1
|
Li Y, Zhu Q, He R, Du J, Qin X, Li Y, Liang X, Wang J. The NFκB Signaling Pathway Is Involved in the Pathophysiological Process of Preeclampsia. Geburtshilfe Frauenheilkd 2024; 84:334-345. [PMID: 38618576 PMCID: PMC11006561 DOI: 10.1055/a-2273-6318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 02/20/2024] [Indexed: 04/16/2024] Open
Abstract
The high prevalence of preeclampsia (PE) is a major cause of maternal and fetal mortality and affects the long-term prognosis of both mother and baby. Termination of pregnancy is currently the only effective treatment for PE, so there is an urgent need for research into its pathogenesis and the development of new therapeutic approaches. The NFκB family of transcription factors has an essential role in inflammation and innate immunity. In this review, we summarize the role of NFκB in normal and preeclampsia pregnancies, the role of NFκB in existing treatment strategies, and potential NFκB treatment strategies.
Collapse
Affiliation(s)
- Yaxi Li
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Qinying Zhu
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Ruifen He
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Junhong Du
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Xue Qin
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Yi Li
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Xiaolei Liang
- Department of Obstetrics and Gynecology, Key Laboratory for Gynecologic Oncology Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Jing Wang
- Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
2
|
Zou J, Niu K, Lu T, Kan J, Cheng H, Xu L. The Multifunction of TRIM26: From Immune Regulation to Oncology. Protein Pept Lett 2024; 31:424-436. [PMID: 38956921 PMCID: PMC11475100 DOI: 10.2174/0109298665311516240621114519] [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: 02/20/2024] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 07/04/2024]
Abstract
Ubiquitination, a crucial post-translational modification, plays a role in nearly all physiological processes. Its functional execution depends on a series of catalytic reactions involving numerous proteases. TRIM26, a protein belonging to the TRIM family, exhibits E3 ubiquitin ligase activity because of its RING structural domain, and is present in diverse cell lineages. Over the last few decades, TRIM26 has been documented to engage in numerous physiological and pathological processes as a controller, demonstrating a diverse array of biological roles. Despite the growing research interest in TRIM26, there has been limited attention given to examining the protein's structure and function in existing reviews. This review begins with a concise overview of the composition and positioning of TRIM26 and then proceeds to examine its roles in immune response, viral invasion, and inflammatory processes. Simultaneously, we demonstrate the contribution of TRIM26 to the progression of various diseases, encompassing numerous malignancies and neurologic conditions. Finally, we have investigated the potential areas for future research on TRIM26.
Collapse
Affiliation(s)
- Jialai Zou
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| | - Kaiyi Niu
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| | - Tao Lu
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| | - Jianxun Kan
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| | - Hao Cheng
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| | - Lijian Xu
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| |
Collapse
|
3
|
Wan Y, Jiang J, Chen M, Han X, Zhong L, Xiao F, Liu J, Liu J, Li H, Huang H, Hou J. Unravelling the imbalanced Th17-like cell differentiation by single-cell RNA sequencing in multiple myeloma. Int Immunopharmacol 2023; 124:110852. [PMID: 37657245 DOI: 10.1016/j.intimp.2023.110852] [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: 06/07/2023] [Revised: 08/08/2023] [Accepted: 08/21/2023] [Indexed: 09/03/2023]
Abstract
Multiple myeloma (MM) is a bone marrow resident hematological malignancy. T helper (Th) cells play an essential role in maladjustment of immune function and promotion of myeloma cell proliferation and survival, which has not been fully elucidated. Here, we compared transcriptome profiles of CD4+ T cells in bone marrow samples of 3 healthy individuals and 10 MM patients before and after treatment using single-cell RNA sequencing. CD4+ T cells were divided into 7 clusters. Imbalanced Th17-like cell differentiation was indicated in MM based on bioinformation analyses, which involved IL2-STAT5 pathways and transcription factors NKFB1, RELA, STAT3, and GTF2A2. Pseudotime trajectory analysis of CD4+ T cell clusters further uncovered the enhanced transition of Th17-like to regulatory T (Treg) cells in MM, which was featured by expression changes of PLAC8, NKFB1, RELA, STAT3, and STAT1 along with the developmental path. Reduced cell-cell interaction between MM cells and CD4+ naïve/recently activated naïve T cells via CD74-APP might lead to imbalanced Th17-like cell differentiation. Checkpoints via TIGIT-NECTIN3 and LGALS9-CD47 in Treg and MM cells were also identified. Our study reveals imbalanced differentiation pattern of Th17-like cells and the immunosuppressive profiles in connection with MM cells, which might help to shed light on CD4+ T cell function in MM.
Collapse
Affiliation(s)
- Yike Wan
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Jinxing Jiang
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Mengping Chen
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Xiaofeng Han
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Lu Zhong
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Fei Xiao
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Jia Liu
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Junling Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hua Li
- Bio-ID Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Honghui Huang
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| | - Jian Hou
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| |
Collapse
|
4
|
Shan F, Cillo AR, Cardello C, Yuan DY, Kunning SR, Cui J, Lampenfeld C, Williams AM, McDonough AP, Pennathur A, Luketich JD, Kirkwood JM, Ferris RL, Bruno TC, Workman CJ, Benos PV, Vignali DAA. Integrated BATF transcriptional network regulates suppressive intratumoral regulatory T cells. Sci Immunol 2023; 8:eadf6717. [PMID: 37713508 PMCID: PMC11045170 DOI: 10.1126/sciimmunol.adf6717] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 08/21/2023] [Indexed: 09/17/2023]
Abstract
Human regulatory T cells (Tregs) are crucial regulators of tissue repair, autoimmune diseases, and cancer. However, it is challenging to inhibit the suppressive function of Tregs for cancer therapy without affecting immune homeostasis. Identifying pathways that may distinguish tumor-restricted Tregs is important, yet the transcriptional programs that control intratumoral Treg gene expression, and that are distinct from Tregs in healthy tissues, remain largely unknown. We profiled single-cell transcriptomes of CD4+ T cells in tumors and peripheral blood from patients with head and neck squamous cell carcinomas (HNSCC) and those in nontumor tonsil tissues and peripheral blood from healthy donors. We identified a subpopulation of activated Tregs expressing multiple tumor necrosis factor receptor (TNFR) genes (TNFR+ Tregs) that is highly enriched in the tumor microenvironment (TME) compared with nontumor tissue and the periphery. TNFR+ Tregs are associated with worse prognosis in HNSCC and across multiple solid tumor types. Mechanistically, the transcription factor BATF is a central component of a gene regulatory network that governs key aspects of TNFR+ Tregs. CRISPR-Cas9-mediated BATF knockout in human activated Tregs in conjunction with bulk RNA sequencing, immunophenotyping, and in vitro functional assays corroborated the central role of BATF in limiting excessive activation and promoting the survival of human activated Tregs. Last, we identified a suite of surface molecules reflective of the BATF-driven transcriptional network on intratumoral Tregs in patients with HNSCC. These findings uncover a primary transcriptional regulator of highly suppressive intratumoral Tregs, highlighting potential opportunities for therapeutic intervention in cancer without affecting immune homeostasis.
Collapse
Affiliation(s)
- Feng Shan
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Integrative Systems Biology Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Anthony R. Cillo
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Carly Cardello
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Daniel Y. Yuan
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sheryl R. Kunning
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Jian Cui
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Caleb Lampenfeld
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Asia M. Williams
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Alexandra P. McDonough
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Arjun Pennathur
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - James D. Luketich
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - John M. Kirkwood
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Robert L. Ferris
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Tullia C. Bruno
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Creg J. Workman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Panayiotis V. Benos
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Epidemiology, University of Florida, Gainesville, FL, USA
| | - Dario A. A. Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| |
Collapse
|
5
|
Pudjihartono N, Ho D, Golovina E, Fadason T, Kempa-Liehr AW, O'Sullivan JM. Juvenile idiopathic arthritis-associated genetic loci exhibit spatially constrained gene regulatory effects across multiple tissues and immune cell types. J Autoimmun 2023; 138:103046. [PMID: 37229810 DOI: 10.1016/j.jaut.2023.103046] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/04/2023] [Accepted: 04/15/2023] [Indexed: 05/27/2023]
Abstract
Juvenile idiopathic arthritis (JIA) is an autoimmune, inflammatory joint disease with complex genetic etiology. Previous GWAS have found many genetic loci associated with JIA. However, the biological mechanism behind JIA remains unknown mainly because most risk loci are located in non-coding genetic regions. Interestingly, increasing evidence has found that regulatory elements in the non-coding regions can regulate the expression of distant target genes through spatial (physical) interactions. Here, we used information on the 3D genome organization (Hi-C data) to identify target genes that physically interact with SNPs within JIA risk loci. Subsequent analysis of these SNP-gene pairs using data from tissue and immune cell type-specific expression quantitative trait loci (eQTL) databases allowed the identification of risk loci that regulate the expression of their target genes. In total, we identified 59 JIA-risk loci that regulate the expression of 210 target genes across diverse tissues and immune cell types. Functional annotation of spatial eQTLs within JIA risk loci identified significant overlap with gene regulatory elements (i.e., enhancers and transcription factor binding sites). We found target genes involved in immune-related pathways such as antigen processing and presentation (e.g., ERAP2, HLA class I and II), the release of pro-inflammatory cytokines (e.g., LTBR, TYK2), proliferation and differentiation of specific immune cell types (e.g., AURKA in Th17 cells), and genes involved in physiological mechanisms related to pathological joint inflammation (e.g., LRG1 in arteries). Notably, many of the tissues where JIA-risk loci act as spatial eQTLs are not classically considered central to JIA pathology. Overall, our findings highlight the potential tissue and immune cell type-specific regulatory changes contributing to JIA pathogenesis. Future integration of our data with clinical studies can contribute to the development of improved JIA therapy.
Collapse
Affiliation(s)
- N Pudjihartono
- The Liggins Institute, The University of Auckland, Auckland, New Zealand.
| | - D Ho
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - E Golovina
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - T Fadason
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - A W Kempa-Liehr
- Department of Engineering Science, The University of Auckland, Auckland, New Zealand
| | - J M O'Sullivan
- The Liggins Institute, The University of Auckland, Auckland, New Zealand; The Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand; MRC Lifecourse Epidemiology Unit, University of Southampton, United Kingdom; Australian Parkinsons Mission, Garvan Institute of Medical Research, Sydney, New South Wales, 384 Victoria Street, Darlinghurst, NSW, 2010, Australia; A*STAR Singapore Institute for Clinical Sciences, Singapore, Singapore.
| |
Collapse
|
6
|
Bariana M, Zhang B, Sun J, Wang W, Wang J, Cassella E, Myint F, Anuncio SA, Ouk S, Liou HC, Tan M, Wang H, Zakrzewski JL. Targeted Lymphoma Therapy Using a Gold Nanoframework-Based Drug Delivery System. ACS APPLIED MATERIALS & INTERFACES 2023; 15:6312-6325. [PMID: 36701696 PMCID: PMC9911369 DOI: 10.1021/acsami.2c17214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Precision nanomedicine can be employed as an alternative to chemo- or radiotherapy to overcome challenges associated with the often narrow therapeutic window of traditional treatment approaches, while safely inducing effective, targeted antitumor responses. Herein, we report the formulation of a therapeutic nanocomposite comprising a hyaluronic acid (HA)-coated gold nanoframework (AuNF) delivery system and encapsulated IT848, a small molecule with potent antilymphoma and -myeloma properties that targets the transcriptional activity of nuclear factor kappa B (NF-κB). The porous AuNFs fabricated via a liposome-templated approach were loaded with IT848 and surface-functionalized with HA to formulate the nanotherapeutics that were able to efficiently deliver the payload with high specificity to myeloma and lymphoma cell lines in vitro. In vivo studies characterized biodistribution, pharmacokinetics, and safety of HA-AuNFs, and we demonstrated superior efficacy of HA-AuNF-formulated IT848 vs free IT848 in lymphoma mouse models. Both in vitro and in vivo results affirm that the AuNF system can be adopted for targeted cancer therapy, improving the drug safety profile, and enhancing its efficacy with minimal dosing. HA-AuNF-formulated IT848 therefore has strong potential for clinical translation.
Collapse
Affiliation(s)
- Manpreet Bariana
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA
| | - Beilu Zhang
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Jingyu Sun
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Weiwei Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07303, USA
| | - Jinping Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07303, USA
| | - Elena Cassella
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA
| | - Faith Myint
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA
| | - Shaina A. Anuncio
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA
| | - Samedy Ouk
- ImmuneTarget Inc., San Diego, CA 92121, USA
| | | | - Ming Tan
- Department of Biostatistics, Bioinformatics and Biomathematics, Georgetown University, Washington, DC 20057, USA
| | - Hongjun Wang
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, NJ 07030, USA
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07303, USA
- Center for Healthcare Innovation, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Johannes L. Zakrzewski
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA
- Department of Oncology, Georgetown University, Washington, DC 20057, USA
- Department of Pediatrics, Hackensack University Medical Center, Hackensack, NJ 07601, USA
| |
Collapse
|
7
|
Zhao W, He X, Liu R, Ruan Q. Accelerating corneal wound healing using exosome-mediated targeting of NF-κB c-Rel. Inflamm Regen 2023; 43:6. [PMID: 36703231 PMCID: PMC9881367 DOI: 10.1186/s41232-023-00260-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 01/18/2023] [Indexed: 01/27/2023] Open
Abstract
The integrity of the corneal epithelium is essential for the maintenance of the physiological function of the cornea. Studies have found that inflammation greatly delays corneal wound healing. NF-κB c-Rel is preferentially expressed by immune cells and promotes the expression of inflammatory cytokines. In the current study, we sought to investigate whether c-Rel could be used as a potential therapeutic target for treating a corneal injury. Our studies reveal that expressions of c-Rel and its inflammatory targets are significantly increased in the cornea of mice with corneal injury. In addition, we find that c-Rel-deficient mice exhibit accelerated corneal wound healing and reduced expression of inflammatory cytokines. Further studies show that topical treatment on the corneal surface using nano-polymers or exosomes loaded with c-Rel-specific siRNA (siRel) can effectively accelerate regular and diabetic corneal wound healing. More importantly, we find that exosomes, as carriers of siRel, showed better efficacy than nano-polymers in treating corneal injury. We further demonstrate that exosomes secreted by mesenchymal stem cells can efficiently transfer siRNA into macrophages and dendritic cells but not T cells. Taken together, these results indicate that blocking c-Rel may represent an attracting strategy for the treatment of both regular and diabetic corneal injury.
Collapse
Affiliation(s)
- Wenbo Zhao
- grid.410587.fShandong First Medical University (Shandong Academy of Medical Sciences), Jinan, 250000 China ,grid.410638.80000 0000 8910 6733Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071 China
| | - Xiaozhen He
- grid.410638.80000 0000 8910 6733Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071 China ,grid.490473.dEye Institute of Shandong First Medical University, Eye Hospital of Shandong First Medical University (Shandong Eye Hospital), Jinan, 250021 China
| | - Ruiling Liu
- grid.410638.80000 0000 8910 6733Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071 China
| | - Qingguo Ruan
- grid.410638.80000 0000 8910 6733Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266071 China
| |
Collapse
|
8
|
Liu Y, Shi JZ, Jiang R, Liu SF, He YY, van der Vorst EPC, Weber C, Döring Y, Yan Y. Regulatory T Cell-Related Gene Indicators in Pulmonary Hypertension. Front Pharmacol 2022; 13:908783. [PMID: 35712711 PMCID: PMC9197497 DOI: 10.3389/fphar.2022.908783] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 04/26/2022] [Indexed: 11/21/2022] Open
Abstract
Objective: Regulatory T cells (Tregs) are critical immune modulators to maintain immune homeostasis and limit pulmonary hypertension (PH). This study was aimed to identify Treg-related genes (TRGs) in PH. Methods: The gene expression profile from lungs of PH patients was retrieved from the Gene Expression Omnibus (GEO) database. The abundance of Tregs was estimated by the xCell algorithm, the correlation of which with differentially expressed genes (DEGs) was performed. DEGs with a |Pearson correlation coefficient| >0.4 were identified as TRGs. Functional annotation and the protein–protein interaction (PPI) network were analyzed. A gene signature for 25 hub TRGs (TRGscore) was generated by a single sample scoring method to determine its accuracy to distinguish PH from control subjects. TRGs were validated in datasets of transcriptional profiling of PH cohorts and in lung tissues of experimental PH mice. Results: A total of 819 DEGs were identified in lungs of 58 PAH patients compared to that of 25 control subjects of dataset GSE117261. In total, 165 of all these DEGs were correlated with the abundance of Tregs and identified as TRGs, with 90 upregulated genes and 75 downregulated genes compared to that of control subjects. The upregulated TRGs were enriched in negative regulation of multiple pathways, such as cAMP-mediated signaling and I-kappaB kinase/NF-kappaB signaling, and regulated by multiple genes encoding transcriptional factors including HIF1A. Furthermore, 25 hub genes categorized into three clusters out of 165 TRGs were derived, and we identified 27 potential drugs targeting 10 hub TRGs. The TRGscore based on 25 hub TRGs was higher in PH patients and could distinguish PH from control subjects (all AUC >0.7). Among them, 10 genes including NCF2, MNDA/Ifi211, HCK, FGR, CSF3R, AQP9, S100A8, G6PD/G6pdx, PGD, and TXNRD1 were significantly reduced in lungs of severe PH patients of dataset GSE24988 as well as in lungs of hypoxic PH mice compared to corresponding controls. Conclusion: Our finding will shed some light on the Treg-associated therapeutic targets in the progression of PH and emphasize on TRGscore as a novel indicator for PH.
Collapse
Affiliation(s)
- Yan Liu
- Department of Nuclear Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jun-Zhuo Shi
- School of Pharmacy, Henan University, Kaifeng, China.,College of Traditional Chinese Medicine, Henan University, Kaifeng, China
| | - Rong Jiang
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Shao-Fei Liu
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Yang-Yang He
- School of Pharmacy, Henan University, Kaifeng, China.,College of Traditional Chinese Medicine, Henan University, Kaifeng, China
| | - Emiel P C van der Vorst
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.,Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany.,Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany.,Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.,Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Yvonne Döring
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.,Department of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Yi Yan
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
9
|
Roberti A, Chaffey LE, Greaves DR. NF-κB Signaling and Inflammation-Drug Repurposing to Treat Inflammatory Disorders? BIOLOGY 2022; 11:372. [PMID: 35336746 PMCID: PMC8945680 DOI: 10.3390/biology11030372] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/12/2022] [Accepted: 02/15/2022] [Indexed: 12/15/2022]
Abstract
NF-κB is a central mediator of inflammation, response to DNA damage and oxidative stress. As a result of its central role in so many important cellular processes, NF-κB dysregulation has been implicated in the pathology of important human diseases. NF-κB activation causes inappropriate inflammatory responses in diseases including rheumatoid arthritis (RA) and multiple sclerosis (MS). Thus, modulation of NF-κB signaling is being widely investigated as an approach to treat chronic inflammatory diseases, autoimmunity and cancer. The emergence of COVID-19 in late 2019, the subsequent pandemic and the huge clinical burden of patients with life-threatening SARS-CoV-2 pneumonia led to a massive scramble to repurpose existing medicines to treat lung inflammation in a wide range of healthcare systems. These efforts continue and have proven to be controversial. Drug repurposing strategies are a promising alternative to de novo drug development, as they minimize drug development timelines and reduce the risk of failure due to unexpected side effects. Different experimental approaches have been applied to identify existing medicines which inhibit NF-κB that could be repurposed as anti-inflammatory drugs.
Collapse
Affiliation(s)
| | | | - David R. Greaves
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK; (A.R.); (L.E.C.)
| |
Collapse
|
10
|
Bian J, Wang T, Sun J, He X, Wu Z, Zhang S, Chi H, Fan T, Wang S, Shi W, Ruan Q. Targeting NF-κB c-Rel in regulatory T cells to treat corneal transplantation rejection. Am J Transplant 2021; 21:3858-3870. [PMID: 34254428 DOI: 10.1111/ajt.16760] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 07/05/2021] [Accepted: 07/08/2021] [Indexed: 01/25/2023]
Abstract
The relevance of Tregs in the induction of tolerance against corneal allografts has been well established. Although it is well known that the conversion of Tregs into effector-like cells contributes to the loss of corneal immune privilege, the underlying mechanism is still not fully understood. Using heterologous penetrating keratoplasty model, we found that Tregs from corneal allograft rejected mice (inflam-Tregs) exhibit impaired function and characteristics of effector T cells. Further study showed that the expression of NF-κB c-Rel, a key mediator of effector T cell function, was significantly increased in inflam-Tregs. Mechanistic study revealed that elevated NF-κB c-Rel level in inflam-Tregs impaired Treg function through the promotion of inflammatory cytokine production and glycolysis. More importantly, we demonstrated that targeting NF-κB c-Rel was able to improve the immune suppressive function of inflam-Tregs in vitro and enhance the potential of them to suppress corneal transplantation rejection. Therefore, our current study identified NF-κB c-Rel as a key mediator of the conversion of Tregs into effector-like cells when under inflammatory environment.
Collapse
Affiliation(s)
- Jiang Bian
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China.,State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China
| | - Ting Wang
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China.,Eye Hospital of Shandong First Medical University, Jinan, China
| | - Jijun Sun
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China.,Eye Hospital of Shandong First Medical University, Jinan, China
| | - Xiaozhen He
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China.,State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China
| | - Zhijiao Wu
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China.,State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China
| | - Songmei Zhang
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China.,State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China
| | - Hao Chi
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China.,State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China
| | - Tingting Fan
- Center for Antibody Drug, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Shaowen Wang
- Center for Antibody Drug, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Weiyun Shi
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China.,Eye Hospital of Shandong First Medical University, Jinan, China
| | - Qingguo Ruan
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China.,State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China.,Center for Antibody Drug, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
11
|
Niu Y, Wang B, Zhou L, Ma C, Waterhouse GIN, Liu Z, Ahmed AF, Sun-Waterhouse D, Kang W. Nigella sativa: A Dietary Supplement as an Immune-Modulator on the Basis of Bioactive Components. Front Nutr 2021; 8:722813. [PMID: 34485368 PMCID: PMC8415885 DOI: 10.3389/fnut.2021.722813] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 07/16/2021] [Indexed: 01/04/2023] Open
Abstract
Nutrients can be considered as functional foods, which exert physiological benefits on immune system. The seeds of Nigella sativa, which have many active constituents, are mainly used for medicine, food spice, and nutritional supplements in Egypt. Much attention has been paid to N. sativa seeds for their anticancer, antibacterial, anti-inflammatory, and immune properties. However, their active constituents and mechanisms underlying functions from N. sativa seeds is unclear. Thus, the bioactive constituents with immune regulation in N. sativa seeds were systematically studied. A new compound (3-methoxythymol-6-O-β-D-apiofuranosyl-(1→6)-β-D-glucopyranoside 1) and 11 known compounds (2–12) were separated from the N. sativa seeds by chromatographic methods. Their structures were then elucidated by spectroscopic analysis of MS, UV, IR, 1H-, and 13C-NMR. Furthermore, immunomodulatory effects of those compounds in RAW 264.7 cells were evaluated by phagocytosis, nitric oxide (NO) and cytokine release, related mRNA transcription, and key proteins expression in vitro. Monosaccharide derivatives, Ethyl-α-D-furaarabinose (5), and Ethyl-β-D-fructofuranoside (8) were shown to played bidirectional regulatory roles in immunity and anti-inflammation through the regulation of nuclear factor-κB (NF-κB) signaling pathways. The results showed the active compounds and mechanisms of immune regulation in N. sativa, thus indicating that N. sativa seeds could be used as dietary supplements in immunomodulation.
Collapse
Affiliation(s)
- Yun Niu
- National R&D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China.,Functional Food Engineering Technology Research Center, Kaifeng, China
| | - Baoguang Wang
- National R&D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China.,Functional Food Engineering Technology Research Center, Kaifeng, China
| | - Li Zhou
- National R&D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China.,Functional Food Engineering Technology Research Center, Kaifeng, China.,Joint International Research Laboratory of Food and Medicine Resource Function, Kaifeng, China
| | - Changyang Ma
- National R&D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China.,Joint International Research Laboratory of Food and Medicine Resource Function, Kaifeng, China
| | - Geoffrey I N Waterhouse
- National R&D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China.,School of Chemical Sciences, University of Auckland, Auckland, New Zealand
| | - Zhenhua Liu
- National R&D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China.,Joint International Research Laboratory of Food and Medicine Resource Function, Kaifeng, China
| | - Adel F Ahmed
- Joint International Research Laboratory of Food and Medicine Resource Function, Kaifeng, China.,Medicinal and Aromatic Plants Researches Department, Agricultural Research Center, Horticulture Research Institute, Giza, Egypt
| | - Dongxiao Sun-Waterhouse
- National R&D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China.,School of Chemical Sciences, University of Auckland, Auckland, New Zealand
| | - Wenyi Kang
- National R&D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China.,Functional Food Engineering Technology Research Center, Kaifeng, China.,Joint International Research Laboratory of Food and Medicine Resource Function, Kaifeng, China
| |
Collapse
|
12
|
Kanapeckaitė A, Beaurivage C, Jančorienė L, Mažeikienė A. In silico drug discovery for a complex immunotherapeutic target - human c-Rel protein. Biophys Chem 2021; 276:106593. [PMID: 34087524 DOI: 10.1016/j.bpc.2021.106593] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/28/2021] [Accepted: 04/12/2021] [Indexed: 12/22/2022]
Abstract
Target evaluation and rational drug design rely on identifying and characterising small-molecule binding sites on therapeutically relevant target proteins. Immunotherapeutics development is especially challenging because of complex disease etiology and heterogenous nature of targets. c-Rel protein, a promising target in many human inflammatory and cancer pathologies, was selected as a case study for an effective in silico screening platform development since this transcription factor currently has no successful therapeutic inhibitors or modulators. This study introduces a novel in silico screening approach to probe binding sites using structural validation sets, molecular modelling and describes a method of a computer-aided drug design when a crystal structure is not available for the target of interest. In addition, we showed that binding sites can be analysed with the machine learning as well as molecular simulation approaches to help assess and systematically analyse how drug candidates can exert their mode of action. Finally, this cutting-edge approach was subjected to a high through-put virtual screen of selected 34 M drug-like compounds filtered from a library of 659 M compounds by identifying the most promising structures and proposing potential action mechanisms for the future development of highly selective human c-Rel inhibitors and/or modulators.
Collapse
Affiliation(s)
| | | | - Ligita Jančorienė
- Vilnius University Medical Faculty InsTtute of Clinical Medicine, Clinic of InfecTous Diseases and Dermatovenerology, Santariškių str. 14, 08406 Vilnius, Lithuania
| | - Asta Mažeikienė
- Department of Physiology, Biochemistry, Microbiology and Laboratory Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, M. K. Čiurlionio g. 21, LT-03101, Vilnius, Lithuania
| |
Collapse
|
13
|
Munmun F, Witt-Enderby PA. Melatonin effects on bone: Implications for use as a therapy for managing bone loss. J Pineal Res 2021; 71:e12749. [PMID: 34085304 DOI: 10.1111/jpi.12749] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/22/2021] [Accepted: 05/31/2021] [Indexed: 02/06/2023]
Abstract
Melatonin is the primary circadian output signal from the brain and is mainly synthesized in pinealocytes. The rhythm and secretion of melatonin are under the control of an endogenous oscillator located in the SCN or the master biological clock. Disruptions in circadian rhythms by shift work, aging, or light at night are associated with bone loss and increased fracture risk. Restoration of nocturnal melatonin peaks to normal levels or therapeutic levels through timed melatonin supplementation has been demonstrated to provide bone-protective actions in various models. Melatonin is a unique molecule with diverse molecular actions targeting melatonin receptors located on the plasma membrane or mitochondria or acting independently of receptors through its actions as an antioxidant or free radical scavenger to stimulate osteoblastogenesis, inhibit osteoclastogenesis, and improve bone density. Its additional actions on entraining circadian rhythms and improving quality of life in an aging population coupled with its safety profile make it an ideal therapeutic candidate for protecting against bone loss in susceptible populations. The intent of this review is to provide a focused discussion on bone loss and disorders of the bone as it relates to melatonin and conditions that modify melatonin levels with the hope that future therapies include those that include melatonin and correct those factors that modify melatonin levels like circadian disruption.
Collapse
Affiliation(s)
- Fahima Munmun
- Division of Pharmaceutical Sciences, Duquesne University School of Pharmacy, Pittsburgh, PA, USA
| | - Paula A Witt-Enderby
- Division of Pharmaceutical Sciences, Duquesne University School of Pharmacy, Pittsburgh, PA, USA
| |
Collapse
|
14
|
Zhang R, Xu K, Shao Y, Sun Y, Saredy J, Cutler E, Yao T, Liu M, Liu L, Drummer Iv C, Lu Y, Saaoud F, Ni D, Wang J, Li Y, Li R, Jiang X, Wang H, Yang X. Tissue Treg Secretomes and Transcription Factors Shared With Stem Cells Contribute to a Treg Niche to Maintain Treg-Ness With 80% Innate Immune Pathways, and Functions of Immunosuppression and Tissue Repair. Front Immunol 2021; 11:632239. [PMID: 33613572 PMCID: PMC7892453 DOI: 10.3389/fimmu.2020.632239] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 12/18/2020] [Indexed: 12/12/2022] Open
Abstract
We used functional -omics angles and examined transcriptomic heterogeneity in CD4+Foxp3+ regulatory T cells (Treg) from spleen (s-Treg), lymph nodes (LN-Treg), intestine (int-Treg), and visceral adipose tissue (VAT-Treg), and made significant findings: 1) Five new shared Treg genes including NIBAN, TNFRSF1b, DUSP4,VAV2, and KLRG1, and 68 new signatures are identified. Among 27 signaling pathways shared in four tissue Treg, 22 pathways are innate immune pathways (81.5%); 2) s-Treg, LN-Treg, int-Treg, and VAT-Treg have zero, 49, 45, and 116 upregulated pathways, respectively; 3) 12, 7, and 15 out of 373 CD markers are identified as specific for LN-Treg, int-Treg, and VAT-Treg, respectively, which may initiate innate immune signaling; 4) 7, 49, 44, and 79 increased cytokines out of 1176 cytokines are identified for four Treg, respectively, suggesting that Treg have much more secretory proteins/cytokines than IL-10, TGF-β, and IL-35; 5) LN-Treg, int-Treg, and VAT-Treg have 13 additional secretory functions more than s-Treg, found by analyzing 1,706 secretomic genes; 6) 2, 20, 25, and 43 increased transcription factors (TFs) out of 1,496 TFs are identified four Treg, respectively; 7) LN-Treg and int-Treg have increased pyroptosis regulators but VAT-Treg have increased apoptosis regulators; 8) 1, 15, 19, and 31 increased kinases out of 661 kinome are identified for s-Treg, LN-Treg, int-Treg, and VAT-Treg, respectively; 9) comparing with that of s-Treg, LN-Treg, int-Treg, and VAT-Treg increase activated cluster (clusters 1–3) markers; and decrease resting cluster (clusters 4–6) markers; and 10) Treg promote tissue repair by sharing secretomes and TFs AHR, ETV5, EGR1, and KLF4 with stem cells, which partially promote upregulation of all the groups of Treg genes. These results suggest that stem cell-shared master genes make tissue Treg as the first T cell type using a Treg niche to maintain their Treg-ness with 80% innate immune pathways, and triple functions of immunosuppression, tissue repair, and homeostasis maintenance. Our results have provided novel insights on the roles of innate immune pathways on Treg heterogeneity and new therapeutic targets for immunosuppression, tissue repair, cardiovascular diseases, chronic kidney disease, autoimmune diseases, transplantation, and cancers.
Collapse
Affiliation(s)
- Ruijing Zhang
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Department of Nephrology, The Second Hospital of Shanxi Medical University, Shanxi, China.,Shanxi Medical University, Shanxi, China.,Department of Nephrology, The Affiliated People's Hospital of Shanxi Medical University, Shanxi, China
| | - Keman Xu
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Ying Shao
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Yu Sun
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Jason Saredy
- Metabolic Disease Research & Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Elizabeth Cutler
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,School of Science and Engineering, Tulane University, New Orleans, LA, United States
| | - Tian Yao
- Shanxi Medical University, Shanxi, China
| | - Ming Liu
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Shanxi Medical University, Shanxi, China
| | - Lu Liu
- Metabolic Disease Research & Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Charles Drummer Iv
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Yifan Lu
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Fatma Saaoud
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Dong Ni
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Jirong Wang
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Department of Nephrology, The Second Hospital of Shanxi Medical University, Shanxi, China
| | - Yafeng Li
- Department of Nephrology, The Affiliated People's Hospital of Shanxi Medical University, Shanxi, China
| | - Rongshan Li
- Department of Nephrology, The Affiliated People's Hospital of Shanxi Medical University, Shanxi, China
| | - Xiaohua Jiang
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Metabolic Disease Research & Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Hong Wang
- Metabolic Disease Research & Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Xiaofeng Yang
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Metabolic Disease Research & Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| |
Collapse
|
15
|
Galgani M, Bruzzaniti S, La Rocca C, Micillo T, de Candia P, Bifulco M, Matarese G. Immunometabolism of regulatory T cells in cancer. Mol Aspects Med 2020; 77:100936. [PMID: 33250195 DOI: 10.1016/j.mam.2020.100936] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/19/2020] [Accepted: 11/19/2020] [Indexed: 02/08/2023]
Abstract
Regulatory T (Treg) cells are known to orchestrate the regulatory mechanisms aimed at suppressing pathological auto-reactive immune responses and are thus key in ensuring the maintenance of immune homeostasis. On the other hand, the presence of Treg cells with enhanced suppressive capability in a plethora of human cancers represents a major obstacle to an effective anti-cancer immune response. A relevant research effort has thus been dedicated to comprehend Treg cell biology, leading to a continuously refining characterization of their phenotype and function and unveiling the central role of metabolism in ensuring Treg cell fitness in cancer. Here we focus on how the peculiar biochemical characteristics of the tumor microenvironment actually support Treg cell metabolic activation and favor their selective survival and proliferation. Moreover, we examine the key metabolic pathways that may become useful targets of novel treatments directed at hampering tumor resident Treg cell proficiency, thus representing the next research frontier in cancer immunotherapy.
Collapse
Affiliation(s)
- Mario Galgani
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II", 80131, Napoli, Italy; Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale Delle Ricerche (IEOS-CNR), 80131, Napoli, Italy
| | - Sara Bruzzaniti
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale Delle Ricerche (IEOS-CNR), 80131, Napoli, Italy; Dipartimento di Biologia, Università Degli Studi di Napoli "Federico II", 80126, Napoli, Italy
| | - Claudia La Rocca
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale Delle Ricerche (IEOS-CNR), 80131, Napoli, Italy
| | - Teresa Micillo
- Unità di Neuroimmunologia, Fondazione Santa Lucia IRCCS, 00179, Roma, Italy
| | | | - Maurizio Bifulco
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II", 80131, Napoli, Italy
| | - Giuseppe Matarese
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II", 80131, Napoli, Italy; Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale Delle Ricerche (IEOS-CNR), 80131, Napoli, Italy.
| |
Collapse
|
16
|
Ayroldi E, Grohmann U. Exemplifying complexity of immune suppression by a "canonical" speech: A glimpse into TNFRSF-activated signaling pathways in Treg cells. Eur J Immunol 2020; 50:944-948. [PMID: 32428262 DOI: 10.1002/eji.202048711] [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: 05/04/2020] [Indexed: 01/03/2023]
Abstract
Regulatory T (Treg) cells are crucial mediators of immune tolerance suppressing self-reactive T cells and preventing autoimmune diseases. Besides activation of the T cell receptor (TCR), empowerment of Treg cell functions requires co-accessory signals, such as those released by the TNF receptor superfamily (TNFRSF) that, however, can also promote immunostimulatory responses when engaged by effector T cells. In this issue of European Journal of Immunology, Lubrano di Ricco et al. [Eur. J. Immunol. 2020. 50: 972-985] have taken a closer look at the important question of the functional meaning of TNFRSF-activated signaling pathways in Treg cells. They have demonstrated that costimulation by TNFR2, 4-1BB, GITR, DR3, but not OX40 in mouse Foxp3+ Treg cells activates the same and unique signaling pathway, i.e., canonical NF-κB, which in turn leads to Foxp3 gene upregulation, cell expansion in vitro and in vivo, and suppressive activity in an experimental model of colitis. Moreover, induction of markers of T helper 2 (Th2) and Th17 as well as of genes encoding proteins involved in noncanonical NF-κΒ was also observed. We here discussed how these findings further highlight the emerging concept of Treg cell plasticity in immune tolerance.
Collapse
Affiliation(s)
- Emira Ayroldi
- Department of Medicine, University of Perugia, Perugia, Italy
| | - Ursula Grohmann
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
17
|
Role of different Th17 and Treg downstream signalling pathways in the pathogenesis of Staphylococcus aureus infection induced septic arthritis in mice. Exp Mol Pathol 2020; 116:104485. [PMID: 32574668 DOI: 10.1016/j.yexmp.2020.104485] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 05/18/2020] [Accepted: 06/17/2020] [Indexed: 12/15/2022]
Abstract
Septic arthritis is a condition of bone disorder caused predominantly by Staphylococcus aureus. Following the bacterial entry activated immune cells specially macrophages and dendritic cells release pro-inflammatory mediators such as IL-6, TNF-α, IL-1β etc., which not only create an inflammatory microenvironment but also play crucial roles in the proliferation of different CD+ T cell subsets. Among them, Th17 and Tregs are of major concern in recent times because of their potential roles in regulating the ongoing inflammation in many diseases including experimental arthritis. But the downstream signalling mechanism of these cells in regulating the severity of inflammation in case of septic arthritis is not known yet. So, here we have established a murine model of S. aureus induced septic arthritis and kept the animal upto 15 days post-infection. To examine the signalling mechanism, Th17 and Treg cells were isolated from blood, spleen and synovial joints of control and infected mice and observed the expression of JNK, NFκB and RANKL in the lysate of isolated Th17 and Tregs. We have also estimated the levels of serum IL-21 and TGF-β. NFκB, JNK and RANKL expression was found to be higher at 3 and 15 days post-infection along with serum IL-21 levels. On the other hand, maximum TGF-β level was observed at 9 days post-infection along with increased Treg population. In conclusion it was hypothesized that bone resorption is related with downstream signalling pathways of Th17 cells, which stimulate osteoclast generation via NFκB/JNK-RANKL axis and helps in the persistence of the disease.
Collapse
|
18
|
Best S, Lam V, Liu T, Bruss N, Kittai A, Danilova OV, Murray S, Berger A, Pennock ND, Lind EF, Danilov AV. Immunomodulatory effects of pevonedistat, a NEDD8-activating enzyme inhibitor, in chronic lymphocytic leukemia-derived T cells. Leukemia 2020; 35:156-168. [PMID: 32203139 DOI: 10.1038/s41375-020-0794-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 02/25/2020] [Accepted: 03/03/2020] [Indexed: 01/28/2023]
Abstract
Novel targeted agents used in therapy of lymphoid malignancies, such as inhibitors of B-cell receptor-associated kinases, are recognized to have complex immune-mediated effects. NEDD8-activating enzyme (NAE) has been identified as a tractable target in chronic lymphocytic leukemia (CLL) and non-Hodgkin lymphoma. We and others have shown that pevonedistat (TAK-924), a small-molecule inhibitor of NAE, abrogates NF-κB signaling in malignant B cells. However, NF-κB pathway activity is indispensable in immune response, and T-cell function is altered in patients with CLL. Using T cells derived from patients with CLL, we demonstrate that although targeting NAE results in markedly differential expression of NF-κB-regulated genes and downregulation of interleukin (IL)-2 signaling during T-cell activation, T cells evade apoptosis. Meanwhile, NAE inhibition favorably modulates polarization of T cells in vitro, with decreased Treg differentiation and a shift toward TH1 phenotype, accompanied by increased interferon-γ production. These findings were recapitulated in vivo in immunocompetent mouse models. T cells exposed to pevonedistat in washout experiments, informed by its human pharmacokinetic profile, recover NAE activity, and maintain their response to T-cell receptor stimulation and cytotoxic potential. Our data shed light on the potential immune implications of targeting neddylation in CLL and lymphoid malignancies.
Collapse
Affiliation(s)
- Scott Best
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Vi Lam
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Tingting Liu
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Nur Bruss
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Adam Kittai
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Olga V Danilova
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | | | | | - Nathan D Pennock
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR, USA
| | - Evan F Lind
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Alexey V Danilov
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA. .,City of Hope National Medical Center, 1500 E Duarte Rd, Duarte, CA, 91010, USA.
| |
Collapse
|
19
|
Fan ZD, Cao Q, Huang N, Ma L, Ma HH, Zhang YY, Yu HG, Zhou GP. MicroRNA-125b regulates Th17/Treg cell differentiation and is associated with juvenile idiopathic arthritis. World J Pediatr 2020; 16:99-110. [PMID: 31102153 DOI: 10.1007/s12519-019-00265-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 05/02/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Juvenile idiopathic arthritis (JIA) is the most common rheumatic disease in childhood driven by aberrant pathways of T-cell activation. T helper 17 (Th17)/regulatory T cell (Treg) imbalance plays critical roles in the pathogenesis of arthritis. MicroRNA-125b (miR-125b) was upregulated after the activation of the initial CD4+ T cells, and could regulate the differentiation of CD4+ T cells. However, the effects of miR-125b on Th17/Treg imbalance and differentiation of Th17/Treg cells remain unknown. METHODS In this study, we evaluated the expression of miR-125b in the peripheral blood mononuclear cells (PBMCs) of children with JIA, and the relationship of miR-125b with Th17/Treg imbalance. Then, we used lentivirus vector-mediated overexpression technology to investigate the regulatory function of miR-125b in CD4+ T cells or dendritic cell/CD4+ T co-culture system. RESULTS Decreased miR-125b expression in PBMCs and CD4+ T cells of JIA patients was negatively correlated with the ratio of Th17/Treg cells. It also correlated negatively with retinoic acid receptor-related orphan receptor γt but positively with Forkhead box protein 3 at transcriptional levels. Furthermore, we found that miR-125b overexpression inhibited Th17 cell differentiation, whereas facilitated the differentiation of Treg cells. MiR-125b upregulation led to the decrease of Th17-secreting cytokines but the increase of the Treg-secreting cytokines. CONCLUSIONS Our results demonstrate that miR-125b participated in regulating Th17/Treg cell differentiation and imbalance in JIA patients. These findings provide novel insight into the critical role of miR-125b in the Th17/Treg imbalance of JIA, and raise the distinct possibility that miR-125b may prove to be a potential therapeutic target for JIA.
Collapse
Affiliation(s)
- Zhi-Dan Fan
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.,Department of Rheumatology and Immunology, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Qian Cao
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Na Huang
- Department of Rheumatology and Immunology, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Le Ma
- Department of Rheumatology and Immunology, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Hui-Hui Ma
- Department of Rheumatology and Immunology, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Ya-Yuan Zhang
- Department of Rheumatology and Immunology, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Hai-Guo Yu
- Department of Rheumatology and Immunology, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Guo-Ping Zhou
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
20
|
Chen M, Zhang C, Zhang J, Kai G, Lu B, Huang Z, Ji L. The involvement of DAMPs-mediated inflammation in cyclophosphamide-induced liver injury and the protection of liquiritigenin and liquiritin. Eur J Pharmacol 2019; 856:172421. [DOI: 10.1016/j.ejphar.2019.172421] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 05/23/2019] [Accepted: 05/24/2019] [Indexed: 02/07/2023]
|
21
|
Chadha S, Wang L, Hancock WW, Beier UH. Sirtuin-1 in immunotherapy: A Janus-headed target. J Leukoc Biol 2019; 106:337-343. [PMID: 30605226 PMCID: PMC7477756 DOI: 10.1002/jlb.2ru1118-422r] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 12/13/2018] [Accepted: 12/16/2018] [Indexed: 12/16/2022] Open
Abstract
Sirtuin-1 (Sirt1), a member of the NAD-dependent sirtuin family of histone/protein deacetylases (HDAC), is an important target for immunotherapy due to its role in deacetylating the transcription factors Foxp3 and thymic retinoid acid receptor related orphan receptor gamma (RORγt). Sirt1 inhibition can increase Foxp3 acetylation and promote the production and functions of Foxp3+ T-regulatory (Treg) cells, whereas the acetylation of RORγt decreases its transcriptional activity DNA binding and decreases the differentiation of proinflammatory Th17 cells. Pharmacologic inhibitors of Sirt1 increase allograft survival and decrease autoimmune colitis and experimental allergic encephalomyelitis. However, in contrast to its role in T cells, Sirt1 has anti-inflammatory effects in myeloid cells, and, context dependent, in Th17 cells. Here, inhibition of Sirt1 can have proinflammatory effects. In addition to effects arising from the central role of Sirt1 in cellular metabolism and NAD-dependent reactions, such proinflammatory effects further complicate the potential of Sirt1 for therapeutic immunosuppression. This review aims to reconcile the opposing literature on pro- and anti-inflammatory effects of Sirt1, provides an overview of the role of Sir1 in the immune system, and discusses the pros and cons associated with inhibiting Sirt1 for control of inflammation and immune responses.
Collapse
Affiliation(s)
- Sakshum Chadha
- Division of Nephrology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, University of Pennsylvania, Philadelphia, PA 19104, USA
- Current address: Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Liqing Wang
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, and Biesecker Center for Pediatric Liver Disease, Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wayne W. Hancock
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, and Biesecker Center for Pediatric Liver Disease, Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ulf H. Beier
- Division of Nephrology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
22
|
Bing Z, Jin-Tao D, Feng L, Ba L, Ya-Feng L, Shi-Xi L. Effect of Astragalus membranaceus in Ovalbumin-Induced Allergic Rhinitis Mouse Model. Am J Rhinol Allergy 2019; 33:420-432. [PMID: 30945558 DOI: 10.1177/1945892419839259] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Background Astragalus membranaceus (AM), a traditional Chinese medicine, has been used to treat allergic diseases, but the mechanism for treating allergic rhinitis (AR) remains unclear. Objective The purpose of this study was to look at the anti-inflammatory effect of AM on AR and the mechanism of anti-allergy. Methods The mouse model of AR was induced by ovalbumin. Allergic symptoms, number of eosinophils in nasal mucosa, and levels of inflammatory cells in nasal lavage fluid were analyzed. We explored the serum immunoglobulin E (IgE), interleukin-4 (IL-4), IL-5, IL-13, interferon-γ (IFN-γ), and IL-10. Besides, the relative mRNA of IL-4, IL-5, and IL-13 was also detected in nasal mucosa tissue. The proportion of CD4+ CD25+ Foxp3+ T cells in the spleen and nasal lymphoid tissue were analyzed. The mRNA levels of nuclear factor-kappa B p65 (NF-κB p65) and inhibitory kappa B alpha (IκBα), as well as NF-κB p65 DNA binding activity, were tested. We also measured the protein levels of NF-κB p65 and p-NF-κB p65 in nasal mucosa. Results AM could reduce the number of eosinophils in the nasal mucosa and decrease the levels of inflammatory cells in nasal lavage fluid. The serum IgE, IL-4, IL-5, and IL-13 were also decreased, while levels of IFN-γ and IL-10 were increased. The relative mRNA of IL-4, IL-5, and IL-13 was decreased by AM. AM increased the proportion of CD4+ CD25+ Foxp3+ T cells in the spleen and nasal lymphoid tissue. In addition, AM could reduce the activity of NF-kB by inhibiting the mRNA expression and DNA binding activity of NF-κB p65. However, AM had no significant effect on mRNA of IκBα. Above all, AM could reduce the p-NF-κB p65 protein expression of nasal mucosa. Conclusions AM could reduce the secretion of inflammatory cytokines by increasing the level of CD4+ CD25+ Foxp3+ T cells and inhibiting the activation of the NF-κB.
Collapse
Affiliation(s)
- Zhong Bing
- 1 Department of Otolaryngology-Head and Neck Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Du Jin-Tao
- 1 Department of Otolaryngology-Head and Neck Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Liu Feng
- 1 Department of Otolaryngology-Head and Neck Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Luo Ba
- 2 Department of Otolaryngology-Head and Neck Surgery, People's Hospital of Tibet Autonomous Region, Lhasa, China
| | - Liu Ya-Feng
- 1 Department of Otolaryngology-Head and Neck Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Liu Shi-Xi
- 1 Department of Otolaryngology-Head and Neck Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| |
Collapse
|
23
|
Nabavi SM, Nabavi SF, Sureda A, Xiao J, Dehpour AR, Shirooie S, Silva AS, Baldi A, Khan H, Daglia M. Anti-inflammatory effects of Melatonin: A mechanistic review. Crit Rev Food Sci Nutr 2019; 59:S4-S16. [DOI: 10.1080/10408398.2018.1487927] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Affiliation(s)
- Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Seyed Fazel Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Antoni Sureda
- Grup de Nutrici_o Comunit_aria i Estr_es Oxidatiu and CIBEROBN (Physiopathology of Obesity and Nutrition), Universitat de les Illes Balears, Palma de E-07122 Mallorca, Spain
| | - Janbo Xiao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Control in Chinese Medicine, University of Macau, Macau SAR, China
| | - Ahmad Reza Dehpour
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Samira Shirooie
- School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ana Sanches Silva
- National Institute for Agricultural and Veterinary Research (INIAV), I.P., Vairão, Vila do Conde, Portugal; Center for Study in Animal Science (CECA), ICETA, University of Oporto, Oporto, Portugal
| | - Alessandra Baldi
- Department of Drug Sciences, Medicinal Chemistry and Pharmaceutical Technology Section, University of Pavia, Pavia, Italy
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan, Pakistan
| | - Maria Daglia
- Department of Drug Sciences, Medicinal Chemistry and Pharmaceutical Technology Section, University of Pavia, Pavia, Italy
| |
Collapse
|
24
|
Bevacizumab-Based Chemotherapy Triggers Immunological Effects in Responding Multi-Treated Recurrent Ovarian Cancer Patients by Favoring the Recruitment of Effector T Cell Subsets. J Clin Med 2019; 8:jcm8030380. [PMID: 30889935 PMCID: PMC6462947 DOI: 10.3390/jcm8030380] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 03/11/2019] [Accepted: 03/13/2019] [Indexed: 12/09/2022] Open
Abstract
Increasing evidence strongly suggests that bevacizumab compound impacts the immunological signature of cancer patients and normalizes tumor vasculature. This study aims to investigate the correlation between the clinical response to bevacizumab-based chemotherapy and the improvement of immune fitness of multi-treated ovarian cancer patients. Peripheral blood mononuclear cells (PBMCs) of 20 consecutive recurrent ovarian cancer patients retrospectively selected to have received bevacizumab or non-bevacizumab-based chemotherapy (Bev group and Ctrl group, respectively) were analyzed. CD4, CD8, and regulatory T cell (Treg) subsets were monitored at the beginning (T0) and after three and six cycles of treatment, together with IL10 production. A lower activated and resting Treg subset was found in the Bev group compared with the Ctrl group until the third therapy cycle, suggesting a reduced immunosuppressive signature. Indeed, clinically responding patients in the Bev group showed a high percentage of non-suppressive Treg and a significant lower IL10 production compared with non-responding patients in the Bev group after three cycles. Furthermore, clinically responding patients showed a discrete population of effector T cell at T0 independent of the therapeutic regimen. This subset was maintained throughout the therapy in only the Bev group. This study evidences that bevacizumab could affect the clinical response of cancer patients, reducing the percentage of Treg and sustaining the circulation of the effector T cells. Results also provide a first rationale regarding the positive immunologic synergism of combining bevacizumab with immunotherapy in multi-treated ovarian cancer patients.
Collapse
|
25
|
Napoletano C, Ruscito I, Bellati F, Zizzari IG, Rahimi H, Gasparri ML, Antonilli M, Panici PB, Rughetti A, Nuti M. Bevacizumab-Based Chemotherapy Triggers Immunological Effects in Responding Multi-Treated Recurrent Ovarian Cancer Patients by Favoring the Recruitment of Effector T Cell Subsets. J Clin Med 2019. [PMID: 30889935 DOI: 10.3390/jcm8030380] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Increasing evidence strongly suggests that bevacizumab compound impacts the immunological signature of cancer patients and normalizes tumor vasculature. This study aims to investigate the correlation between the clinical response to bevacizumab-based chemotherapy and the improvement of immune fitness of multi-treated ovarian cancer patients. Peripheral blood mononuclear cells (PBMCs) of 20 consecutive recurrent ovarian cancer patients retrospectively selected to have received bevacizumab or non-bevacizumab-based chemotherapy (Bev group and Ctrl group, respectively) were analyzed. CD4, CD8, and regulatory T cell (Treg) subsets were monitored at the beginning (T0) and after three and six cycles of treatment, together with IL10 production. A lower activated and resting Treg subset was found in the Bev group compared with the Ctrl group until the third therapy cycle, suggesting a reduced immunosuppressive signature. Indeed, clinically responding patients in the Bev group showed a high percentage of non-suppressive Treg and a significant lower IL10 production compared with non-responding patients in the Bev group after three cycles. Furthermore, clinically responding patients showed a discrete population of effector T cell at T0 independent of the therapeutic regimen. This subset was maintained throughout the therapy in only the Bev group. This study evidences that bevacizumab could affect the clinical response of cancer patients, reducing the percentage of Treg and sustaining the circulation of the effector T cells. Results also provide a first rationale regarding the positive immunologic synergism of combining bevacizumab with immunotherapy in multi-treated ovarian cancer patients.
Collapse
Affiliation(s)
- Chiara Napoletano
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy.
| | - Ilary Ruscito
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy.
- Tumor Bank Ovarian Cancer Network (TOC), Department of Gynecology, European Competence Center for Ovarian Cancer, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Augustenburger Platz 1, D-13353 Berlin, Germany.
| | - Filippo Bellati
- Department of Medical and Surgical Sciences and Translational Medicine, Sant'Andrea University Hospital, Sapienza University of Rome, Via di Grottarossa 1035, 00189 Rome, Italy.
| | - Ilaria Grazia Zizzari
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy.
| | - Hassan Rahimi
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy.
| | - Maria Luisa Gasparri
- Department of Maternal and Child and Urological Sciences, Policlinico Umberto I "Sapienza" University of Rome, Viale del Policlinico 155, 00161 Rome, Italy.
| | - Morena Antonilli
- Department of Maternal and Child and Urological Sciences, Policlinico Umberto I "Sapienza" University of Rome, Viale del Policlinico 155, 00161 Rome, Italy.
| | - Pierluigi Benedetti Panici
- Department of Maternal and Child and Urological Sciences, Policlinico Umberto I "Sapienza" University of Rome, Viale del Policlinico 155, 00161 Rome, Italy.
| | - Aurelia Rughetti
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy.
| | - Marianna Nuti
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy.
| |
Collapse
|
26
|
Russo MA, Fiore NT, van Vreden C, Bailey D, Santarelli DM, McGuire HM, Fazekas de St Groth B, Austin PJ. Expansion and activation of distinct central memory T lymphocyte subsets in complex regional pain syndrome. J Neuroinflammation 2019; 16:63. [PMID: 30885223 PMCID: PMC6423749 DOI: 10.1186/s12974-019-1449-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 02/28/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Complex regional pain syndrome (CRPS) is a debilitating condition where trauma to a limb results in devastating persistent pain that is disproportionate to the initial injury. The pathophysiology of CRPS remains unknown; however, accumulating evidence suggests it is an immunoneurological disorder, especially in light of evidence of auto-antibodies in ~ 30% of patients. Despite this, a systematic assessment of all circulating leukocyte populations in CRPS has never been performed. METHODS We characterised 14 participants as meeting the Budapest clinical criteria for CRPS and assessed their pain ratings and psychological state using a series of questionnaires. Next, we performed immunophenotyping on blood samples from the 14 CRPS participants as well as 14 healthy pain-free controls using mass cytometry. Using a panel of 38 phenotypic and activation markers, we characterised the numbers and intracellular activation status of all major leukocyte populations using manual gating strategies and unsupervised cluster analysis. RESULTS We have shown expansion and activation of several distinct populations of central memory T lymphocytes in CRPS. The number of central memory CD8+ T cells was increased 2.15-fold; furthermore, this cell group had increased phosphorylation of NFkB and STAT1 compared to controls. Regarding central memory CD4+ T lymphocytes, the number of Th1 and Treg cells was increased 4.98-fold and 2.18-fold respectively, with increased phosphorylation of NFkB in both populations. We also found decreased numbers of CD1c+ myeloid dendritic cells, although with increased p38 phosphorylation. These changes could indicate dendritic cell tissue trafficking, as well as their involvement in lymphocyte activation. CONCLUSIONS These findings represent the first mass cytometry immunophenotyping study in any chronic pain state and provide preliminary evidence of an antigen-mediated T lymphocyte response in CRPS. In particular, the presence of increased numbers of long-lived central memory CD4+ and CD8+ T lymphocytes with increased activation of pro-inflammatory signalling pathways may indicate ongoing inflammation and cellular damage in CRPS.
Collapse
Affiliation(s)
- Marc A. Russo
- Hunter Pain Clinic, 91 Chatham Street, Broadmeadow, NSW 2292 Australia
- Genesis Research Services, 220 Denison St, Broadmeadow, NSW 2292 Australia
| | - Nathan T. Fiore
- Discipline of Anatomy & Histology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Room E513, Anderson Stuart Building, Sydney, NSW 2006 Australia
| | - Caryn van Vreden
- Ramaciotti Centre for Human Systems Biology, Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006 Australia
- Sydney Cytometry, Centenary Institute and the Charles Perkins Centre, John Hopkins Drive, Camperdown, NSW 2050 Australia
| | - Dominic Bailey
- Genesis Research Services, 220 Denison St, Broadmeadow, NSW 2292 Australia
| | | | - Helen M. McGuire
- Ramaciotti Centre for Human Systems Biology, Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006 Australia
- Discipline of Pathology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006 Australia
| | - Barbara Fazekas de St Groth
- Ramaciotti Centre for Human Systems Biology, Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006 Australia
- Discipline of Pathology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006 Australia
| | - Paul J. Austin
- Discipline of Anatomy & Histology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Room E513, Anderson Stuart Building, Sydney, NSW 2006 Australia
| |
Collapse
|
27
|
Comito G, Iscaro A, Bacci M, Morandi A, Ippolito L, Parri M, Montagnani I, Raspollini MR, Serni S, Simeoni L, Giannoni E, Chiarugi P. Lactate modulates CD4 + T-cell polarization and induces an immunosuppressive environment, which sustains prostate carcinoma progression via TLR8/miR21 axis. Oncogene 2019; 38:3681-3695. [PMID: 30664688 DOI: 10.1038/s41388-019-0688-7] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 12/07/2018] [Accepted: 12/25/2018] [Indexed: 01/01/2023]
Abstract
Leukocyte infiltration plays an active role in controlling tumor development. In the early stages of carcinogenesis, T cells counteract tumor growth. However, in advanced stages, cancer cells and infiltrating stromal components interfere with the immune control and instruct immune cells to support, rather than counteract, tumor malignancy, via cell-cell contact or soluble mediators. In particular, metabolites are emerging as active players in driving immunosuppression. Here we demonstrate that in a prostate cancer model lactate released by glycolytic cancer-associated fibroblasts (CAFs) acts on CD4+ T cells, shaping T-cell polarization. In particular, CAFs exposure (i) reduces the percentage of the antitumoral Th1 subset, inducing a lactate-dependent, SIRT1-mediated deacetylation/degradation of T-bet transcription factor; (ii) increases Treg cells, driving naive T cells polarization, through a lactate-based NF-kB activation and FoxP3 expression. In turn, this metabolic-based CAF-immunomodulated environment exerts a pro-invasive effect on prostate cancer cells, by activating a previously unexplored miR21/TLR8 axis that sustains cancer malignancy.
Collapse
Affiliation(s)
- G Comito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134, Florence, Italy
| | - A Iscaro
- Department of Oncology and Metabolism, University of Sheffield, Medical School, Beech Hill Road, Sheffield, UK
| | - M Bacci
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134, Florence, Italy
| | - A Morandi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134, Florence, Italy
| | - L Ippolito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134, Florence, Italy
| | - M Parri
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134, Florence, Italy
| | - I Montagnani
- Histopathology and Molecular Diagnostics, University Hospital Careggi, Largo Brambilla, 3, 50134, Florence, Italy
| | - M R Raspollini
- Histopathology and Molecular Diagnostics, University Hospital Careggi, Largo Brambilla, 3, 50134, Florence, Italy
| | - S Serni
- Department of Urological Robotic Surgery and Renal Transplantation, University of Florence, Careggi Hospital, Florence, 50134, Italy
| | - L Simeoni
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University, Leipziger Str. 44, D-39120, Magdeburg, Germany
| | - E Giannoni
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134, Florence, Italy.
| | - P Chiarugi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134, Florence, Italy.,Tuscany Tumor Institute (ITT) and Excellence Centre for Research, Transfer and High Education DenoTHE, Florence, 50134, Italy
| |
Collapse
|
28
|
PD-1 immunobiology in systemic lupus erythematosus. J Autoimmun 2018; 97:1-9. [PMID: 30396745 DOI: 10.1016/j.jaut.2018.10.025] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 10/22/2018] [Accepted: 10/28/2018] [Indexed: 01/22/2023]
Abstract
Programmed death (PD)-1 receptors and their ligands have been identified in the pathogenesis and development of systemic lupus erythematosus (SLE). Two key pathways, toll-like receptor and type I interferon, are significant to SLE pathogenesis and modulate the expression of PD-1 and the ligands (PD-L1, PD-L2) through activation of NF-κB and/or STAT1. These cell signals are regulated by tyrosine kinase (Tyro, Axl, Mer) receptors (TAMs) that are aberrantly activated in SLE. STAT1 and NF-κB also exhibit crosstalk with the aryl hydrocarbon receptor (AHR). Ligands to AHR are identified in SLE etiology and pathogenesis. These ligands also regulate the activity of the Epstein-Barr virus (EBV), which is an identified factor in SLE and PD-1 immunobiology. AHR is important in the maintenance of immune tolerance and the development of distinct immune subsets, highlighting a potential role of AHR in PD-1 immunobiology. Understanding the functions of AHR ligands as well as AHR crosstalk with STAT1, NF-κB, and EBV may provide insight into disease development, the PD-1 axis and immunotherapies that target PD-1 and its ligand, PD-L1.
Collapse
|
29
|
Torabi S, Tamaddon M, Asadolahi M, Shokri G, Tavakoli R, Tasharrofi N, Rezaei R, Tavakolpour V, Sazegar H, Kouhkan F. miR-455-5p downregulation promotes inflammation pathways in the relapse phase of relapsing-remitting multiple sclerosis disease. Immunogenetics 2018; 71:87-95. [DOI: 10.1007/s00251-018-1087-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 09/21/2018] [Indexed: 01/01/2023]
|
30
|
Wu X, Wang W, Chen Y, Liu X, Wang J, Qin X, Yuan D, Yu T, Chen G, Mi Y, Mou J, Cui J, Hu A, E Y, Pei D. High Mobility Group Box Protein 1 Serves as a Potential Prognostic Marker of Lung Cancer and Promotes Its Invasion and Metastasis by Matrix Metalloproteinase-2 in a Nuclear Factor- κB-Dependent Manner. BIOMED RESEARCH INTERNATIONAL 2018; 2018:3453706. [PMID: 29850505 PMCID: PMC5933054 DOI: 10.1155/2018/3453706] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 12/01/2017] [Accepted: 02/04/2018] [Indexed: 12/15/2022]
Abstract
Several studies have reported a significant role of high mobility group box protein 1 (HMGB1) in lung cancer. Nevertheless, there is a lack of knowledge regarding the expression of HMGB1 and its correlation with the clinicopathological features of lung cancer. In addition, the potential molecular mechanisms underlying the role of HMGB1 in lung cancer are still unknown. We therefore investigated the clinicopathological and prognostic significance as well as the potential role of HMGB1 in the development and progression of lung cancer. HMGB1 expression in the tumor tissues of the cohort correlated with clinicopathological features. Moreover, lung cell migration and invasion were significantly increased after treatment with HMGB1. The matrix metalloproteinase-2 (MMP-2) expression and activity were upregulated after treatment with HMGB1, while the upregulated expression of MMP-2 stimulated by HMGB1 in lung cancer cells was significantly reduced with the blockage of si-p65. These results indicated that HMGB1 expression was significantly associated with lung cancer progression. We also showed that HMGB1 promoted lung cancer invasion and metastasis by upregulating the expression and activity of MMP-2 in an NF-κB-dependent manner. Taken together, these data suggested that HMGB1 may be a potential prognosis and therapeutic marker for lung cancer.
Collapse
Affiliation(s)
- Xiaojin Wu
- Department of Radiation Oncology, The First People's Hospital of Xuzhou, Xuzhou, Jiangsu 221002, China
| | - Weitao Wang
- Geneis Beijing Co., Ltd., Beijing 100102, China
| | - Yuanyuan Chen
- Department of Radiation Oncology, The First People's Hospital of Xuzhou, Xuzhou, Jiangsu 221002, China
| | - Xiangqun Liu
- Department of Respiratory Diseases, The First People's Hospital of Xuzhou, Xuzhou, Jiangsu 221002, China
| | - Jindong Wang
- Department of Chest Surgery, The First People's Hospital of Xuzhou, Xuzhou, Jiangsu 221002, China
| | - Xiaobin Qin
- Department of Tumor, The First People's Hospital of Xuzhou, Xuzhou, Jiangsu 221002, China
| | - Dawei Yuan
- Geneis Beijing Co., Ltd., Beijing 100102, China
| | - Tao Yu
- Department of Tumor, The First People's Hospital of Xuzhou, Xuzhou, Jiangsu 221002, China
| | - Guangxia Chen
- Department of Gastroenterology, The First People's Hospital of Xuzhou, Xuzhou, Jiangsu 221002, China
| | - Yanyan Mi
- Department of Pharmacy, Xuzhou Medical College, Xuzhou, Jiangsu 221004, China
| | - Jie Mou
- Department of Pharmacy, Xuzhou Medical College, Xuzhou, Jiangsu 221004, China
| | - Jinpeng Cui
- Clinical Laboratory of Yantaishan Hospital, No. 91, Jiefang Road, Yantai, Shandong 264001, China
| | - Ankang Hu
- Laboratory Animal Center, Xuzhou Medical College, Xuzhou, Jiangsu 221004, China
| | - Yunxiang E
- Department of Tumor, The First People's Hospital of Xuzhou, Xuzhou, Jiangsu 221002, China
| | - Dongsheng Pei
- Department of Pathology, Xuzhou Medical College, Xuzhou, Jiangsu 221004, China
| |
Collapse
|
31
|
Fan T, Zhong F, Liu R, Chen YH, Wang T, Ruan Q. siRNA-mediated c-Rel knockdown ameliorates collagen-induced arthritis in mice. Int Immunopharmacol 2018; 56:9-17. [DOI: 10.1016/j.intimp.2018.01.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/19/2017] [Accepted: 01/04/2018] [Indexed: 12/18/2022]
|
32
|
Jevtić B, Djedović N, Stanisavljević S, Gašić U, Mišić D, Despotović J, Samardžić J, Miljković D, Timotijević G. Anti-encephalitogenic effects of cucumber leaf extract. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.07.060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
33
|
Limagne E, Thibaudin M, Euvrard R, Berger H, Chalons P, Végan F, Humblin E, Boidot R, Rébé C, Derangère V, Ladoire S, Apetoh L, Delmas D, Ghiringhelli F. Sirtuin-1 Activation Controls Tumor Growth by Impeding Th17 Differentiation via STAT3 Deacetylation. Cell Rep 2017; 19:746-759. [DOI: 10.1016/j.celrep.2017.04.004] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 02/24/2017] [Accepted: 03/31/2017] [Indexed: 01/01/2023] Open
|
34
|
Silencing c‐Rel in macrophages dampens Th1 and Th17 immune responses and alleviates experimental autoimmune encephalomyelitis in mice. Immunol Cell Biol 2017; 95:593-600. [DOI: 10.1038/icb.2017.11] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 02/13/2017] [Accepted: 02/13/2017] [Indexed: 12/25/2022]
|
35
|
Li J, Jing J, Bai Y, Li Z, Xing R, Tan B, Ma X, Qiu WW, Du C, Du B, Yang F, Tang J, Siwko S, Liu M, Chen H, Luo J. SH479, a Betulinic Acid Derivative, Ameliorates Experimental Autoimmune Encephalomyelitis by Regulating the T Helper 17/Regulatory T Cell Balance. Mol Pharmacol 2017; 91:464-474. [PMID: 28213589 DOI: 10.1124/mol.116.107136] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 02/15/2017] [Indexed: 12/19/2022] Open
Abstract
CD4+ T helper cells, especially T helper 17 (TH17) cells, combined with immune regulatory network dysfunction, play key roles in autoimmune diseases including multiple sclerosis (MS). Betulinic acid (BA), a natural pentacyclic triterpenoid, has been reported to be involved in anti-inflammation, in particular having an inhibitory effect on proinflammatory cytokine interleukin 17 (IL-17) and interferon-γ (IFN-γ) production. In this study, we screened BA derivatives and found a BA derivative, SH479, that had a greater inhibitory effect on TH17 differentiation. Our further analysis showed that SH479 had a greater inhibitory effect on TH17 and TH1, and a more stimulatory effect on regulatory T (Treg) cells. To evaluate the effects of SH479 on autoimmune diseases in vivo, we employed the extensively used MS mouse model experimental autoimmune encephalomyelitis (EAE). Our results showed that SH479 ameliorated clinical and histologic signs of EAE in both prevention and therapeutic protocols by regulating the TH17/Treg balance. SH479 dose-dependently reduced splenic lymphocyte proinflammatory factors and increased anti-inflammatory factors. Moreover, SH479 specifically inhibited splenic lymphocyte viability from EAE mice but not normal splenic lymphocyte viability. At the molecular level, SH479 inhibited TH17 differentiation by regulating signal transducer and activator of transcription-3 (STAT3) phosphorylation, DNA binding activity, and recruitment to the Il-17a promoter in CD4+ T cells. Furthermore, SH479 promoted the STAT5 signaling pathway and inhibited the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathway. Together, our data demonstrated that SH479 ameliorated EAE by regulating the TH17/Treg balance through inhibiting the STAT3 and NF-κB pathways while activating the STAT5 pathway, suggesting that SH479 is a potential novel drug candidate for autoimmune diseases including MS.
Collapse
Affiliation(s)
- Jing Li
- Shanghai Fengxian District Central Hospital, Shanghai, People's Republic of China (J.Li); Shanghai Fengxian District Central Hospital and East China Normal University Joint Center for Translational Medicine, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, People's Republic of China (J.J., Y.B., Z.L., R.X., B.T., X.M., B.D., M.L., H.C., J.Luo); Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, People's Republic of China (W.-W.Q., F.Y., J.T.); Laboratory of Receptor-Based Bio-medicine, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China (C.D.); Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, Texas (S.S., M.L.)
| | - Ji Jing
- Shanghai Fengxian District Central Hospital, Shanghai, People's Republic of China (J.Li); Shanghai Fengxian District Central Hospital and East China Normal University Joint Center for Translational Medicine, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, People's Republic of China (J.J., Y.B., Z.L., R.X., B.T., X.M., B.D., M.L., H.C., J.Luo); Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, People's Republic of China (W.-W.Q., F.Y., J.T.); Laboratory of Receptor-Based Bio-medicine, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China (C.D.); Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, Texas (S.S., M.L.)
| | - Yang Bai
- Shanghai Fengxian District Central Hospital, Shanghai, People's Republic of China (J.Li); Shanghai Fengxian District Central Hospital and East China Normal University Joint Center for Translational Medicine, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, People's Republic of China (J.J., Y.B., Z.L., R.X., B.T., X.M., B.D., M.L., H.C., J.Luo); Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, People's Republic of China (W.-W.Q., F.Y., J.T.); Laboratory of Receptor-Based Bio-medicine, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China (C.D.); Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, Texas (S.S., M.L.)
| | - Zhen Li
- Shanghai Fengxian District Central Hospital, Shanghai, People's Republic of China (J.Li); Shanghai Fengxian District Central Hospital and East China Normal University Joint Center for Translational Medicine, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, People's Republic of China (J.J., Y.B., Z.L., R.X., B.T., X.M., B.D., M.L., H.C., J.Luo); Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, People's Republic of China (W.-W.Q., F.Y., J.T.); Laboratory of Receptor-Based Bio-medicine, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China (C.D.); Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, Texas (S.S., M.L.)
| | - Roumei Xing
- Shanghai Fengxian District Central Hospital, Shanghai, People's Republic of China (J.Li); Shanghai Fengxian District Central Hospital and East China Normal University Joint Center for Translational Medicine, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, People's Republic of China (J.J., Y.B., Z.L., R.X., B.T., X.M., B.D., M.L., H.C., J.Luo); Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, People's Republic of China (W.-W.Q., F.Y., J.T.); Laboratory of Receptor-Based Bio-medicine, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China (C.D.); Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, Texas (S.S., M.L.)
| | - Binhe Tan
- Shanghai Fengxian District Central Hospital, Shanghai, People's Republic of China (J.Li); Shanghai Fengxian District Central Hospital and East China Normal University Joint Center for Translational Medicine, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, People's Republic of China (J.J., Y.B., Z.L., R.X., B.T., X.M., B.D., M.L., H.C., J.Luo); Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, People's Republic of China (W.-W.Q., F.Y., J.T.); Laboratory of Receptor-Based Bio-medicine, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China (C.D.); Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, Texas (S.S., M.L.)
| | - Xueyun Ma
- Shanghai Fengxian District Central Hospital, Shanghai, People's Republic of China (J.Li); Shanghai Fengxian District Central Hospital and East China Normal University Joint Center for Translational Medicine, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, People's Republic of China (J.J., Y.B., Z.L., R.X., B.T., X.M., B.D., M.L., H.C., J.Luo); Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, People's Republic of China (W.-W.Q., F.Y., J.T.); Laboratory of Receptor-Based Bio-medicine, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China (C.D.); Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, Texas (S.S., M.L.)
| | - Wen-Wei Qiu
- Shanghai Fengxian District Central Hospital, Shanghai, People's Republic of China (J.Li); Shanghai Fengxian District Central Hospital and East China Normal University Joint Center for Translational Medicine, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, People's Republic of China (J.J., Y.B., Z.L., R.X., B.T., X.M., B.D., M.L., H.C., J.Luo); Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, People's Republic of China (W.-W.Q., F.Y., J.T.); Laboratory of Receptor-Based Bio-medicine, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China (C.D.); Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, Texas (S.S., M.L.)
| | - Changsheng Du
- Shanghai Fengxian District Central Hospital, Shanghai, People's Republic of China (J.Li); Shanghai Fengxian District Central Hospital and East China Normal University Joint Center for Translational Medicine, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, People's Republic of China (J.J., Y.B., Z.L., R.X., B.T., X.M., B.D., M.L., H.C., J.Luo); Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, People's Republic of China (W.-W.Q., F.Y., J.T.); Laboratory of Receptor-Based Bio-medicine, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China (C.D.); Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, Texas (S.S., M.L.)
| | - Bing Du
- Shanghai Fengxian District Central Hospital, Shanghai, People's Republic of China (J.Li); Shanghai Fengxian District Central Hospital and East China Normal University Joint Center for Translational Medicine, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, People's Republic of China (J.J., Y.B., Z.L., R.X., B.T., X.M., B.D., M.L., H.C., J.Luo); Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, People's Republic of China (W.-W.Q., F.Y., J.T.); Laboratory of Receptor-Based Bio-medicine, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China (C.D.); Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, Texas (S.S., M.L.)
| | - Fan Yang
- Shanghai Fengxian District Central Hospital, Shanghai, People's Republic of China (J.Li); Shanghai Fengxian District Central Hospital and East China Normal University Joint Center for Translational Medicine, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, People's Republic of China (J.J., Y.B., Z.L., R.X., B.T., X.M., B.D., M.L., H.C., J.Luo); Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, People's Republic of China (W.-W.Q., F.Y., J.T.); Laboratory of Receptor-Based Bio-medicine, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China (C.D.); Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, Texas (S.S., M.L.)
| | - Jie Tang
- Shanghai Fengxian District Central Hospital, Shanghai, People's Republic of China (J.Li); Shanghai Fengxian District Central Hospital and East China Normal University Joint Center for Translational Medicine, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, People's Republic of China (J.J., Y.B., Z.L., R.X., B.T., X.M., B.D., M.L., H.C., J.Luo); Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, People's Republic of China (W.-W.Q., F.Y., J.T.); Laboratory of Receptor-Based Bio-medicine, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China (C.D.); Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, Texas (S.S., M.L.)
| | - Stefan Siwko
- Shanghai Fengxian District Central Hospital, Shanghai, People's Republic of China (J.Li); Shanghai Fengxian District Central Hospital and East China Normal University Joint Center for Translational Medicine, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, People's Republic of China (J.J., Y.B., Z.L., R.X., B.T., X.M., B.D., M.L., H.C., J.Luo); Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, People's Republic of China (W.-W.Q., F.Y., J.T.); Laboratory of Receptor-Based Bio-medicine, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China (C.D.); Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, Texas (S.S., M.L.)
| | - Mingyao Liu
- Shanghai Fengxian District Central Hospital, Shanghai, People's Republic of China (J.Li); Shanghai Fengxian District Central Hospital and East China Normal University Joint Center for Translational Medicine, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, People's Republic of China (J.J., Y.B., Z.L., R.X., B.T., X.M., B.D., M.L., H.C., J.Luo); Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, People's Republic of China (W.-W.Q., F.Y., J.T.); Laboratory of Receptor-Based Bio-medicine, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China (C.D.); Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, Texas (S.S., M.L.)
| | - Huaqing Chen
- Shanghai Fengxian District Central Hospital, Shanghai, People's Republic of China (J.Li); Shanghai Fengxian District Central Hospital and East China Normal University Joint Center for Translational Medicine, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, People's Republic of China (J.J., Y.B., Z.L., R.X., B.T., X.M., B.D., M.L., H.C., J.Luo); Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, People's Republic of China (W.-W.Q., F.Y., J.T.); Laboratory of Receptor-Based Bio-medicine, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China (C.D.); Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, Texas (S.S., M.L.)
| | - Jian Luo
- Shanghai Fengxian District Central Hospital, Shanghai, People's Republic of China (J.Li); Shanghai Fengxian District Central Hospital and East China Normal University Joint Center for Translational Medicine, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, People's Republic of China (J.J., Y.B., Z.L., R.X., B.T., X.M., B.D., M.L., H.C., J.Luo); Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, People's Republic of China (W.-W.Q., F.Y., J.T.); Laboratory of Receptor-Based Bio-medicine, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China (C.D.); Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, Texas (S.S., M.L.)
| |
Collapse
|
36
|
Jevtić B, Djedović N, Stanisavljević S, Despotović J, Miljković D, Timotijević G. Cucurbitacin E Potently Modulates the Activity of Encephalitogenic Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:4900-7. [PMID: 27225664 DOI: 10.1021/acs.jafc.6b00951] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Cucurbitacin E (CucE) is a highly oxidized steroid consisting of a tetracyclic triterpene. It is a member of a Cucurbitacin family of biomolecules that are predominantly found in Cucurbitaceae plants. CucE has already been identified as a potent anti-inflammatory compound. Here, its effects on CD4(+) T helper (Th) cells and macrophages, as the major encephalitogenic cells in the autoimmunity of the central nervous system, were investigated. Production of major pathogenic Th cell cytokines: interferon-gamma and interleukin-17 were inhibited under the influence of CucE. The effects of CucE on CD4(+) T cells were mediated through the modulation of aryl hydrocarbon receptor, STAT3, NFκB, p38 MAPK, and miR-146 signaling. Further, production of nitric oxide and reactive oxygen species, as well as phagocytic ability, were inhibited in macrophages treated with CucE. These results imply that CucE possesses powerful antiencephalitogenic activity.
Collapse
Affiliation(s)
- Bojan Jevtić
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade , Belgrade, Serbia
| | - Neda Djedović
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade , Belgrade, Serbia
| | - Suzana Stanisavljević
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade , Belgrade, Serbia
| | - Jovana Despotović
- Laboratory for Molecular Biology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade , Belgrade, Serbia
| | - Djordje Miljković
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade , Belgrade, Serbia
| | - Gordana Timotijević
- Laboratory for Plant Molecular Biology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade , Belgrade, Serbia
| |
Collapse
|
37
|
Hussman JP, Beecham AH, Schmidt M, Martin ER, McCauley JL, Vance JM, Haines JL, Pericak-Vance MA. GWAS analysis implicates NF-κB-mediated induction of inflammatory T cells in multiple sclerosis. Genes Immun 2016; 17:305-12. [PMID: 27278126 PMCID: PMC4956564 DOI: 10.1038/gene.2016.23] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 04/22/2016] [Accepted: 04/29/2016] [Indexed: 12/13/2022]
Abstract
To identify genes and biologically relevant pathways associated with risk to develop multiple sclerosis (MS), the Genome-Wide Association Studies noise reduction method (GWAS-NR) was applied to MS genotyping data. Regions of association were defined based on the significance of linkage disequilibrium blocks. Candidate genes were cross-referenced based on a review of current literature, with attention to molecular function and directly interacting proteins. Supplementary annotations and pathway enrichment scores were generated using The Database for Annotation, Visualization and Integrated Discovery. The candidate set of 220 MS susceptibility genes prioritized by GWAS-NR was highly enriched with genes involved in biological pathways related to positive regulation of cell, lymphocyte and leukocyte activation (P=6.1E-15, 1.2E-14 and 5.0E-14, respectively). Novel gene candidates include key regulators of NF-κB signaling and CD4+ T helper type 1 (Th1) and T helper type 17 (Th17) lineages. A large subset of MS candidate genes prioritized by GWAS-NR were found to interact in a tractable pathway regulating the NF-κB-mediated induction and infiltration of pro-inflammatory Th1/Th17 T-cell lineages, and maintenance of immune tolerance by T-regulatory cells. This mechanism provides a biological context that potentially links clinical observations in MS to the underlying genetic landscape that may confer susceptibility.
Collapse
Affiliation(s)
| | - A H Beecham
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - M Schmidt
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - E R Martin
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL, USA.,Dr John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - J L McCauley
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL, USA.,Dr John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - J M Vance
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL, USA.,Dr John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, USA.,Department of Neurology, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - J L Haines
- Department of Epidemiology & Biostatistics, Case Western Reserve University, Cleveland, OH, USA
| | - M A Pericak-Vance
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL, USA.,Dr John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, USA.,Department of Neurology, University of Miami, Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
38
|
Choudhury S, Gupta P, Ghosh S, Mukherjee S, Chakraborty P, Chatterji U, Chattopadhyay S. Arsenic-induced dose-dependent modulation of the NF-κB/IL-6 axis in thymocytes triggers differential immune responses. Toxicology 2016; 357-358:85-96. [PMID: 27289040 DOI: 10.1016/j.tox.2016.06.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 06/01/2016] [Accepted: 06/08/2016] [Indexed: 12/22/2022]
Abstract
Arsenic contamination of drinking water is a matter of global concern. Arsenic intake impairs immune responses and leads to a variety of pathological conditions including cancer. In order to understand the intricate tuning of immune responses elicited by chronic exposure to arsenic, a mouse model was established by subjecting mice to different environmentally relevant concentrations of arsenic in drinking water for 30days. Detailed study of the thymus, a primary immune organ, revealed arsenic-mediated tissue damage in both histological specimens and scanning electron micrographs. Analysis of molecular markers of apoptosis by Western blot revealed a dose-dependent activation of the apoptotic cascade. Enzymatic assays supported oxidative stress as an instigator of cell death. Interestingly, assessment of inflammatory responses revealed disparity in the NF-κB/IL-6/STAT3 axis, where it was found that in animals consuming higher amounts of arsenic NF-κB activation did not lead to the classical IL-6 upregulation response. This deviation from the canonical pathway was accompanied with a significant rise in numbers of CD4+ CD25+ FoxP3 expressing cells in the thymus. The cytokine profile of the animals exposed to higher doses of arsenic also indicated an immune-suppressed milieu, thus validating that arsenic shapes the immune environment in context to its dose of exposure and that at higher doses it leads to immune-suppression. Our study establishes a novel role of arsenic in regulating immune homeostasis in context to its dose, where, at higher doses, arsenic related upregulation of NF-κB cascade takes on an alternative role that is correlated with increased immune-suppression.
Collapse
Affiliation(s)
- Sreetama Choudhury
- Department of Physiology, UCSTA, University of Calcutta, 92, A.P.C. Road, Kolkata 700009, India
| | - Payal Gupta
- Department of Physiology, UCSTA, University of Calcutta, 92, A.P.C. Road, Kolkata 700009, India
| | - Sayan Ghosh
- Department of Physiology, UCSTA, University of Calcutta, 92, A.P.C. Road, Kolkata 700009, India
| | - Sudeshna Mukherjee
- Department of Physiology, UCSTA, University of Calcutta, 92, A.P.C. Road, Kolkata 700009, India
| | - Priyanka Chakraborty
- Department of Physiology, UCSTA, University of Calcutta, 92, A.P.C. Road, Kolkata 700009, India
| | - Urmi Chatterji
- Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, India
| | - Sreya Chattopadhyay
- Department of Physiology, UCSTA, University of Calcutta, 92, A.P.C. Road, Kolkata 700009, India.
| |
Collapse
|
39
|
Fan T, Wang S, Yu L, Yi H, Liu R, Geng W, Wan X, Ma Y, Cai L, Chen YH, Ruan Q. Treating psoriasis by targeting its susceptibility gene Rel. Clin Immunol 2016; 165:47-54. [DOI: 10.1016/j.clim.2016.03.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 03/02/2016] [Accepted: 03/13/2016] [Indexed: 12/22/2022]
|
40
|
Shi JH, Sun SC. TCR signaling to NF-κB and mTORC1: Expanding roles of the CARMA1 complex. Mol Immunol 2015; 68:546-57. [PMID: 26260210 PMCID: PMC4679546 DOI: 10.1016/j.molimm.2015.07.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 06/07/2015] [Accepted: 07/19/2015] [Indexed: 12/25/2022]
Abstract
Naïve T-cell activation requires signals from both the T-cell receptor (TCR) and the costimulatory molecule CD28. A central mediator of the TCR and CD28 signals is the scaffold protein CARMA1, which functions by forming a complex with partner proteins, Bcl10 and MALT1. A well-known function of the CARMA1 signaling complex is to mediate activation of IκB kinase (IKK) and its target transcription factor NF-κB, thereby promoting T-cell activation and survival. Recent evidence suggests that CARMA1 also mediates TCR/CD28-stimulated activation of the IKK-related kinase TBK1, which plays a role in regulating the homeostasis and migration of T cells. Moreover, the CARMA1 complex connects the TCR/CD28 signals to the activation of mTORC1, a metabolic kinase regulating various aspects of T-cell functions. This review will discuss the mechanism underlying the activation of the CARMA1-dependent signaling pathways and their roles in regulating T-cell functions.
Collapse
Affiliation(s)
- Jian-hong Shi
- Central Laboratory, Affiliated Hospital of Hebei University, 212 Yuhua East Road, Baoding 071000, China
| | - Shao-Cong Sun
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030, USA.
| |
Collapse
|
41
|
Sasaki CY, Chen G, Munk R, Eitan E, Martindale J, Longo DL, Ghosh P. p(⁷⁰S⁶K¹) in the TORC1 pathway is essential for the differentiation of Th17 Cells, but not Th1, Th2, or Treg cells in mice. Eur J Immunol 2015; 46:212-22. [PMID: 26514620 DOI: 10.1002/eji.201445422] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 09/24/2015] [Accepted: 10/12/2015] [Indexed: 12/24/2022]
Abstract
The TORC1 pathway is necessary for ribosomal biogenesis and initiation of protein translation. Furthermore, the differentiation of Th1 and Th17 cells requires TORC1 activity. To investigate the role of the TORC1 pathway in the differentiation of Th1 and/or Th17 cells in more detail, we compared the differentiation capacity of naïve T cells from wild type and p70(S6K1) knockout mice. Expression of many of the genes associated with Th17-cell differentiation, such as IL17a, IL17f, and IL-23R, were reduced in p70(S6K1) knockout mice. In contrast, the development of Th1, Th2, and Treg cells was unaffected in the absence of p70(S6K1) . Furthermore, expression of the major transcription factor in Th17-cell differentiation, retinoic acid receptor-related orphan receptor gamma T, remained unchanged. However, the acetylation of histone 3 at the promoters of IL17a and IL17f was reduced in the absence of p70(S6K1) . In accordance with the in vitro data, the kinetics, but not the development, of EAE was affected with the loss of p70(S6K1) expression. Collectively, our findings suggested that both in vitro and in vivo differentiation of Th17 cells were positively regulated by p70(S6K1) .
Collapse
Affiliation(s)
- Carl Y Sasaki
- Laboratory of Genetics, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Gang Chen
- Laboratory of Genetics, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Rachel Munk
- Laboratory of Genetics, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Erez Eitan
- Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Jennifer Martindale
- Laboratory of Genetics, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Dan L Longo
- Laboratory of Genetics, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Paritosh Ghosh
- Laboratory of Genetics, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| |
Collapse
|
42
|
Min HK, Kim SM, Baek SY, Woo JW, Park JS, Cho ML, Lee J, Kwok SK, Kim SW, Park SH. Anthocyanin Extracted from Black Soybean Seed Coats Prevents Autoimmune Arthritis by Suppressing the Development of Th17 Cells and Synthesis of Proinflammatory Cytokines by Such Cells, via Inhibition of NF-κB. PLoS One 2015; 10:e0138201. [PMID: 26544846 PMCID: PMC4636296 DOI: 10.1371/journal.pone.0138201] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 08/26/2015] [Indexed: 12/29/2022] Open
Abstract
INTRODUCTION Oxidative stress plays a role in the pathogenesis of rheumatoid arthritis (RA). Anthocyanin is a plant antioxidant. We investigated the therapeutic effects of anthocyanin extracted from black soybean seed coats (AEBS) in a murine model of collagen-induced arthritis (CIA) and human peripheral blood mononuclear cells (PBMCs) and explored possible mechanisms by which AEBS might exert anti-arthritic effects. MATERIAL AND METHODS CIA was induced in DBA/1J mice. Cytokine levels were measured via enzyme-linked immunosorbent assays. Joints were assessed in terms of arthritis incidence, clinical arthritis scores, and histological features. The extent of oxidative stress in affected joints was determined by measuring the levels of nitrotyrosine and inducible nitric oxide synthase. NF-κB activity was assayed by measuring the ratio of phosphorylated IκB to total IκB via Western blotting. Th17 cells were stained with antibodies against CD4, IL-17, and STAT3. Osteoclast formation was assessed via TRAP staining and measurement of osteoclast-specific mRNA levels. RESULTS In the CIA model, AEBS decreased the incidence of arthritis, histological inflammation, cartilage scores, and oxidative stress. AEBS reduced the levels of proinflammatory cytokines in affected joints of CIA mice and suppressed NF-κB signaling. AEBS decreased Th17 cell numbers in spleen of CIA mice. Additionally, AEBS repressed differentiation of Th17 cells and expression of Th17-associated genes in vitro, in both splenocytes of naïve DBA/1J mice and human PBMCs. In vitro, the numbers of both human and mouse tartrate-resistant acid phosphatase+ (TRAP) multinucleated cells fell, in a dose-dependent manner, upon addition of AEBS. CONCLUSIONS The anti-arthritic effects of AEBS were associated with decreases in Th17 cell numbers, and the levels of proinflammatory cytokines synthesized by such cells, mediated via suppression of NF-κB signaling. Additionally, AEBS suppressed osteoclastogenesis and reduced oxidative stress levels.
Collapse
MESH Headings
- Animals
- Anthocyanins/pharmacology
- Arthritis, Experimental/chemically induced
- Arthritis, Experimental/drug therapy
- Arthritis, Experimental/metabolism
- Arthritis, Experimental/pathology
- Autoimmune Diseases/chemically induced
- Autoimmune Diseases/drug therapy
- Autoimmune Diseases/metabolism
- Autoimmune Diseases/pathology
- Blotting, Western
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Cell Differentiation/drug effects
- Cells, Cultured
- Collagen/toxicity
- Cytokines/genetics
- Cytokines/metabolism
- Enzyme-Linked Immunosorbent Assay
- Flow Cytometry
- Humans
- Immunoenzyme Techniques
- Leukocytes, Mononuclear/cytology
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/metabolism
- Male
- Mice
- Mice, Inbred DBA
- NF-kappa B/antagonists & inhibitors
- NF-kappa B/genetics
- NF-kappa B/metabolism
- Osteoclasts/cytology
- Osteoclasts/drug effects
- Osteoclasts/metabolism
- Plant Extracts/pharmacology
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Reverse Transcriptase Polymerase Chain Reaction
- Seeds/chemistry
- Signal Transduction/drug effects
- Glycine max/chemistry
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Th17 Cells/drug effects
- Th17 Cells/immunology
- Th17 Cells/metabolism
Collapse
Affiliation(s)
- Hong Ki Min
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Sung-Min Kim
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| | - Seung-Ye Baek
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| | - Jung-Won Woo
- Office of Clinical Development, Genexine Inc., Korea Bio Park, Seongnam, Gyeonggi-do, South Korea
| | - Jin-Sil Park
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| | - Mi-La Cho
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| | - Jennifer Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Seung-Ki Kwok
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Sae Woong Kim
- Catholic Integrative Medicine Research Institute, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Sung-Hwan Park
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
43
|
Hsu SY, Liou JW, Cheng TL, Peng SY, Lin CC, Chu YY, Luo WC, Huang ZK, Jiang SJ. beta-Naphthoflavone protects from peritonitis by reducing TNF-alpha-induced endothelial cell activation. Pharmacol Res 2015; 102:192-9. [PMID: 26453957 DOI: 10.1016/j.phrs.2015.10.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Revised: 10/01/2015] [Accepted: 10/02/2015] [Indexed: 01/06/2023]
Abstract
β-Naphthoflavone (β-NF), a ligand of the aryl hydrocarbon receptor, has been shown to possess anti-oxidative properties. We investigated the anti-oxidative and anti-inflammatory potential of β-NF in human microvascular endothelial cells treated with tumor necrosis factor-alpha (TNF-α). Pretreatment with β-NF significantly inhibited TNF-α-induced intracellular reactive oxygen species, translocation of p67(phox), and TNF-α-induced monocyte binding and transmigration. In addition, β-NF significantly inhibited TNF-α-induced ICAM-1 and VCAM-1 expression. The mRNA expression levels of the inflammatory cytokines TNF-α and IL-6 were reduced by β-NF, as was the infiltration of white blood cells, in a peritonitis model. The inhibition of adhesion molecules was associated with suppressed nuclear translocation of NF-κB p65 and Akt, and suppressed phosphorylation of ERK1/2 and p38. The translocation of Egr-1, a downstream transcription factor involved in the MEK-ERK signaling pathway, was suppressed by β-NF treatment. Our findings show that β-NF inhibits TNF-α-induced NF-kB and ERK1/2 activation and ROS generation, thereby suppressing the expression of adhesion molecules. This results in reduced adhesion and transmigration of leukocytes in vitro and prevents the infiltration of leukocytes in a peritonitis model. Our findings also suggest that β-NF might prevent TNF-α-induced inflammation.
Collapse
Affiliation(s)
- Sheng-Yao Hsu
- Department ofOphthalmology,ChinaMedicalUniversity-AnNan Hospital,Tainan,Taiwan.; School of Medicine, China Medical University, Taichung, Taiwan
| | - Je-Wen Liou
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Tsung-Lin Cheng
- Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shih-Yi Peng
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Chi-Chen Lin
- Institute of Biomedical Sciences, College of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Yuan-Yuan Chu
- Postgraduate program in Biochemistry, School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Wei-Cheng Luo
- Master program in Microbiology, Immunology and Biochemistry, School of Medicine Master Thesis, Tzu Chi University, Hualien, Taiwan
| | - Zheng-Kai Huang
- Bachelor in Department of Molecular Biology and Human Genetics, College of Life Sciences, Tzu Chi University, Hualien, Taiwan
| | - Shinn-Jong Jiang
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, Taiwan.
| |
Collapse
|
44
|
Zhou X, Dong L, Yang B, He Z, Chen Y, Deng T, Huang B, Lan C. Preinduction of heat shock protein 70 protects mice against post-infection irritable bowel syndrome via NF-κB and NOS/NO signaling pathways. Amino Acids 2015. [PMID: 26215736 DOI: 10.1007/s00726-015-2056-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
This study aimed to investigate the protective effects of preinduction of heat shock protein 70 (HSP70) on Trichinella spiralis infection-induced post-infectious irritable bowel syndrome (PI-IBS) in mice. Trichinella spiralis infection significantly reduced HSP70 abundance, ileal villus height and crypt depth, expression of tight junctions, serum lysine and arginine concentrations, and ileal SCL7A6 and SCL7A7 mRNA levels, induced inflammatory response, and activated NF-κB signaling pathway. Meanwhile, the heat treatment upregulated HSP70 expression, and then reversed intestinal dysfunction and inflammatory response. Preinduction of HSP70 enhanced serum arginine and intestinal SCL7A7 expression and inhibited NF-κB activation compared with PI-IBS model. Treatment with pyrrolidine dithiocarbamate (PDTC, an NF-κB inhibitor) and N-nitro-L-arginine methyl ester hydrochloride (L-NAME, a nitric oxide synthase inhibitor, NOS) further demonstrated that preinduction of HSP70 might inhibit NF-κB and activated NOS/nitric oxide (NO) signaling pathways. In conclusion, preinduction of HSP70 by heat treatment may confer beneficial effects on Trichinella spiralis infection-induced PI-IBS in mice, and the protective effect of HSP70 may be associated with inhibition of NF-κB and stimulation of NOS/NO signaling pathways.
Collapse
Affiliation(s)
- Xuchun Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Liwei Dong
- Department of Gastroenterology, Hainan Provincial General Hospital, Haikou, 570311, China
| | - Bo Yang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhoutao He
- Department of Gastroenterology, Hainan Provincial General Hospital, Haikou, 570311, China
| | - Yiyao Chen
- Department of Gastroenterology, Hainan Provincial General Hospital, Haikou, 570311, China
| | - Taozhi Deng
- Department of Gastroenterology, Hainan Provincial General Hospital, Haikou, 570311, China
| | - Baili Huang
- Department of Gastroenterology, Hainan Provincial General Hospital, Haikou, 570311, China
| | - Cheng Lan
- Department of Gastroenterology, Hainan Provincial General Hospital, Haikou, 570311, China.
| |
Collapse
|
45
|
Lipopolysaccharide directly stimulates Th17 differentiation in vitro modulating phosphorylation of RelB and NF-κB1. Immunol Lett 2015; 165:10-9. [PMID: 25794633 DOI: 10.1016/j.imlet.2015.03.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 02/15/2015] [Accepted: 03/09/2015] [Indexed: 12/31/2022]
Abstract
Toll-like receptors (TLRs) recognize a wide range of pathogen-associated molecular patterns (PAMP) and are preferentially expressed in innate immune cells. TLR-mediated activation of these cells activates the adaptive immune system. However, it has become clear that TLRs are not only expressed but also functionally active in CD4 T cells. The intestines are continuously exposed to TLR ligands, including lipopolysaccharide (LPS), a TLR4 ligand, and TLR4 is expressed higher in Th17 cells than Th1 and Th2 cells. In addition, development of Th17 cells in the gut mucosa is more dependent on gut microbiota than Th1, Th2, and Treg. Thus, we examined whether LPS directly regulates Th17 differentiation. LPS directly stimulated Th17 differentiation in vitro. In Th17 cells, LPS increased phosphorylation of NF-κB1, resulting in an increase of p50, the processed form of NF-κB1, whereas it decreased phosphorylation of RelB, leading to the up-regulation of RelB. Subcutaneous injection of LPS increased the frequency of IL-17 producing cells in inguinal lymph nodes, worsening experimental autoimmune encephalomyelitis (EAE). Additionally, expression of TLR1, TLR2, TLR4, and TLR5 was reduced upon T cell activation and LPS showed modest effect on TLR4 expression. These findings provide the first evidence that TLR4 activation directly regulate Th17 differentiation.
Collapse
|
46
|
Annemann M, Wang Z, Plaza-Sirvent C, Glauben R, Schuster M, Ewald Sander F, Mamareli P, Kühl AA, Siegmund B, Lochner M, Schmitz I. IκBNS Regulates Murine Th17 Differentiation during Gut Inflammation and Infection. THE JOURNAL OF IMMUNOLOGY 2015; 194:2888-98. [DOI: 10.4049/jimmunol.1401964] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
47
|
Lou Q, Zhang W, Liu G, Ma Y. The C-type lectin OCILRP2 costimulates EL4 T cell activation via the DAP12-Raf-MAP kinase pathway. PLoS One 2014; 9:e113218. [PMID: 25411776 PMCID: PMC4239057 DOI: 10.1371/journal.pone.0113218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 10/21/2014] [Indexed: 11/30/2022] Open
Abstract
OCILRP2 is a typical Type-II transmembrane protein that is selectively expressed in activated T lymphocytes, dendritic cells, and B cells and functions as a novel co-stimulator of T cell activation. However, the signaling pathways underlying OCILRP2 in T cell activation are still not completely understood. In this study, we found that the knockdown of OCILRP2 expression with shRNA or the blockage of its activity by an anti-OCILRP2 antagonist antibody reduced CD3/CD28-costimulated EL4 T cell viability and IL-2 production, inhibit Raf1, MAPK3, and MAPK8 activation, and impair NFAT and NF-κB transcriptional activities. Furthermore, immunoprecipitation results indicated that OCILRP2 could interact with the DAP12 protein, an adaptor containing an intracellular ITAM motif that can transduce signals to induce MAP kinase activation for T cell activation. Our data reveal that after binding with DAP12, OCILRP2 activates the Raf-MAP kinase pathways, resulting in T cell activation.
Collapse
Affiliation(s)
- Qiang Lou
- Henan Engineering Lab of Antibody Medicine, Key Laboratory of Cellular and Molecular Immunology, Medical College of Henan University, Kaifeng 475004, China
| | - Wei Zhang
- Henan Engineering Lab of Antibody Medicine, Key Laboratory of Cellular and Molecular Immunology, Medical College of Henan University, Kaifeng 475004, China
| | - Guangchao Liu
- Henan Engineering Lab of Antibody Medicine, Key Laboratory of Cellular and Molecular Immunology, Medical College of Henan University, Kaifeng 475004, China
| | - Yuanfang Ma
- Henan Engineering Lab of Antibody Medicine, Key Laboratory of Cellular and Molecular Immunology, Medical College of Henan University, Kaifeng 475004, China
- * E-mail:
| |
Collapse
|
48
|
Lutz ER, Wu AA, Bigelow E, Sharma R, Mo G, Soares K, Solt S, Dorman A, Wamwea A, Yager A, Laheru D, Wolfgang CL, Wang J, Hruban RH, Anders RA, Jaffee EM, Zheng L. Immunotherapy converts nonimmunogenic pancreatic tumors into immunogenic foci of immune regulation. Cancer Immunol Res 2014; 2:616-31. [PMID: 24942756 PMCID: PMC4082460 DOI: 10.1158/2326-6066.cir-14-0027] [Citation(s) in RCA: 376] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is considered a "nonimmunogenic" neoplasm. Single-agent immunotherapies have failed to demonstrate significant clinical activity in PDAC and other "nonimmunogenic" tumors, in part due to a complex tumor microenvironment (TME) that provides a formidable barrier to immune infiltration and function. We designed a neoadjuvant and adjuvant clinical trial comparing an irradiated, granulocyte-macrophage colony-stimulating factor (GM-CSF)-secreting, allogeneic PDAC vaccine (GVAX) given as a single agent or in combination with low-dose cyclophosphamide to deplete regulatory T cells (Treg) as a means to study how the TME is altered by immunotherapy. Examination of resected PDACs revealed the formation of vaccine-induced intratumoral tertiary lymphoid aggregates in 33 of 39 patients 2 weeks after vaccine treatment. Immunohistochemical analysis showed these aggregates to be regulatory structures of adaptive immunity. Microarray analysis of microdissected aggregates identified gene-expression signatures in five signaling pathways involved in regulating immune-cell activation and trafficking that were associated with improved postvaccination responses. A suppressed Treg pathway and an enhanced Th17 pathway within these aggregates were associated with improved survival, enhanced postvaccination mesothelin-specific T-cell responses, and increased intratumoral Teff:Treg ratios. This study provides the first example of immune-based therapy converting a "nonimmunogenic" neoplasm into an "immunogenic" neoplasm by inducing infiltration of T cells and development of tertiary lymphoid structures in the TME. Post-GVAX T-cell infiltration and aggregate formation resulted in the upregulation of immunosuppressive regulatory mechanisms, including the PD-1-PD-L1 pathway, suggesting that patients with vaccine-primed PDAC may be better candidates than vaccine-naïve patients for immune checkpoint and other immunomodulatory therapies.
Collapse
Affiliation(s)
- Eric R Lutz
- Authors' Affiliations: Departments of Oncology, The Sidney Kimmel Cancer Center; The Skip Viragh Center for Pancreatic Cancer Research and Clinical Care; The Sol Goldman Pancreatic Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | - Annie A Wu
- Authors' Affiliations: Departments of Oncology, Surgery; The Sidney Kimmel Cancer Center
| | - Elaine Bigelow
- Authors' Affiliations: Departments of Oncology, The Sidney Kimmel Cancer Center
| | | | - Guanglan Mo
- Authors' Affiliations: Departments of Oncology, The Sidney Kimmel Cancer Center; The Skip Viragh Center for Pancreatic Cancer Research and Clinical Care
| | - Kevin Soares
- Authors' Affiliations: Departments of Oncology, Surgery; The Sidney Kimmel Cancer Center; The Skip Viragh Center for Pancreatic Cancer Research and Clinical Care; The Sol Goldman Pancreatic Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | - Sara Solt
- Authors' Affiliations: Departments of Oncology, The Sidney Kimmel Cancer Center; The Skip Viragh Center for Pancreatic Cancer Research and Clinical Care
| | - Alvin Dorman
- Authors' Affiliations: Departments of Oncology, The Sidney Kimmel Cancer Center; The Skip Viragh Center for Pancreatic Cancer Research and Clinical Care
| | - Anthony Wamwea
- Authors' Affiliations: Departments of Oncology, The Sidney Kimmel Cancer Center; The Skip Viragh Center for Pancreatic Cancer Research and Clinical Care
| | - Allison Yager
- Authors' Affiliations: Departments of Oncology, The Sidney Kimmel Cancer Center
| | - Daniel Laheru
- Authors' Affiliations: Departments of Oncology, The Sidney Kimmel Cancer Center; The Skip Viragh Center for Pancreatic Cancer Research and Clinical Care
| | - Christopher L Wolfgang
- Authors' Affiliations: Departments of Oncology, Surgery; The Sidney Kimmel Cancer Center; The Sol Goldman Pancreatic Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | - Jiang Wang
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Ralph H Hruban
- Authors' Affiliations: Departments of Oncology, Pathology, and The Sidney Kimmel Cancer Center; The Sol Goldman Pancreatic Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | - Robert A Anders
- Authors' Affiliations: Departments of Oncology, Pathology, and The Sidney Kimmel Cancer Center; The Sol Goldman Pancreatic Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | - Elizabeth M Jaffee
- Authors' Affiliations: Departments of Oncology, Pathology, and The Sidney Kimmel Cancer Center; The Skip Viragh Center for Pancreatic Cancer Research and Clinical Care; The Sol Goldman Pancreatic Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | - Lei Zheng
- Authors' Affiliations: Departments of Oncology, Surgery; The Sidney Kimmel Cancer Center; The Skip Viragh Center for Pancreatic Cancer Research and Clinical Care; The Sol Goldman Pancreatic Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| |
Collapse
|
49
|
Abstract
The ability of adaptive immune system to protect higher vertebrates from pathogens resides in the ability of B and T cells to express different antigen specific receptors and to respond to different threats by activating distinct differentiation and/or activation pathways. In the past 10 years, the major role of epigenetics in controlling molecular mechanisms responsible for these peculiar features and, more in general, for lymphocyte development has become evident. KRAB-ZFPs is the widest family of mammalian transcriptional repressors, which function through the recruitment of the co-factor KRAB-Associated Protein 1 (KAP1) that in turn engages histone modifiers inducing heterochromatin formation. Although most of the studies on KRAB proteins have been performed in embryonic cells, more recent reports highlighted a relevant role for these proteins also in adult tissues. This article will review the role of KRAB-ZFP and KAP1 in the epigenetic control of mouse and human adaptive immune cells.
Collapse
|
50
|
A novel synthetic dibenzocyclooctadiene lignan analog XLYF-104-6 attenuates lipopolysaccharide-induced inflammatory response in RAW264.7 macrophage cells and protects BALB/c mice from sepsis. Eur J Pharmacol 2014; 729:22-9. [DOI: 10.1016/j.ejphar.2014.01.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 12/31/2013] [Accepted: 01/09/2014] [Indexed: 11/19/2022]
|