland
| |
Collapse
|
12
|
Ong GH, Ori D, Kawasaki T, Kawai T. Inhibition of lipopolysaccharide-induced inflammatory responses by 1'-acetoxychavicol acetate. Genes Cells 2022; 27:482-492. [PMID: 35467779 DOI: 10.1111/gtc.12943] [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: 03/28/2022] [Revised: 04/12/2022] [Accepted: 04/19/2022] [Indexed: 11/26/2022]
Abstract
Lipopolysaccharide on gram negative bacteria can be detected by Toll-like receptor 4 (TLR4) to elicit a series of innate immune responses, leading to inflammation to eliminate the targeted pathogen. However, dysregulation in the responses results in excessive inflammation. The 1'-acetoxychavicol acetate (ACA) is a bioactive compound originated from Alpinia species known to have anti-inflammatory and apoptosis-inducing properties. Here, we found that ACA inhibits lipopolysaccharide-induced expression and production of proinflammatory cytokines such as interleukin 6 and TNFα by macrophages. ACA suppresses the activation of NF-κB and MAP kinases in TLR4 signaling. Moreover, ACA also inhibits TLR4-mediated induction of type I interferon by suppressing IRF3 activation. In lipopolysaccharide-challenged mice, ACA treatment successfully increased the survival of mice and alleviated inflammation in the lung. Thus, ACA is a potential anti-inflammatory agent to regulate excessive inflammation. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Guang Han Ong
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), 8916-5 Takayama-cho, Ikoma, Nara, Japan
| | - Daisuke Ori
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), 8916-5 Takayama-cho, Ikoma, Nara, Japan
| | - Takumi Kawasaki
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), 8916-5 Takayama-cho, Ikoma, Nara, Japan
| | - Taro Kawai
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), 8916-5 Takayama-cho, Ikoma, Nara, Japan
| |
Collapse
|
13
|
Potjewyd FM, Axtman AD. Exploration of Aberrant E3 Ligases Implicated in Alzheimer's Disease and Development of Chemical Tools to Modulate Their Function. Front Cell Neurosci 2021; 15:768655. [PMID: 34867205 PMCID: PMC8637409 DOI: 10.3389/fncel.2021.768655] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/28/2021] [Indexed: 11/24/2022] Open
Abstract
The Ubiquitin Proteasome System (UPS) is responsible for the degradation of misfolded or aggregated proteins via a multistep ATP-dependent proteolytic mechanism. This process involves a cascade of ubiquitin (Ub) transfer steps from E1 to E2 to E3 ligase. The E3 ligase transfers Ub to a targeted protein that is brought to the proteasome for degradation. The inability of the UPS to remove misfolded or aggregated proteins due to UPS dysfunction is commonly observed in neurodegenerative diseases, such as Alzheimer's disease (AD). UPS dysfunction in AD drives disease pathology and is associated with the common hallmarks such as amyloid-β (Aβ) accumulation and tau hyperphosphorylation, among others. E3 ligases are key members of the UPS machinery and dysfunction or changes in their expression can propagate other aberrant processes that accelerate AD pathology. The upregulation or downregulation of expression or activity of E3 ligases responsible for these processes results in changes in protein levels of E3 ligase substrates, many of which represent key proteins that propagate AD. A powerful way to better characterize UPS dysfunction in AD and the role of individual E3 ligases is via the use of high-quality chemical tools that bind and modulate specific E3 ligases. Furthermore, through combining gene editing with recent advances in 3D cell culture, in vitro modeling of AD in a dish has become more relevant and possible. These cell-based models of AD allow for study of specific pathways and mechanisms as well as characterization of the role E3 ligases play in driving AD. In this review, we outline the key mechanisms of UPS dysregulation linked to E3 ligases in AD and highlight the currently available chemical modulators. We present several key approaches for E3 ligase ligand discovery being employed with respect to distinct classes of E3 ligases. Where possible, specific examples of the use of cultured neurons to delineate E3 ligase biology have been captured. Finally, utilizing the available ligands for E3 ligases in the design of proteolysis targeting chimeras (PROTACs) to degrade aberrant proteins is a novel strategy for AD, and we explore the prospects of PROTACs as AD therapeutics.
Collapse
|
14
|
Human Monocytes Plasticity in Neurodegeneration. Biomedicines 2021; 9:biomedicines9070717. [PMID: 34201693 PMCID: PMC8301413 DOI: 10.3390/biomedicines9070717] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/11/2021] [Accepted: 06/21/2021] [Indexed: 01/09/2023] Open
Abstract
Monocytes play a crucial role in immunity and tissue homeostasis. They constitute the first line of defense during the inflammatory process, playing a role in the pathogenesis and progression of diseases, making them an attractive therapeutic target. They are heterogeneous in morphology and surface marker expression, which suggest different molecular and physiological properties. Recent evidences have demonstrated their ability to enter the brain, and, as a consequence, their hypothetical role in different neurodegenerative diseases. In this review, we will discuss the current knowledge about the correlation between monocyte dysregulation in the brain and/or in the periphery and neurological diseases in humans. Here we will focus on the most common neurodegenerative disorders, such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and multiple sclerosis.
Collapse
|
15
|
Najafi S, Saadat P, Beladi Moghadam N, Manoucherinia A, Aghazadeh Z, Vali Mohammadi A, Pashaiefar H, Hosseini M, Mirshafiey A. Evaluation of the Effect of Mannuronic Acid as a Novel NSAID With Immunosuppressive Properties on Expression of SOCS1, SOCS3, SHIP1, and TRAF6 Genes and Serum Levels of IL-6 and TNF-α in Patients With Multiple Sclerosis. J Clin Pharmacol 2021; 61:1303-1310. [PMID: 33908653 DOI: 10.1002/jcph.1879] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 04/18/2021] [Indexed: 11/12/2022]
Abstract
Multiple sclerosis (MS) is described as a chronic inflammatory, demyelinating disease of the central nervous system on an autoimmune basis, which is the most frequent reason for nontraumatic disability in youth. The efficacy and safety of β-D-nannuronic acid (M2000) as a novel immunosuppressive drug (patented PCT/EP2017/067920) has been shown in an experimental model of MS and also in a phase 2 clinical trial. The effects of M2000 on SOCS1, SOCS3, TRAF6, and SHIP1 gene expression and also serum levels of IL-6 and TNF-α in secondary progressive multiple sclerosis patients have been assessed in this study. In this study, 14 secondary progressive multiple sclerosis patients and 14 healthy subjects (as the control group) were recruited from the phase 2 clinical trial (Clinical Trial identifier, IRCT2016111313739N6). Gene expression of SOCS1, SOCS3, TRAF6, and SHIP1 was measured at baseline and after 6 months of therapy with M2000 using a quantitative real-time polymerase chain reaction method. Furthermore, the serum levels of IL-6 and TNF-α were assessed by the enzyme-linked immunosorbent assay method. Our results showed that the gene expression of SOCS1, SOCS3, and SHIP1 was increased after 6 months of therapy with M2000 in MS patients. Moreover, the serum levels of IL-6 and TNF-α of patients declined compared with baseline, but this was not statistically significant. The results of this study demonstrated that M2000, with immunosuppressive properties, could upregulate SOCS1, SOCS3, and SHIP1 genes in patients with secondary progressive multiple sclerosis.
Collapse
Affiliation(s)
- Soheil Najafi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Payam Saadat
- Mobility Impairment Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Nahid Beladi Moghadam
- Department of Neurology, Imam Hossein Hospital, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Ali Manoucherinia
- Department of Clinical Neuroscience (CNS), Karolinska Institutet, Stockholm, Sweden
| | - Zahra Aghazadeh
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Hossein Pashaiefar
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mostafa Hosseini
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Mirshafiey
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Huang H, Xia A, Sun L, Lu C, Liu Y, Zhu Z, Wang S, Cai J, Zhou X, Liu S. Pathogenic Functions of Tumor Necrosis Factor Receptor- Associated Factor 6 Signaling Following Traumatic Brain Injury. Front Mol Neurosci 2021; 14:629910. [PMID: 33967693 PMCID: PMC8096983 DOI: 10.3389/fnmol.2021.629910] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/29/2021] [Indexed: 01/25/2023] Open
Abstract
Neuroinflammation contributes to delayed (secondary) neurodegeneration following traumatic brain injury (TBI). Tumor necrosis factor receptor-associated factor 6 (TRAF6) signaling may promote post-TBI neuroinflammation, thereby exacerbating secondary injury. This study investigated the pathogenic functions of TRAF6 signaling following TBI in vivo and in vitro. A rat TBI model was established by air pressure contusion while lipopolysaccharide (LPS) exposure was used to induce inflammatory-like responses in cultured astrocytes. Model rats were examined for cell-specific expression of TRAF6, NF-κB, phosphorylated (p)-NF-κB, MAPKs (ERK, JNK, and p38), p-MAPKs, chemokines (CCL2 and CXCL1), and chemokine receptors (CCR2 and CXCR2) by immunofluorescence, RT-qPCR, western blotting, and ELISA, for apoptosis by TUNEL staining, and spatial cognition by Morris water maze testing. These measurements were compared between TBI model rats receiving intracerebral injections of TRAF6-targeted RNAi vector (AAV9-TRAF6-RNAi), empty vector, MAPK/NF-κB inhibitors, or vehicle. Primary astrocytes were stimulated with LPS following TRAF6 siRNA or control transfection, and NF-κB, MAPKs, chemokine, and chemokine receptor expression levels evaluated by western blotting and ELISA. TRAF6 was expressed mainly in astrocytes and neurons of injured cortex, peaking 3 days post-TBI. Knockdown by AAV9-TRAF6-RNAi improved spatial learning and memory, decreased TUNEL-positive cell number in injured cortex, and downregulated expression levels of p-NF-κB, p-ERK, p-JNK, p-p38, CCL2, CCR2, CXCL1, and CXCR2 post-TBI. Inhibitors of NF-κB, ERK, JNK, and p38 significantly suppressed CCL2, CCR2, CXCL1, and CXCR2 expression following TBI. Furthermore, TRAF6-siRNA inhibited LPS-induced NF-κB, ERK, JNK, p38, CCL2, and CXCL1 upregulation in cultured astrocytes. Targeting TRAF6-MAPKs/NF-κB-chemokine signaling pathways may provide a novel therapeutic approach for reducing post-TBI neuroinflammation and concomitant secondary injury.
Collapse
Affiliation(s)
- Huan Huang
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, China.,School of Medicine, Nantong University, Nantong, China
| | - Anqi Xia
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, China.,School of Medicine, Nantong University, Nantong, China
| | - Li Sun
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Chun Lu
- Department of Rehabilitation Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Liu
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China
| | - Zhenjie Zhu
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Siye Wang
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Junyan Cai
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Xiaoyun Zhou
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Su Liu
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
17
|
Mechanism by which TRAF6 Participates in the Immune Regulation of Autoimmune Diseases and Cancer. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4607197. [PMID: 33294443 PMCID: PMC7714562 DOI: 10.1155/2020/4607197] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 11/06/2020] [Accepted: 11/17/2020] [Indexed: 11/24/2022]
Abstract
Tumor necrosis factor (TNF) receptor-associated factor 6 (TRAF6), an E3 ubiquitin ligase, is a signal transduction molecule shared by the interleukin-1 receptor (IL-1R)/Toll-like receptor (TLR) family and the TNFR superfamily. TRAF6 has a unique TRAF domain and RING finger domain that mediate intracellular signaling events. In the immune system, TRAF6-mediated signaling has been shown to be critical for the development, homeostasis, and activation of a variety of immune cells, including B cells, T cells, dendritic cells, and macrophages. Although the pathogenesis and etiology of autoimmune diseases and cancer are not fully understood, it is worth noting that existing studies have shown that TRAF6 is involved in the pathogenesis and development of a variety of these diseases. Herein, we reviewed the role of TRAF6 in certain immune cells, as well as the function and potential effect of TRAF6 in autoimmune diseases and cancer. Our review indicates that TRAF6 may be a novel target for autoimmune diseases and cancer.
Collapse
|
18
|
Lutgens E, Atzler D, Döring Y, Duchene J, Steffens S, Weber C. Immunotherapy for cardiovascular disease. Eur Heart J 2020; 40:3937-3946. [PMID: 31121017 DOI: 10.1093/eurheartj/ehz283] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 02/11/2019] [Accepted: 04/17/2019] [Indexed: 12/21/2022] Open
Abstract
The outcomes of the Canakinumab Anti-inflammatory Thrombosis Outcome Study (CANTOS) trial have unequivocally proven that inflammation is a key driver of atherosclerosis and that targeting inflammation, in this case by using an anti-interleukin-1β antibody, improves cardiovascular disease (CVD) outcomes. This is especially true for CVD patients with a pro-inflammatory constitution. Although CANTOS has epitomized the importance of targeting inflammation in atherosclerosis, treatment with canakinumab did not improve CVD mortality, and caused an increase in infections. Therefore, the identification of novel drug targets and development of novel therapeutics that block atherosclerosis-specific inflammatory pathways and exhibit limited immune-suppressive side effects, as pursued in our collaborative research centre, are required to optimize immunotherapy for CVD. In this review, we will highlight the potential of novel immunotherapeutic targets that are currently considered to become a future treatment for CVD.
Collapse
Affiliation(s)
- Esther Lutgens
- Institute for Cardiovascular Prevention (IPEK), CRC 1123 Atherosclerosis - Mechanisms and Networks of novel therapeutic Targets, Ludwig-Maximilians-Universität, Ludwig-Maximilians-University Munich, Pettenkoferstraße 9, Munich 80336, Germany.,Department of Medical Biochemistry, Amsterdam University Medical Centers, Location AMC, Amsterdam Cardiovascular Sciences (ACS), University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, the Netherlands.,German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Dorothee Atzler
- Institute for Cardiovascular Prevention (IPEK), CRC 1123 Atherosclerosis - Mechanisms and Networks of novel therapeutic Targets, Ludwig-Maximilians-Universität, Ludwig-Maximilians-University Munich, Pettenkoferstraße 9, Munich 80336, Germany.,Department of Medical Biochemistry, Amsterdam University Medical Centers, Location AMC, Amsterdam Cardiovascular Sciences (ACS), University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, the Netherlands.,German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany.,Walther-Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Goethestraße 33, Munich 80336, Germany
| | - Yvonne Döring
- Institute for Cardiovascular Prevention (IPEK), CRC 1123 Atherosclerosis - Mechanisms and Networks of novel therapeutic Targets, Ludwig-Maximilians-Universität, Ludwig-Maximilians-University Munich, Pettenkoferstraße 9, Munich 80336, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Johan Duchene
- Institute for Cardiovascular Prevention (IPEK), CRC 1123 Atherosclerosis - Mechanisms and Networks of novel therapeutic Targets, Ludwig-Maximilians-Universität, Ludwig-Maximilians-University Munich, Pettenkoferstraße 9, Munich 80336, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Sabine Steffens
- Institute for Cardiovascular Prevention (IPEK), CRC 1123 Atherosclerosis - Mechanisms and Networks of novel therapeutic Targets, Ludwig-Maximilians-Universität, Ludwig-Maximilians-University Munich, Pettenkoferstraße 9, Munich 80336, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), CRC 1123 Atherosclerosis - Mechanisms and Networks of novel therapeutic Targets, Ludwig-Maximilians-Universität, Ludwig-Maximilians-University Munich, Pettenkoferstraße 9, Munich 80336, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany.,Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitsingel 50, 6229 ER Maastricht, the Netherlands
| |
Collapse
|
19
|
Combined administration of a small-molecule inhibitor of TRAF6 and Docetaxel reduces breast cancer skeletal metastasis and osteolysis. Cancer Lett 2020; 488:27-39. [PMID: 32474152 DOI: 10.1016/j.canlet.2020.05.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/08/2020] [Accepted: 05/18/2020] [Indexed: 12/20/2022]
Abstract
Tumour necrosis factor receptor-associated factor 6 (TRAF6) has been implicated in breast cancer and osteoclastic bone destruction. Here, we report that 6877002, a verified small-molecule inhibitor of TRAF6, reduced metastasis, osteolysis and osteoclastogenesis in models of osteotropic human and mouse breast cancer. First, we observed that TRAF6 is highly expressed in osteotropic breast cancer cells and its level of expression was higher in patients with bone metastasis. Pre-exposure of osteoclasts and osteoblasts to non-cytotoxic concentrations of 6877002 inhibited cytokine-induced NFκB activation and osteoclastogenesis, and reduced the ability of osteotropic human MDA-MB-231 and mouse 4T1 breast cancer cells to support bone cell activity. 6877002 inhibited human MDA-MB-231-induced osteolysis in the mouse calvaria organ system, and reduced soft tissue and bone metastases in immuno-competent mice following intra-cardiac injection of mouse 4T1-Luc2 cells. Of clinical relevance, combined administration of 6877002 with Docetaxel reduced metastasis and inhibited osteolytic bone damage in mice bearing 4T1-Luc2 cells. Thus, TRAF6 inhibitors such as 6877002 - alone or in combination with conventional chemotherapy - show promise for the treatment of metastatic breast cancer.
Collapse
|
20
|
Identification of a pro-elongation effect of diallyl disulfide, a major organosulfur compound in garlic oil, on microglial process. J Nutr Biochem 2020; 78:108323. [DOI: 10.1016/j.jnutbio.2019.108323] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 10/30/2019] [Accepted: 12/06/2019] [Indexed: 12/11/2022]
|
21
|
Neuroinflammation in CNS diseases: Molecular mechanisms and the therapeutic potential of plant derived bioactive molecules. PHARMANUTRITION 2020. [DOI: 10.1016/j.phanu.2020.100176] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
22
|
Haghmorad D, Yazdanpanah E, Jadid Tavaf M, Zargarani S, Soltanmohammadi A, Mahmoudi MB, Mahmoudi M. Prevention and treatment of experimental autoimmune encephalomyelitis induced mice with 1, 25-dihydroxyvitamin D 3. Neurol Res 2019; 41:943-957. [PMID: 31402771 DOI: 10.1080/01616412.2019.1650218] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Multiple sclerosis (MS) is a complex inflammatory and demyelinating disease of the central nervous system (CNS) frequently starts in young adulthood. Demyelination, inflammatory and axonal damage in the CNS is the pathological hallmark of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. 1, 25-dihydroxyvitamin D3 (Vitamin D3) is involved in calcium regulation, phosphorus homeostasis, and bone mineralization. In addition, vitamin D3 has potential inhibitory effects on immune cells in various inflammatory and autoimmunity disease. C57BL/6 female mice were divided into prevention groups (low, middle and high doses) and treatment groups (middle and high doses). Prevention groups received vitamin D3 2 weeks before EAE induction, and treatment groups were treated with vitamin D3 simultaneous with EAE induction. Vitamin D3 inhibits the development of EAE in a dose-dependent manner. Histological studies revealed reduced demyelination and limited infiltration into CNS, moreover vitamin D3 increased the production of IL-4, IL-10, and TGF-β, while a significant reduction in the production of IFN-γ, IL-6, TNF-α, and IL-17 was observed. Flow cytometry results for CD4+ T cell subsets in compliance with ELISA cytokine assay results showed a significant decrease in the percentage of Th1 and Th17, but also a significant increase in the percentage of Th2 and Treg for middle and high dose vitamin D3 treated mice. Real-time PCR results indicated that middle and high dose vitamin D3 treatment reduced T-bet and ROR-γt expression, but enhanced GATA3 and Foxp3 expression. Real-Time PCR results in CNS for T cell subsets related cytokines and transcription factors supported the results of flow cytometry and ELISA. This study indicated that middle and high doses of vitamin D3 deviate the balance between Th1/Th2 and Th17/Treg to Th2 and Treg. Moreover, vitamin D3 could reduce the incidence and severity of EAE clinical disease.
Collapse
Affiliation(s)
- Dariush Haghmorad
- Department of Pathology and Laboratory Medicine, School of Medicine, Semnan University of Medical Sciences , Semnan , Iran.,Department of Immunology, School of Medicine, Semnan University of Medical Sciences , Semnan , Iran
| | - Esmaeil Yazdanpanah
- Immunology Research Center, Department of Immunology and Allergy, Mashhad University of Medical Sciences , Mashhad , Iran
| | - Maryam Jadid Tavaf
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences , Semnan , Iran
| | - Simin Zargarani
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences , Semnan , Iran
| | - Azita Soltanmohammadi
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences , Semnan , Iran
| | | | - Mahmoud Mahmoudi
- Immunology Research Center, Department of Immunology and Allergy, Mashhad University of Medical Sciences , Mashhad , Iran
| |
Collapse
|
23
|
Seijkens TTP, van Tiel CM, Kusters PJH, Atzler D, Soehnlein O, Zarzycka B, Aarts SABM, Lameijer M, Gijbels MJ, Beckers L, den Toom M, Slütter B, Kuiper J, Duchene J, Aslani M, Megens RTA, van 't Veer C, Kooij G, Schrijver R, Hoeksema MA, Boon L, Fay F, Tang J, Baxter S, Jongejan A, Moerland PD, Vriend G, Bleijlevens B, Fisher EA, Duivenvoorden R, Gerdes N, de Winther MPJ, Nicolaes GA, Mulder WJM, Weber C, Lutgens E. Targeting CD40-Induced TRAF6 Signaling in Macrophages Reduces Atherosclerosis. J Am Coll Cardiol 2019; 71:527-542. [PMID: 29406859 PMCID: PMC5800892 DOI: 10.1016/j.jacc.2017.11.055] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 11/02/2017] [Accepted: 11/16/2017] [Indexed: 02/05/2023]
Abstract
Background Disrupting the costimulatory CD40-CD40L dyad reduces atherosclerosis, but can result in immune suppression. The authors recently identified small molecule inhibitors that block the interaction between CD40 and tumor necrosis factor receptor-associated factor (TRAF) 6 (TRAF-STOPs), while leaving CD40-TRAF2/3/5 interactions intact, thereby preserving CD40-mediated immunity. Objectives This study evaluates the potential of TRAF-STOP treatment in atherosclerosis. Methods The effects of TRAF-STOPs on atherosclerosis were investigated in apolipoprotein E deficient (Apoe−/−) mice. Recombinant high-density lipoprotein (rHDL) nanoparticles were used to target TRAF-STOPs to macrophages. Results TRAF-STOP treatment of young Apoe−/− mice reduced atherosclerosis by reducing CD40 and integrin expression in classical monocytes, thereby hampering monocyte recruitment. When Apoe−/− mice with established atherosclerosis were treated with TRAF-STOPs, plaque progression was halted, and plaques contained an increase in collagen, developed small necrotic cores, and contained only a few immune cells. TRAF-STOP treatment did not impair “classical” immune pathways of CD40, including T-cell proliferation and costimulation, Ig isotype switching, or germinal center formation, but reduced CD40 and β2-integrin expression in inflammatory monocytes. In vitro testing and transcriptional profiling showed that TRAF-STOPs are effective in reducing macrophage migration and activation, which could be attributed to reduced phosphorylation of signaling intermediates of the canonical NF-κB pathway. To target TRAF-STOPs specifically to macrophages, TRAF-STOP 6877002 was incorporated into rHDL nanoparticles. Six weeks of rHDL-6877002 treatment attenuated the initiation of atherosclerosis in Apoe−/− mice. Conclusions TRAF-STOPs can overcome the current limitations of long-term CD40 inhibition in atherosclerosis and have the potential to become a future therapeutic for atherosclerosis.
Collapse
Affiliation(s)
- Tom T P Seijkens
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, the Netherlands; Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
| | - Claudia M van Tiel
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Pascal J H Kusters
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Dorothee Atzler
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, the Netherlands; Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany; Walther-Straub-Institut for Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich, Germany; German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Oliver Soehnlein
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany; German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Barbara Zarzycka
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Suzanne A B M Aarts
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Marnix Lameijer
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Marion J Gijbels
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, the Netherlands; Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands; Department of Molecular Genetics, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Linda Beckers
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Myrthe den Toom
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Bram Slütter
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Johan Kuiper
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Johan Duchene
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
| | - Maria Aslani
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
| | - Remco T A Megens
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany; Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Cornelis van 't Veer
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Gijs Kooij
- Department of Molecular Cell Biology and Immunology, Neuroscience Campus Amsterdam, VU Medical Center, Amsterdam, the Netherlands
| | - Roy Schrijver
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Marten A Hoeksema
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, the Netherlands
| | | | - Francois Fay
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jun Tang
- Bioceros BV, Utrecht, the Netherlands; Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Samantha Baxter
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Aldo Jongejan
- Department of Bioinformatics, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Perry D Moerland
- Department of Bioinformatics, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Gert Vriend
- Centre for Molecular and Biomolecular Informatics (CMBI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Boris Bleijlevens
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Edward A Fisher
- Division of Cardiology, Department of Medicine, Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York, New York
| | - Raphael Duivenvoorden
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, the Netherlands
| | - Norbert Gerdes
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany; Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Menno P J de Winther
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, the Netherlands; Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
| | - Gerry A Nicolaes
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Willem J M Mulder
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, the Netherlands; Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany; German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, the Netherlands; Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany.
| |
Collapse
|
24
|
Kusters PJH, Lutgens E, Seijkens TTP. Exploring immune checkpoints as potential therapeutic targets in atherosclerosis. Cardiovasc Res 2019; 114:368-377. [PMID: 29309533 DOI: 10.1093/cvr/cvx248] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 12/21/2017] [Indexed: 12/20/2022] Open
Abstract
In the past decades, the inflammatory nature of atherosclerosis has been well-recognized and despite the development of therapeutic strategies targeted at its classical risk factors such as dyslipidemia and hypertension, atherosclerosis remains a major cause of morbidity and mortality. Additional strategies targeting the chronic inflammatory pathways underlying the development of atherosclerosis are therefore required. Interactions between different immune cells result in the secretion of inflammatory mediators, such as cytokines and chemokines, and fuel atherogenesis. Immune checkpoint proteins have a critical role in facilitating immune cell interactions and play an essential role in the development of atherosclerosis. Although the therapeutic potential of these molecules is well-recognized in clinical oncology, the use of immune checkpoint modulators in atherosclerosis is still limited to experimental models. Here, we review recent insights on the role of immune checkpoint proteins in atherosclerosis. Additionally, we explore the therapeutic potential and challenges of immune checkpoint modulating strategies in cardiovascular medicine and we discuss novel therapeutic approaches to target these proteins in atherosclerosis.
Collapse
Affiliation(s)
- Pascal J H Kusters
- Department of Medical Biochemistry, Experimental Vascular Biology, Academic Medical Center (AMC), University of Amsterdam, Meibergdreef 15, 1105 CZ Amsterdam, The Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Experimental Vascular Biology, Academic Medical Center (AMC), University of Amsterdam, Meibergdreef 15, 1105 CZ Amsterdam, The Netherlands.,Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian's University (LMU), Pettenkoferstraße 8a, 80336 Munich, Germany
| | - Tom T P Seijkens
- Department of Medical Biochemistry, Experimental Vascular Biology, Academic Medical Center (AMC), University of Amsterdam, Meibergdreef 15, 1105 CZ Amsterdam, The Netherlands.,Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian's University (LMU), Pettenkoferstraße 8a, 80336 Munich, Germany
| |
Collapse
|
25
|
Arditi M, Shah PK. STOP the TRAFfic and Reduce the Plaque. J Am Coll Cardiol 2019; 71:543-546. [PMID: 29406860 DOI: 10.1016/j.jacc.2017.12.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 12/19/2017] [Indexed: 10/18/2022]
Affiliation(s)
- Moshe Arditi
- Department of Biomedical Sciences, Division of Immunology, Infectious and Immunologic Diseases Research Center (IIDRC), Cedars-Sinai Medical Center, Los Angeles, California.
| | - Prediman Krishan Shah
- Department of Medicine, Division of Cardiology, Oppenheimer Atherosclerosis Research Center, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|
26
|
Aarts SABM, Seijkens TTP, Kusters PJH, van Tiel CM, Reiche ME, den Toom M, Beckers L, van Roomen CPAA, de Winther MPJ, Kooij G, Lutgens E. Macrophage CD40 signaling drives experimental autoimmune encephalomyelitis. J Pathol 2019; 247:471-480. [PMID: 30471110 PMCID: PMC6519352 DOI: 10.1002/path.5205] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 10/23/2018] [Accepted: 11/19/2018] [Indexed: 01/06/2023]
Abstract
The costimulatory CD40L-CD40 dyad plays a major role in multiple sclerosis (MS). CD40 is highly expressed on MHCII+ B cells, dendritic cells and macrophages in human MS lesions. Here we investigated the role of the CD40 downstream signaling intermediates TNF receptor-associated factor 2 (TRAF2) and TRAF6 in MHCII+ cells in experimental autoimmune encephalomyelitis (EAE). Both MHCII-CD40-Traf2-/- and MHCII-CD40-Traf6-/- mice showed a reduction in clinical signs of EAE and prevented demyelination. However, only MHCII-CD40-Traf6-/- mice displayed a decrease in myeloid and lymphoid cell infiltration into the CNS that was accompanied by reduced levels of TNF-α, IL-6 and IFN-γ. As CD40-TRAF6 interactions predominantly occur in macrophages, we subjected CD40flfl LysMcre mice to EAE. This myeloid-specific deletion of CD40 resulted in a significant reduction in EAE severity, reduced CNS inflammation and demyelination. In conclusion, the CD40-TRAF6 signaling pathway in MHCII+ cells plays a key role in neuroinflammation and demyelination during EAE. Concomitant with the fact that CD40-TRAF6 interactions are predominant in macrophages, depletion of myeloid CD40 also reduces neuroinflammation. CD40-TRAF6 interactions thus represent a promising therapeutic target for MS. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Suzanne ABM Aarts
- Department of Medical Biochemistry, Subdivision of Experimental Vascular BiologyAmsterdam University Medical Centers, Amsterdam Cardiovascular Sciences (ACS), University of AmsterdamArizonaAmsterdamThe Netherlands
| | - Tom TP Seijkens
- Department of Medical Biochemistry, Subdivision of Experimental Vascular BiologyAmsterdam University Medical Centers, Amsterdam Cardiovascular Sciences (ACS), University of AmsterdamArizonaAmsterdamThe Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University (LMU)MunichGermany
| | - Pascal JH Kusters
- Department of Medical Biochemistry, Subdivision of Experimental Vascular BiologyAmsterdam University Medical Centers, Amsterdam Cardiovascular Sciences (ACS), University of AmsterdamArizonaAmsterdamThe Netherlands
| | - Claudia M van Tiel
- Department of Medical Biochemistry, Subdivision of Experimental Vascular BiologyAmsterdam University Medical Centers, Amsterdam Cardiovascular Sciences (ACS), University of AmsterdamArizonaAmsterdamThe Netherlands
| | - Myrthe E Reiche
- Department of Medical Biochemistry, Subdivision of Experimental Vascular BiologyAmsterdam University Medical Centers, Amsterdam Cardiovascular Sciences (ACS), University of AmsterdamArizonaAmsterdamThe Netherlands
| | - Myrthe den Toom
- Department of Medical Biochemistry, Subdivision of Experimental Vascular BiologyAmsterdam University Medical Centers, Amsterdam Cardiovascular Sciences (ACS), University of AmsterdamArizonaAmsterdamThe Netherlands
| | - Linda Beckers
- Department of Medical Biochemistry, Subdivision of Experimental Vascular BiologyAmsterdam University Medical Centers, Amsterdam Cardiovascular Sciences (ACS), University of AmsterdamArizonaAmsterdamThe Netherlands
| | - Cindy PAA van Roomen
- Department of Medical Biochemistry, Subdivision of Experimental Vascular BiologyAmsterdam University Medical Centers, Amsterdam Cardiovascular Sciences (ACS), University of AmsterdamArizonaAmsterdamThe Netherlands
| | - Menno PJ de Winther
- Department of Medical Biochemistry, Subdivision of Experimental Vascular BiologyAmsterdam University Medical Centers, Amsterdam Cardiovascular Sciences (ACS), University of AmsterdamArizonaAmsterdamThe Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University (LMU)MunichGermany
- Department of Molecular Cell Biology and ImmunologyAmsterdam University Medical Centers, MS Center Amsterdam, Amsterdam NeuroscienceAmsterdamThe Netherlands
| | - Gijs Kooij
- Department of Molecular Cell Biology and ImmunologyAmsterdam University Medical Centers, MS Center Amsterdam, Amsterdam NeuroscienceAmsterdamThe Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Subdivision of Experimental Vascular BiologyAmsterdam University Medical Centers, Amsterdam Cardiovascular Sciences (ACS), University of AmsterdamArizonaAmsterdamThe Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University (LMU)MunichGermany
| |
Collapse
|
27
|
Li M, Zhang D, Ge X, Zhu X, Zhou Y, Zhang Y, Peng X, Shen A. TRAF6-p38/JNK-ATF2 axis promotes microglial inflammatory activation. Exp Cell Res 2019; 376:133-148. [PMID: 30763583 DOI: 10.1016/j.yexcr.2019.02.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 02/02/2019] [Accepted: 02/09/2019] [Indexed: 12/18/2022]
Abstract
Activating transcription factor 2 (ATF2), a member of the alkaline-leucine zipper family, is widely expressed in various tissues, and reportedly involved in inflammatory responses to various irritates, but its role in the central nervous system (CNS) remains unclear. This study aimed to investigate the expression and biological function of ATF2 in CNS inflammation. Utilizing the LPS-induced neuroinflammation model on mice, we first found ATF2 up-regulation and its co-localization with microglia in inflamed mice brain. In vitro, we revealed an increased expression, phosphorylation, and nuclear accumulation of ATF2 in LPS-treated BV2 microglia cells. Inhibiting ATF2 significantly decreased the expression of pro-inflammatory factors in LPS-treated microglia, and alleviated neuronal apoptosis induced by the conditioned medium of activated microglia. Knocking down TRAF6, an important adaptor of the TLR4/MAPK/NF-κB signaling pathway, suppressed the LPS-induced ATF2 expression and phosphorylation, accompanied by the decreased p38/JNK phosphorylation, in microglia. Blocking p38 or JNK signaling pathway by the specific inhibitors reversed the TRAF6-overexpression mediated ATF2 activation. Taken together, our data first proved the pro-inflammatory function of ATF2 in microglia, and suggested that the TRAF6-JNK/p38-ATF2 axis might promote microglial inflammatory activation and thus aggravate neuronal injury in brain, which might become a potential therapeutic target for CNS diseases.
Collapse
Affiliation(s)
- Mengmeng Li
- Clinical Medicine Research Center, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China; Jiangsu Key Laboratory of Neurogeneration, Nantong University, Nantong 226001, People's Republic of China
| | - Dongmei Zhang
- Clinical Medicine Research Center, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China
| | - Xin Ge
- Clinical Medicine Research Center, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China; Jiangsu Key Laboratory of Neurogeneration, Nantong University, Nantong 226001, People's Republic of China
| | - Xiangyang Zhu
- Neurology Department, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China
| | - Yong Zhou
- Neurology Department, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China
| | - Yi Zhang
- Neurosurgery Department, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China
| | - Xiao Peng
- Clinical Medicine Research Center, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China
| | - Aiguo Shen
- Clinical Medicine Research Center, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China; Jiangsu Key Laboratory of Neurogeneration, Nantong University, Nantong 226001, People's Republic of China; Cancer Research Center of Nantong, Tumor Hospital Affiliated to Nantong University, Nantong 226361, People's Republic of China.
| |
Collapse
|
28
|
Abstract
Innate and adaptive immune effector mechanisms, in conjunction with hyperlipidemia, are important drivers of atherosclerosis. The interaction between the different immune cells and the secretion of cytokines and chemokines determine the progression of atherosclerosis. The activation or dampening of the immune response is tightly controlled by immune checkpoints. Costimulatory and coinhibitory immune checkpoints represent potential targets for immune modulatory therapies for atherosclerosis. This review will discuss the current knowledge on immune checkpoints in atherosclerosis and the clinical potential of immune checkpoint targeted therapy for atherosclerosis.
Collapse
Affiliation(s)
- Ellen Rouwet
- From the Department of Surgery and Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands (E.R.)
| | - Esther Lutgens
- Department of Medical Biochemistry, Experimental Vascular Biology Laboratory, Academic Medical Center, Amsterdam, The Netherlands (E.L.)
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian's University (LMU), Munich, Germany (E.L.)
| |
Collapse
|
29
|
Abdullah M, Berthiaume JM, Willis MS. Tumor necrosis factor receptor-associated factor 6 as a nuclear factor kappa B-modulating therapeutic target in cardiovascular diseases: at the heart of it all. Transl Res 2018; 195:48-61. [PMID: 29175266 PMCID: PMC5898986 DOI: 10.1016/j.trsl.2017.10.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 10/17/2017] [Accepted: 10/30/2017] [Indexed: 02/06/2023]
Abstract
Inflammatory and immune signaling has been documented as a root cause of many cardiovascular pathologies. In this review, we explore the emerging role of tumor necrosis factor receptor-associated factor 6 (TRAF6)-nuclear factor kappa B (NF-κB) signaling axis in atherosclerosis, ischemic heart disease, pathologic cardiac hypertrophy or heart failure, myocarditis, and sepsis-induced cardiomyopathy. We discuss the current understanding of cardiac inflammation in heart disease, present the TRAF6 signaling axis in the heart, then summarize what is known about TRAF6 in pathophysiology of heart disease including proof-of-concept studies that identify the utility of blocking TRAF6 to attenuate cardiac dysfunction, which suggests that TRAF6 is a novel, druggable target in treating cardiovascular disease incurred by inflammatory processes.
Collapse
Affiliation(s)
- Muhammad Abdullah
- Department of Biochemistry, QuaidiAzam University, Islamabad, Pakistan; Department of Pathology & Laboratory Medicine, University of North Carolina, Chapel Hill, NC
| | - Jessica M Berthiaume
- Department of Physiology & Biophysics, Case Western Reserve University, Cleveland, Ohio
| | - Monte S Willis
- Department of Pathology & Laboratory Medicine, University of North Carolina, Chapel Hill, NC; Department of Pharmacology, University of North Carolina, Chapel Hill, NC.
| |
Collapse
|
30
|
Bojadzic D, Buchwald P. Toward Small-Molecule Inhibition of Protein-Protein Interactions: General Aspects and Recent Progress in Targeting Costimulatory and Coinhibitory (Immune Checkpoint) Interactions. Curr Top Med Chem 2018; 18:674-699. [PMID: 29848279 PMCID: PMC6067980 DOI: 10.2174/1568026618666180531092503] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 02/27/2018] [Accepted: 05/11/2018] [Indexed: 02/06/2023]
Abstract
Protein-Protein Interactions (PPIs) that are part of the costimulatory and coinhibitory (immune checkpoint) signaling are critical for adequate T cell response and are important therapeutic targets for immunomodulation. Biologics targeting them have already achieved considerable clinical success in the treatment of autoimmune diseases or transplant recipients (e.g., abatacept, belatacept, and belimumab) as well as cancer (e.g., ipilimumab, nivolumab, pembrolizumab, atezolizumab, durvalumab, and avelumab). In view of such progress, there have been only relatively limited efforts toward developing small-molecule PPI inhibitors (SMPPIIs) targeting these cosignaling interactions, possibly because they, as all other PPIs, are difficult to target by small molecules and were not considered druggable. Nevertheless, substantial progress has been achieved during the last decade. SMPPIIs proving the feasibility of such approaches have been identified through various strategies for a number of cosignaling interactions including CD40-CD40L, OX40-OX40L, BAFFR-BAFF, CD80-CD28, and PD-1-PD-L1s. Here, after an overview of the general aspects and challenges of SMPPII-focused drug discovery, we review them briefly together with relevant structural, immune-signaling, physicochemical, and medicinal chemistry aspects. While so far only a few of these SMPPIIs have shown activity in animal models (DRI-C21045 for CD40-D40L, KR33426 for BAFFR-BAFF) or reached clinical development (RhuDex for CD80-CD28, CA-170 for PD-1-PD-L1), there is proof-of-principle evidence for the feasibility of such approaches in immunomodulation. They can result in products that are easier to develop/ manufacture and are less likely to be immunogenic or encounter postmarket safety events than corresponding biologics, and, contrary to them, can even become orally bioavailable.
Collapse
Affiliation(s)
- Damir Bojadzic
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Peter Buchwald
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida, USA
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| |
Collapse
|
31
|
Dou Y, Tian X, Zhang J, Wang Z, Chen G. Roles of TRAF6 in Central Nervous System. Curr Neuropharmacol 2018; 16:1306-1313. [PMID: 29651950 PMCID: PMC6251041 DOI: 10.2174/1570159x16666180412094655] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 07/22/2017] [Accepted: 02/28/2018] [Indexed: 12/30/2022] Open
Abstract
Tumor necrosis factor receptor-associated factor (TRAF) is an important binding protein of tumor necrosis factor (TNF) superfamily and the toll/IL-1 receptor (TIR) superfamily, which play an important role in innate immunity and acquired immunity. TRAFs family have 7 members (TRAF1-7), and TRAF6 has its special facture and biological function. TRAF6 has two special domains: C-terminal domain and N-terminal domain, which could integrate with multiple kinases and regulate signaling pathway function as an E3 ubiquitin ligase. Studies have increasingly found that TRAF6 is closely related to central nervous system diseases, such as stroke, Traumatic brain injury, neurodegenerative diseases and neuropathic pain. Further research on the pathophysiological mechanism may be expected to become the new targets for the treatment of central nervous system diseases.
Collapse
Affiliation(s)
| | | | | | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou215006, P.R. China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou215006, P.R. China
| |
Collapse
|
32
|
Lalani AI, Zhu S, Gokhale S, Jin J, Xie P. TRAF molecules in inflammation and inflammatory diseases. ACTA ACUST UNITED AC 2017. [PMID: 29527458 DOI: 10.1007/s40495-017-0117-y] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Purpose of Review This review presents an overview of the current knowledge of TRAF molecules in inflammation with an emphasis on available human evidence and direct in vivo evidence of mouse models that demonstrate the contribution of TRAF molecules in the pathogenesis of inflammatory diseases. Recent Findings The tumor necrosis factor receptor (TNF-R)-associated factor (TRAF) family of cytoplasmic proteins was initially identified as signaling adaptors that bind directly to the intracellular domains of receptors of the TNF-R superfamily. It is now appreciated that TRAF molecules are widely employed in signaling by a variety of adaptive and innate immune receptors as well as cytokine receptors. TRAF-dependent signaling pathways typically lead to the activation of nuclear factor-κBs (NF-κBs), mitogen-activated protein kinases (MAPKs), or interferon-regulatory factors (IRFs). Most of these signaling pathways have been linked to inflammation, and therefore TRAF molecules were expected to regulate inflammation and inflammatory responses since their discovery in 1990s. However, direct in vivo evidence of TRAFs in inflammation and especially in inflammatory diseases had been lacking for many years, partly due to the difficulty imposed by early lethality of TRAF2-/-, TRAF3-/-, and TRAF6-/- mice. With the creation of conditional knockout and lineage-specific transgenic mice of different TRAF molecules, our understanding about TRAFs in inflammation and inflammatory responses has rapidly advanced during the past decade. Summary Increasing evidence indicates that TRAF molecules are versatile and indispensable regulators of inflammation and inflammatory responses and that aberrant expression or function of TRAFs contributes to the pathogenesis of inflammatory diseases.
Collapse
Affiliation(s)
- Almin I Lalani
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey 08854
- Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, New Jersey 08854
| | - Sining Zhu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey 08854
- Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, New Jersey 08854
| | - Samantha Gokhale
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey 08854
- Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, New Jersey 08854
| | - Juan Jin
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey 08854
- Department of Pharmacology, Anhui Medical University, Meishan Road 81st, Shushan District, Hefei, Anhui province, China
| | - Ping Xie
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey 08854
- Member, Rutgers Cancer Institute of New Jersey
| |
Collapse
|
33
|
Aarts SABM, Seijkens TTP, van Dorst KJF, Dijkstra CD, Kooij G, Lutgens E. The CD40-CD40L Dyad in Experimental Autoimmune Encephalomyelitis and Multiple Sclerosis. Front Immunol 2017; 8:1791. [PMID: 29312317 PMCID: PMC5732943 DOI: 10.3389/fimmu.2017.01791] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 11/29/2017] [Indexed: 12/16/2022] Open
Abstract
The CD40-CD40L dyad is an immune checkpoint regulator that promotes both innate and adaptive immune responses and has therefore an essential role in the development of inflammatory diseases, including multiple sclerosis (MS). In MS, CD40 and CD40L are expressed on immune cells present in blood and lymphoid organs, affected resident central nervous system (CNS) cells, and inflammatory cells that have infiltrated the CNS. CD40-CD40L interactions fuel the inflammatory response underlying MS, and both genetic deficiency and antibody-mediated inhibition of the CD40-CD40L dyad reduce disease severity in experimental autoimmune encephalomyelitis (EAE). Both proteins are therefore attractive therapeutic candidates to modulate aberrant inflammatory responses in MS. Here, we discuss the genetic, experimental and clinical studies on the role of CD40 and CD40L interactions in EAE and MS and we explore novel approaches to therapeutically target this dyad to combat neuroinflammatory diseases.
Collapse
Affiliation(s)
- Suzanne A. B. M. Aarts
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Tom T. P. Seijkens
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University (LMU), Munich, Germany
| | | | - Christine D. Dijkstra
- Department of Molecular Cell Biology and Immunology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, Netherlands
| | - Gijs Kooij
- Department of Molecular Cell Biology and Immunology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University (LMU), Munich, Germany
| |
Collapse
|
34
|
Interaction of curcumin and capsaicin with LPS induced TRAF6 expression in peripheral blood mononuclear cells. Med Chem Res 2017. [DOI: 10.1007/s00044-017-1940-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|