51
|
Sabir JSM, El Omri A, Banaganapalli B, Al-Shaeri MA, Alkenani NA, Sabir MJ, Hajrah NH, Zrelli H, Ciesla L, Nasser KK, Elango R, Shaik NA, Khan M. Dissecting the Role of NF-κb Protein Family and Its Regulators in Rheumatoid Arthritis Using Weighted Gene Co-Expression Network. Front Genet 2019; 10:1163. [PMID: 31824568 PMCID: PMC6879671 DOI: 10.3389/fgene.2019.01163] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 10/23/2019] [Indexed: 12/26/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic synovial autoinflammatory disease that destructs the cartilage and bone, leading to disability. The functional regulation of major immunity-related pathways like nuclear factor kappa B (NF-κB), which is involved in the chronic inflammatory reactions underlying the development of RA, remains to be explored. Therefore, this study has adopted statistical and knowledge-based systemic investigations (like gene correlation, semantic similarity, and topological parameters based on graph theory) to study the gene expression status of NF-κB protein family (NKPF) and its regulators in synovial tissues to trace the molecular pathways through which these regulators contribute to RA. A complex protein–protein interaction map (PPIM) of 2,742 genes and 37,032 interactions was constructed from differentially expressed genes (p ≤ 0.05). PPIM was further decomposed into a Regulator Allied Protein Interaction Network (RAPIN) based on the interaction between genes (5 NKPF, 31 seeds, 131 hubs, and 652 bottlenecks). Pathway network analysis has shown the RA-specific disturbances in the functional connectivity between seed genes (RIPK1, ATG7, TLR4, TNFRSF1A, KPNA1, CFLAR, SNW1, FOSB, PARVA, CX3CL1, and TRPC6) and NKPF members (RELA, RELB, NFKB2, and REL). Interestingly, these genes are known for their involvement in inflammation and immune system (signaling by interleukins, cytokine signaling in immune system, NOD-like receptor signaling, MAPK signaling, Toll-like receptor signaling, and TNF signaling) pathways connected to RA. This study, for the first time, reports that SNW1, along with other NK regulatory genes, plays an important role in RA pathogenesis and might act as potential biomarker for RA. Additionally, these genes might play important roles in RA pathogenesis, as well as facilitate the development of effective targeted therapies. Our integrative data analysis and network-based methods could accelerate the identification of novel drug targets for RA from high-throughput genomic data.
Collapse
Affiliation(s)
- Jamal S M Sabir
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Genomics and Biotechnology Section and Research Group, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdelfatteh El Omri
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Genomics and Biotechnology Section and Research Group, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Babajan Banaganapalli
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Majed A Al-Shaeri
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Naser A Alkenani
- Biology-Zoology Division, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mumdooh J Sabir
- Department of Computer Sciences, Faculty of Computers and Information Technology, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nahid H Hajrah
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Genomics and Biotechnology Section and Research Group, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Houda Zrelli
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Genomics and Biotechnology Section and Research Group, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Lukasz Ciesla
- Department of Biological Sciences, Science and Engineering Complex, The University of Alabama, Tuscaloosa, AL, United States
| | - Khalidah K Nasser
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ramu Elango
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Noor Ahmad Shaik
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Muhummadh Khan
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Genomics and Biotechnology Section and Research Group, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
52
|
Zhang J, Xu R, Wu L, Jiang J. Expression and function of Toll‑like receptors in peripheral blood mononuclear cells in patients with ankylosing spondylitis. Mol Med Rep 2019; 20:3565-3572. [PMID: 31485664 PMCID: PMC6755152 DOI: 10.3892/mmr.2019.10631] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 05/29/2019] [Indexed: 12/11/2022] Open
Abstract
Ankylosing spondylitis (AS) is a common chronic inflammatory autoimmune disease. Toll‑like receptors (TLRs) are involved in non‑specific immunity. In the present study, the roles of TLRs in AS were investigated. The levels of inflammatory cytokines were detected by ELISA and reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR). The expression levels of TLRs and nuclear factor‑κB (NF‑κB) signaling‑associated factors were determined via RT‑qPCR and western blot analyses. It was observed that the levels of interleukin (IL)‑6, tumor necrosis factor‑α (TNF‑α), C‑reactive protein, TLR4 and TLR5 were increased in patients with AS, whereas those of IL‑10 and TLR3 were decreased. Pomalidomide, a TNF‑α release inhibitor, reduced the expression of IL‑6, TNF‑α, TLR4, TLR5 and phosphorylated‑p65, and upregulated that of IL‑10, TLR3 and p65 in peripheral blood mononuclear cells from patients with AS. Treatment of patients with infliximab, an anti‑TNF‑α monoclonal antibody, induced similar effects in vivo. In conclusion, it was revealed that inhibition of TNF‑α suppressed inflammatory responses in patients with AS, increased the expression of TLR3 and decreased NF‑κB signaling, and the expression of TLR4 and TLR5. The results indicated that TLRs and the NF‑κB signaling pathway were involved in the regulation of inflammatory responses in AS. These findings provided insight into the mechanisms underlying the development of AS and potential novel therapeutic approaches.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Orthopedics, Mingzhou Hospital of Zhejiang University, Ningbo, Zhejiang 315000, P.R. China
| | - Rongming Xu
- Department of Orthopedics, Mingzhou Hospital of Zhejiang University, Ningbo, Zhejiang 315000, P.R. China
| | - Lei Wu
- Department of Orthopedics, Mingzhou Hospital of Zhejiang University, Ningbo, Zhejiang 315000, P.R. China
| | - Jihong Jiang
- Department of Orthopedics, Mingzhou Hospital of Zhejiang University, Ningbo, Zhejiang 315000, P.R. China
| |
Collapse
|
53
|
Akhmetzyanova E, Kletenkov K, Mukhamedshina Y, Rizvanov A. Different Approaches to Modulation of Microglia Phenotypes After Spinal Cord Injury. Front Syst Neurosci 2019; 13:37. [PMID: 31507384 PMCID: PMC6718713 DOI: 10.3389/fnsys.2019.00037] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 07/29/2019] [Indexed: 01/04/2023] Open
Abstract
Microglial cells, which are highly plastic, immediately respond to any change in the microenvironment by becoming activated and shifting the phenotype toward neurotoxicity or neuroprotection. The polarization of microglia/macrophages after spinal cord injury (SCI) seems to be a dynamic process and can change depending on the microenvironment, stage, course, and severity of the posttraumatic process. Effective methods to modulate microglia toward a neuroprotective phenotype in order to stimulate neuroregeneration are actively sought for. In this context, available approaches that can selectively impact the polarization of microglia/macrophages regulate synthesis of trophic factors and cytokines/chemokines in them, and their phagocytic function and effects on the course and outcome of SCI are discussed in this review.
Collapse
Affiliation(s)
- Elvira Akhmetzyanova
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Konstantin Kletenkov
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Yana Mukhamedshina
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.,Department of Histology, Cytology and Embryology, Kazan State Medical University, Kazan, Russia
| | - Albert Rizvanov
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| |
Collapse
|
54
|
Morris G, Berk M, Maes M, Carvalho AF, Puri BK. Socioeconomic Deprivation, Adverse Childhood Experiences and Medical Disorders in Adulthood: Mechanisms and Associations. Mol Neurobiol 2019; 56:5866-5890. [PMID: 30685844 PMCID: PMC6614134 DOI: 10.1007/s12035-019-1498-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 01/15/2019] [Indexed: 12/30/2022]
Abstract
Severe socioeconomic deprivation (SED) and adverse childhood experiences (ACE) are significantly associated with the development in adulthood of (i) enhanced inflammatory status and/or hypothalamic-pituitary-adrenal (HPA) axis dysfunction and (ii) neurological, neuroprogressive, inflammatory and autoimmune diseases. The mechanisms by which these associations take place are detailed. The two sets of consequences are themselves strongly associated, with the first set likely contributing to the second. Mechanisms enabling bidirectional communication between the immune system and the brain are described, including complex signalling pathways facilitated by factors at the level of immune cells. Also detailed are mechanisms underpinning the association between SED, ACE and the genesis of peripheral inflammation, including epigenetic changes to immune system-related gene expression. The duration and magnitude of inflammatory responses can be influenced by genetic factors, including single nucleotide polymorphisms, and by epigenetic factors, whereby pro-inflammatory cytokines, reactive oxygen species, reactive nitrogen species and nuclear factor-κB affect gene DNA methylation and histone acetylation and also induce several microRNAs including miR-155, miR-181b-1 and miR-146a. Adult HPA axis activity is regulated by (i) genetic factors, such as glucocorticoid receptor polymorphisms; (ii) epigenetic factors affecting glucocorticoid receptor function or expression, including the methylation status of alternative promoter regions of NR3C1 and the methylation of FKBP5 and HSD11β2; (iii) chronic inflammation and chronic nitrosative and oxidative stress. Finally, it is shown how severe psychological stress adversely affects mitochondrial structure and functioning and is associated with changes in brain mitochondrial DNA copy number and transcription; mitochondria can act as couriers of childhood stress into adulthood.
Collapse
Affiliation(s)
- Gerwyn Morris
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Barwon Health, P.O. Box 291, Geelong, Victoria, Australia
| | - Michael Berk
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Barwon Health, P.O. Box 291, Geelong, Victoria, Australia
- Department of Psychiatry, Level 1 North, Main Block, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Kenneth Myer Building, 30 Royal Parade, Parkville, Victoria, Australia
- Orygen, The National Centre of Excellence in Youth Mental Health, 35 Poplar Rd, Parkville, Victoria, Australia
| | - Michael Maes
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Barwon Health, P.O. Box 291, Geelong, Victoria, Australia
- Department of Psychiatry, Chulalongkorn University, Bangkok, Thailand
| | - André F Carvalho
- Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Centre for Addiction & Mental Health (CAMH), Toronto, ON, Canada
| | - Basant K Puri
- Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK.
| |
Collapse
|
55
|
Singh G, Singh A, Singh P, Bhatti R. Bergapten Ameliorates Vincristine-Induced Peripheral Neuropathy by Inhibition of Inflammatory Cytokines and NFκB Signaling. ACS Chem Neurosci 2019; 10:3008-3017. [PMID: 31064179 DOI: 10.1021/acschemneuro.9b00206] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Bergapten, a furanocoumarin derivative found in a variety of medicinal plants, is documented to possess anti-inflammatory activity. However, whether bergapten is useful in alleviating the symptoms as well as the progress of peripheral neuropathy is not yet studied. The current investigation has been designed to explore the effect of bergapten on vincristine-induced neuropathic pain. Rats were grouped as normal, neuropathic control (vincristine), gabapentin, and bergapten treated groups with five animals in each group. Vincristine (100 μg/kg, i.p.) was administered for 10 days with 2 days break. Gabapentin (60 mg/kg, i.p.) and bergapten (10 mg/kg i.p.) treatments were given once daily for 14 days. The animals were assessed for hyperalgesia and allodynia. After 14 days, animals were sacrificed to detect plasma pro-inflammatory cytokines (TNF α, IL-1β), spinal cord, and sciatic nerve oxidative stress and expression of iNOS, COX-2, and NFkB in the spinal cord. There was a marked reduction in pain behaviors in the bergapten group as compared to the vincristine group. Bergapten also attenuated pro-inflammatory cytokines (TNFα and IL-1β), oxidative stress, and expression of NFkB, COX-2, and iNOS. Overall the current study concludes that bergapten could serve as a potential lead to drug development for the treatment of neuropathic pain.
Collapse
|
56
|
Wang J, Zhao Q. Kaempferitrin inhibits proliferation, induces apoptosis, and ameliorates inflammation in human rheumatoid arthritis fibroblast-like synoviocytes. Phytother Res 2019; 33:1726-1735. [PMID: 31155798 DOI: 10.1002/ptr.6364] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 03/12/2019] [Accepted: 03/16/2019] [Indexed: 12/31/2022]
Abstract
Rheumatoid arthritis (RA) is a complex chronic inflammatory disease that is associated with the aberrant activation of fibroblast-like synoviocytes (FLS). Kaempferitrin is a natural flavonoid glycoside that possesses anti-inflammatory bioactivity. However, the effect of kaempferitrin on RA has not yet been revealed. The aim of the present study was to investigate the effect of kaempferitrin on human RA-FLS MH7A cell line. We found that kaempferitrin inhibited proliferation and induced apoptosis of MH7A cells. Kaempferitrin decreased the levels of interleukin (IL)-1β, IL-6, tumor necrosis factor (TNF)-α, matrix metalloproteinase (MMP)-1, and MMP-3 in MH7A cells. Moreover, kaempferitrin blocked the activation of nuclear factor-κB (NF-κB) and protein kinase B (Akt)/mammalian target of rapamycin (mTOR) pathways. Furthermore, treatment with kaempferitrin decreased paw thickness and arthritis scores, and reduced the serum levels of IL-1β, IL-6, and TNF-α in a collagen-induced arthritis mouse model. In conclusion, kaempferitrin inhibited cell proliferation, induced cell apoptosis, and ameliorated inflammation of RA-FLS by suppressing the NF-κB and Akt/mTOR pathways.
Collapse
Affiliation(s)
- Jing Wang
- Department of Rheumatology and Immunology, Huaihe Hospital of Henan University, Kaifeng, Henan, China
| | - Qing Zhao
- Department of Rheumatology and Immunology, Huaihe Hospital of Henan University, Kaifeng, Henan, China
| |
Collapse
|
57
|
Ding Y, Zhao Q, Wang L. Pro-apoptotic and anti-inflammatory effects of araloside A on human rheumatoid arthritis fibroblast-like synoviocytes. Chem Biol Interact 2019; 306:131-137. [PMID: 31004595 DOI: 10.1016/j.cbi.2019.04.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 03/29/2019] [Accepted: 04/17/2019] [Indexed: 02/06/2023]
Abstract
Rheumatoid arthritis fibroblast-like synoviocytes play an essential role in the occurrence and progression of rheumatoid arthritis. As the main pharmacologically active components of Aralia taibaiensis, total saponins, particularly triterpenoid saponins, have been shown to possess multiple pharmacological activities including relieving rheumatism. However, the effect of araloside A, a triterpenoid saponin extracted from the root bark of Aralia taibaiensis, on rheumatoid arthritis remains unknown. Cell counting kit-8 assay was employed to determine cell viability. Flow cytometry analysis, caspase-3/7 activity assay and Western blot analysis of cytochrome c and B-cell lymphoma 2 were conducted to evaluate cell apoptosis. Inflammation was assessed by detecting the production of inflammatory cytokines including interleukin-6 and interleukin-8, as well as inflammatory mediators including nitric oxide and prostaglandin E2. The changes of the nuclear factor kappa B pathway were examined by Western blot. Results showed that araloside A concentration-dependently inhibited the proliferation of MH7A cells. Meanwhile, araloside A dose-dependently augmented the apoptotic rate and caspase-3/7 activity, increased cytochrome c level and decreased B-cell lymphoma 2 level in MH7A cells. Araloside A concentration-dependently curbed the production of interleukin-6, interleukin-8, prostaglandin E2 and nitric oxide in MH7A cells. In addition, we found that araloside A inhibited the nuclear factor kappa B pathway and inhibition of the nuclear factor kappa B pathway by BAY11-7082 and PDTC showed a similar role to araloside A in MH7A cells. Taken together, araloside A exerted pro-apoptotic and anti-inflammatory effects in rheumatoid arthritis fibroblast-like synoviocytes via inhibition of the nuclear factor kappa B pathway.
Collapse
Affiliation(s)
- Yanjie Ding
- Department of Rheumatology and Immunology, Huaihe Hospital of Henan University, Kaifeng, 475000, Henan, China
| | - Qing Zhao
- Department of Rheumatology and Immunology, Huaihe Hospital of Henan University, Kaifeng, 475000, Henan, China
| | - Laifang Wang
- Department of Rheumatology and Immunology, Huaihe Hospital of Henan University, Kaifeng, 475000, Henan, China.
| |
Collapse
|
58
|
Kucharzewska P, Maracle CX, Jeucken KCM, van Hamburg JP, Israelsson E, Furber M, Tas SW, Olsson HK. NIK-IKK complex interaction controls NF-κB-dependent inflammatory activation of endothelium in response to LTβR ligation. J Cell Sci 2019; 132:jcs225615. [PMID: 30837284 DOI: 10.1242/jcs.225615] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 02/14/2019] [Indexed: 12/26/2022] Open
Abstract
NF-κB-inducing kinase (NIK; also known as MAP3K14) is a central regulator of non-canonical NF-κB signaling in response to stimulation of TNF receptor superfamily members, such as the lymphotoxin-β receptor (LTβR), and is implicated in pathological angiogenesis associated with chronic inflammation and cancer. Here, we identify a previously unrecognized role of the LTβR-NIK axis during inflammatory activation of human endothelial cells (ECs). Engagement of LTβR-triggered canonical and non-canonical NF-κB signaling promoted expression of inflammatory mediators and adhesion molecules, and increased immune cell adhesion to ECs. Sustained LTβR-induced inflammatory activation of ECs was NIK dependent, but independent of p100, indicating that the non-canonical arm of NF-κB is not involved. Instead, prolonged activation of canonical NF-κB signaling, through the interaction of NIK with IκB kinase α and β (also known as CHUK and IKBKB, respectively), was required for the inflammatory response. Endothelial inflammatory activation induced by synovial fluid from rheumatoid arthritis patients was significantly reduced by NIK knockdown, suggesting that NIK-mediated alternative activation of canonical NF-κB signaling is a key driver of pathological inflammatory activation of ECs. Targeting NIK could thus provide a novel approach for treating chronic inflammatory diseases.
Collapse
Affiliation(s)
- Paulina Kucharzewska
- Respiratory, Inflammation and Autoimmunity IMED Biotech Unit, AstraZeneca, Gothenburg, SE-431 83 Mölndal, Sweden
| | - Chrissta X Maracle
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology and Rheumatology and Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Kim C M Jeucken
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology and Rheumatology and Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Jan Piet van Hamburg
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology and Rheumatology and Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Elisabeth Israelsson
- Respiratory, Inflammation and Autoimmunity IMED Biotech Unit, AstraZeneca, Gothenburg, SE-431 83 Mölndal, Sweden
| | - Mark Furber
- Respiratory, Inflammation and Autoimmunity IMED Biotech Unit, AstraZeneca, Gothenburg, SE-431 83 Mölndal, Sweden
| | - Sander W Tas
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology and Rheumatology and Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Henric K Olsson
- Respiratory, Inflammation and Autoimmunity IMED Biotech Unit, AstraZeneca, Gothenburg, SE-431 83 Mölndal, Sweden
| |
Collapse
|
59
|
Wang KZ, Feng L, Jiang WD, Wu P, Liu Y, Jiang J, Kuang SY, Tang L, Zhang YA, Zhou XQ. Dietary gossypol reduced intestinal immunity and aggravated inflammation in on-growing grass carp (Ctenopharyngodon idella). FISH & SHELLFISH IMMUNOLOGY 2019; 86:814-831. [PMID: 30543935 DOI: 10.1016/j.fsi.2018.12.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 11/25/2018] [Accepted: 12/09/2018] [Indexed: 06/09/2023]
Abstract
The present study explored the effects of dietary gossypol on the gut health of on-growing grass carp. The fish were fed six diets containing different levels of free gossypol (0, 121.38, 243.94, 363.89, 759.93 and 1162.06 mg/kg diet) from gossypol-acetic acid for 60 days and then challenged with Aeromonas hydrophila for 14 days. The results showed that dietary gossypol (1) could aggravate enteritis and damage the structure of intestinal epithelial cells, (2) decreased the lysozyme (LZ) and Acid phosphatase (ACP) activities, complement 3 (C3), C4 and immunoglobulin M (IgM) contents, and it down-regulated the Hepcidin (rather than distal intestine (DI)), immunoglobulin Z (IgZ), liver-expressed antimicrobial peptide (LEAP)-2B, Mucin2 and β-defensin-1 mRNA levels in the proximal intestine (PI), mid intestine (MI) and DI, (3) up-regulated intestinal pro-inflammatory cytokines tumor necrosis factor α (TNF-α), interferon γ2 (IFN-γ2), interleukin 1β (IL-1β), IL-6 (only in PI), IL-8 and IL-12p35 mRNA levels partly related to nuclear factor kappa B (NF-κB) signalling, and (4) down-regulated the mRNA levels of anti-inflammatory cytokines such as transforming growth factor (TGF)-β1, TGF-β2, interleukin 4/13A (IL-4/13A) (except IL-4/13B), IL-10 and IL-11 partly relating to target of rapamycin (TOR) signalling in the intestines of on-growing grass carp. Moreover, the dietary gossypol had no impact on the LEAP-2A, IL-12P40, IL-17D, IL-10, NF-κBp52, IKKα and eIF4E-binding proteins 2 (4E-BP2) mRNA levels in the intestines. Finally, based on the intestinal histopathological results, enteritis morbidity, LZ activity and IgM content, the safe dose of gossypol in the diets for on-growing grass carp should be less than 103.42 mg/kg diet.
Collapse
Affiliation(s)
- Kai-Zhuo Wang
- Animal Nutrition Institute, Sichuan Agricultural University, Sichuan, Chengdu, 611130, China
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Sichuan, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, Chengdu, 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan, Agricultural University, Sichuan, Chengdu, 611130, China
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Sichuan, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, Chengdu, 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan, Agricultural University, Sichuan, Chengdu, 611130, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Sichuan, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, Chengdu, 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan, Agricultural University, Sichuan, Chengdu, 611130, China
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Sichuan, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, Chengdu, 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan, Agricultural University, Sichuan, Chengdu, 611130, China
| | - Jun Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Sichuan, Chengdu, 611130, China
| | - Sheng-Yao Kuang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu, 610066, China
| | - Ling Tang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu, 610066, China
| | - Yong-An Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Sichuan, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, Chengdu, 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan, Agricultural University, Sichuan, Chengdu, 611130, China.
| |
Collapse
|
60
|
Cavalli M, Baltzer N, Umer HM, Grau J, Lemnian I, Pan G, Wallerman O, Spalinskas R, Sahlén P, Grosse I, Komorowski J, Wadelius C. Allele specific chromatin signals, 3D interactions, and motif predictions for immune and B cell related diseases. Sci Rep 2019; 9:2695. [PMID: 30804403 PMCID: PMC6389883 DOI: 10.1038/s41598-019-39633-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 01/24/2019] [Indexed: 12/20/2022] Open
Abstract
Several Genome Wide Association Studies (GWAS) have reported variants associated to immune diseases. However, the identified variants are rarely the drivers of the associations and the molecular mechanisms behind the genetic contributions remain poorly understood. ChIP-seq data for TFs and histone modifications provide snapshots of protein-DNA interactions allowing the identification of heterozygous SNPs showing significant allele specific signals (AS-SNPs). AS-SNPs can change a TF binding site resulting in altered gene regulation and are primary candidates to explain associations observed in GWAS and expression studies. We identified 17,293 unique AS-SNPs across 7 lymphoblastoid cell lines. In this set of cell lines we interrogated 85% of common genetic variants in the population for potential regulatory effect and we identified 237 AS-SNPs associated to immune GWAS traits and 714 to gene expression in B cells. To elucidate possible regulatory mechanisms we integrated long-range 3D interactions data to identify putative target genes and motif predictions to identify TFs whose binding may be affected by AS-SNPs yielding a collection of 173 AS-SNPs associated to gene expression and 60 to B cell related traits. We present a systems strategy to find functional gene regulatory variants, the TFs that bind differentially between alleles and novel strategies to detect the regulated genes.
Collapse
Affiliation(s)
- Marco Cavalli
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Nicholas Baltzer
- Department of Cell and Molecular Biology, Computational Biology and Bioinformatics, Uppsala University, Uppsala, Sweden
| | - Husen M Umer
- Department of Cell and Molecular Biology, Computational Biology and Bioinformatics, Uppsala University, Uppsala, Sweden
| | - Jan Grau
- Institute of Computer Science, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Ioana Lemnian
- Institute of Computer Science, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Gang Pan
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Ola Wallerman
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Rapolas Spalinskas
- Science for Life Laboratory, Division of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Pelin Sahlén
- Science for Life Laboratory, Division of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Ivo Grosse
- Institute of Computer Science, Martin Luther University Halle-Wittenberg, Halle, Germany.,German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, Leipzig, Germany
| | - Jan Komorowski
- Department of Cell and Molecular Biology, Computational Biology and Bioinformatics, Uppsala University, Uppsala, Sweden.,Institute of Computer Science, Polish Academy of Sciences, Warszawa, Poland
| | - Claes Wadelius
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
61
|
Zhang N, Zhang S, Xu C, Fu L, Liu T, Zhao Y. Retracted: Decoy Oligodeoxynucleotides, Polysaccharides, and Targeted Peptide‐Functionalized Gold Nanorods for the Combined Treatment of Rheumatoid Arthritis. Adv Healthc Mater 2018; 7:e1800982. [DOI: 10.1002/adhm.201800982] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 10/24/2018] [Indexed: 01/04/2023]
Affiliation(s)
- Nan Zhang
- Department of PharmaceuticsSchool of Pharmaceutical SciencesZhengzhou University Zhengzhou HeNan 450001 P. R. China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesHeNan Province Zhengzhou HeNan 450001 P. R. China
- Key Laboratory of Advanced Pharmaceutical TechnologyMinistry of Education of ChinaHeNan Province Zhengzhou HeNan 450001 P. R. China
| | - Shasha Zhang
- Department of PharmaceuticsSchool of Pharmaceutical SciencesZhengzhou University Zhengzhou HeNan 450001 P. R. China
| | - Chunyu Xu
- Department of PharmaceuticsSchool of Pharmaceutical SciencesZhengzhou University Zhengzhou HeNan 450001 P. R. China
| | - Lingling Fu
- Department of PharmaceuticsSchool of Pharmaceutical SciencesZhengzhou University Zhengzhou HeNan 450001 P. R. China
| | - Tuanbing Liu
- Department of PharmaceuticsSchool of Pharmaceutical SciencesZhengzhou University Zhengzhou HeNan 450001 P. R. China
| | - Yongxing Zhao
- Department of PharmaceuticsSchool of Pharmaceutical SciencesZhengzhou University Zhengzhou HeNan 450001 P. R. China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesHeNan Province Zhengzhou HeNan 450001 P. R. China
- Key Laboratory of Advanced Pharmaceutical TechnologyMinistry of Education of ChinaHeNan Province Zhengzhou HeNan 450001 P. R. China
| |
Collapse
|
62
|
Chen W, Yi C, Jin L. The Role of Nicotinamide Adenine Dinucleotide in the Pathogenesis of Rheumatoid Arthritis: Potential Implications for Treatment. EUROPEAN MEDICAL JOURNAL 2018. [DOI: 10.33590/emj/10312205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic, systemic, inflammatory, autoimmune disease characterised by small joint swelling, deformity, and dysfunction. Its exact aetiology is unclear. Current treatment approaches do not control harmful autoimmune attacks or prevent irreversible damage without considerable side effects. Nicotinamide adenine dinucleotide (NAD+), an important hydrogen carrier in mitochondrial respiration and oxidative phosphorylation, is the major determinant of redox state in the cell. NAD+ metabolites act as degradation substrates for a wide range of enzymes, such as sirtuins, poly-ADP-ribose polymerases, ADP-ribosyltransferases, and CD38. The roles of NAD+ have expanded beyond its role as a coenzyme, linking cellular metabolism to inflammation signalling and immune response. The aim of this review is to illustrate the role of NAD+-related enzymes in the pathogenesis of RA and highlight the potential therapeutic role of NAD+ in RA.
Collapse
Affiliation(s)
- Weiqian Chen
- Department of Rheumatology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Caihong Yi
- Department of Rheumatology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Lin Jin
- Department of Rheumatology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
63
|
Seleznik GM, Reding T, Peter L, Gupta A, Steiner SG, Sonda S, Verbeke CS, Dejardin E, Khatkov I, Segerer S, Heikenwalder M, Graf R. Development of autoimmune pancreatitis is independent of CDKN1A/p21-mediated pancreatic inflammation. Gut 2018; 67:1663-1673. [PMID: 28774888 DOI: 10.1136/gutjnl-2016-313458] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 06/07/2017] [Accepted: 06/19/2017] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Chronic pancreatitis (CP) and autoimmune pancreatitis (AIP) are characterised by different inflammatory processes. If pancreatic inflammation is a prerequisite for autoimmunity is still unclear. AIP is considered mostly a T cell-mediated disease; however, in induction of CP, macrophages play a pivotal role. p21-a member of cyclin-dependent kinase inhibitors-can influence inflammatory processes, in particular can regulate T cell activation and promote macrophage development. We therefore examined the role of p21-mediated inflammation in AIP. DESIGN We intercrossed lymphotoxin (LT) overexpressing mice (Tg(Ela1-LTa,b))-a model to study AIP development-with p21-deficient mice. Furthermore, we characterised p21 expression in human AIP and non-AIP specimens. RESULTS p21 deficiency in LT mice (LTp21-/-) prevented early pancreatic injury and reduced inflammation. In acinar cells, diminished proliferation and abrogated activation of non-canonical nuclear factor kappa-light-chain-enhancer of activated B cell (NF-κB) pathway was observed. In contrast, 12-month-old LT mice with and without p21 had similar inflammatory signatures and T-B cell infiltration. Interestingly, LT and LTp21-/- mice had comparable tertiary lymphoid organs (TLOs), autoantibodies and elevated IgG levels. However, acinar cell proliferation, acinar-to-ductal metaplasia and acinar non-canonical NF-κB pathway activation remained impaired in LTp21-/- pancreata. CONCLUSIONS Our findings indicate that p21 is crucial for pancreatic inflammation in LT-driven pancreatic injury. p21 is involved in early acinar secretion of inflammatory mediators that attract innate immune cells. However, p21 is not essential for humoral immune response, accountable for autoimmunity. Remarkably, p21 renders acinar cells less susceptible to proliferation and transdifferentiation. We therefore suggest that AIP can also develop independent of chronic inflammatory processes.
Collapse
Affiliation(s)
- Gitta M Seleznik
- Department of Visceral and Transplantation Surgery, Swiss HPB Centre, University Hospital Zurich, Zurich, Switzerland
| | - Theresia Reding
- Department of Visceral and Transplantation Surgery, Swiss HPB Centre, University Hospital Zurich, Zurich, Switzerland
| | - Lukas Peter
- Department of Visceral and Transplantation Surgery, Swiss HPB Centre, University Hospital Zurich, Zurich, Switzerland
| | - Anurag Gupta
- Department of Visceral and Transplantation Surgery, Swiss HPB Centre, University Hospital Zurich, Zurich, Switzerland
| | - Sabrina G Steiner
- Department of Visceral and Transplantation Surgery, Swiss HPB Centre, University Hospital Zurich, Zurich, Switzerland
| | - Sabrina Sonda
- Department of Visceral and Transplantation Surgery, Swiss HPB Centre, University Hospital Zurich, Zurich, Switzerland
| | - Caroline S Verbeke
- Department of Pathology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Emmanuel Dejardin
- Laboratory of Molecular Immunology and Signal Transduction, GIGA-Research, University of Liège, Liège, Belgium
| | - Igor Khatkov
- Department of Medicine and Dentistry, Moscow State University, Moscow, Russia
| | - Stephan Segerer
- Division of Nephrology, University Hospital, Zurich, Switzerland.,Division of Nephrology, Dialysis and Transplantation, Kantonsspital Aarau, Aarau, Switzerland
| | - Mathias Heikenwalder
- School of Medicine, Institute of Virology, TUM-Helmholtz Zentrum Munich, Munich, Germany.,Department of Chronic Inflammation and Cancer, German Cancer Center (DKFZ), Heidelberg, Germany
| | - Rolf Graf
- Department of Visceral and Transplantation Surgery, Swiss HPB Centre, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
64
|
Dong H, Liu F, Ma F, Xu L, Pang L, Li X, Liu B, Wang L. Montelukast inhibits inflammatory response in rheumatoid arthritis fibroblast-like synoviocytes. Int Immunopharmacol 2018; 61:215-221. [PMID: 29890415 DOI: 10.1016/j.intimp.2018.04.042] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 04/22/2018] [Accepted: 04/24/2018] [Indexed: 02/08/2023]
Abstract
Montelukast, a potent selective antagonist of cysteinyl leukotriene (cysLT) receptors, has displayed its important anti-oxidative and anti-inflammatory effects in various tissues and organs. Rheumatoid arthritis (RA) is an immune disease defined by hyperplastic synovitis and joint destruction. Fibroblast-like synoviocytes in RA (RA-FLSs) are the main cell component of the hyperplastic synovium. Whether montelukast can protect against the inflammatory milieu of RA remains unclear. Here, it is shown that cysLT1R is present in FLSs and unregulated in RA-FLSs. Montelukast has an inhibitory effect on the inflammatory microenvironment of RA by decreasing the secretion of IL-6, IL-8, MMP-3 and MMP-13 in FLSs induced by IL-1β. Notably, treatment with montelukast attenuated IL-1β-induced phosphorylation of IκBα, IκBα degradation, nuclear translocation of p65 and NF-κB activity to express a luciferase reporter gene in FLSs. The findings of the current study provide evidence for a novel therapeutic strategy for RA using montelukast.
Collapse
Affiliation(s)
- Hongyu Dong
- Department of Rheumatology and Immunology, Shijingshan teaching hospital of Capital Medical University, Beijing Shijingshan Hospital, Beijing 100043, China
| | - Feng Liu
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China.
| | - Fengyun Ma
- Department of Rheumatology and Immunology, Shijingshan teaching hospital of Capital Medical University, Beijing Shijingshan Hospital, Beijing 100043, China
| | - Lianna Xu
- Department of Rheumatology and Immunology, Shijingshan teaching hospital of Capital Medical University, Beijing Shijingshan Hospital, Beijing 100043, China
| | - Linna Pang
- Department of Rheumatology and Immunology, Shijingshan teaching hospital of Capital Medical University, Beijing Shijingshan Hospital, Beijing 100043, China
| | - Xuyan Li
- Department of Rheumatology and Immunology, Shijingshan teaching hospital of Capital Medical University, Beijing Shijingshan Hospital, Beijing 100043, China
| | - Bo Liu
- Department of Rheumatology and Immunology, Shijingshan teaching hospital of Capital Medical University, Beijing Shijingshan Hospital, Beijing 100043, China
| | - Liang Wang
- Center of Orthopedics, The 309th Hospital of PLA, Beijing 100091, China
| |
Collapse
|
65
|
Xia ZB, Meng FR, Fang YX, Wu X, Zhang CW, Liu Y, Liu D, Li GQ, Feng FB, Qiu HY. Inhibition of NF-κB signaling pathway induces apoptosis and suppresses proliferation and angiogenesis of human fibroblast-like synovial cells in rheumatoid arthritis. Medicine (Baltimore) 2018; 97:e10920. [PMID: 29879032 PMCID: PMC5999456 DOI: 10.1097/md.0000000000010920] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is the most common inflammatory arthritis and is a major cause of disability. The nuclear factor-kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathway has been reported to be involved in the pathogenesis of RA with unclear mechanisms. Therefore, this study aims to explore the effect of NF-κB pathway on proliferation, apoptosis, and angiogenesis of human fibroblast-like synovial cells (HFLS) in RA. METHODS Normal HFLS and RA-HFLS were selected as the normal and control groups, respectively. RA-HFLS were treated by BAY11-7082 (an inhibitor of NF-κB) in different concentrations, namely 2.5 μmol/L BAY11-7082, 5 μmol/LBAY11-7082 and 10 μmol/L BAY11-7082. MTT assay was employed to detect cell proliferation. Cell apoptosis was determined by flow cytometry at 24, 48, and 72 hours after culture. Western blot analysis was employed to detect the expressions of NF-κB, angiogenesis-related factors (VEGF, Ang1, and Ang2). RESULTS Initially, we found that BAY11-7082 inhibited NF-κB expression in a concentration-dependent manner. According to the findings of MTT assay and flow cytometry, we understood that RA-HFLS treated by BAY11-7082 (an inhibitor of NF-κB), the inhibition of NF-κB pathway, suppressed RA-HFLS proliferation and induced RA-HFLS apoptosis in a concentration and time-dependent manner. Furthermore, RA-HFLS treated by BAY11-7082 presented decreased VEGF, Ang1 and Ang2 expressions in a concentration-dependent manner. CONCLUSION The study concluded that inhibition of NF-κB pathway induced cell apoptosis and suppressed proliferation and angiogenesis of RA-HFLS, which could serve as a novel target in the treatment of RA.
Collapse
Affiliation(s)
- Zhong-Bin Xia
- Department of Rheumatology, Clinical Medical College of Yangzhou University (Northern Jiangsu People's Hospital Affiliated to Yangzhou University), Yangzhou
| | - Fan-Ru Meng
- Department of Rheumatology, Clinical Medical College of Yangzhou University (Northern Jiangsu People's Hospital Affiliated to Yangzhou University), Yangzhou
- Clinical Medical College, Dalian Medical University, Dalian
| | - Yu-Xuan Fang
- Department of Rheumatology, Clinical Medical College of Yangzhou University (Northern Jiangsu People's Hospital Affiliated to Yangzhou University), Yangzhou
- Clinical Medical College, Dalian Medical University, Dalian
| | - Xia Wu
- Department of Rheumatology, Clinical Medical College of Yangzhou University (Northern Jiangsu People's Hospital Affiliated to Yangzhou University), Yangzhou
- Clinical Medical College, Dalian Medical University, Dalian
| | - Chun-Wang Zhang
- Department of Rheumatology, Clinical Medical College of Yangzhou University (Northern Jiangsu People's Hospital Affiliated to Yangzhou University), Yangzhou
- Clinical Medical College, Dalian Medical University, Dalian
| | - Ying Liu
- Department of Rheumatology, Clinical Medical College of Yangzhou University (Northern Jiangsu People's Hospital Affiliated to Yangzhou University), Yangzhou
- Clinical Medical College, Dalian Medical University, Dalian
| | - Dan Liu
- Department of Pathology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, PR China
| | - Guo-Qing Li
- Department of Rheumatology, Clinical Medical College of Yangzhou University (Northern Jiangsu People's Hospital Affiliated to Yangzhou University), Yangzhou
| | - Fan-Bo Feng
- Department of Rheumatology, Clinical Medical College of Yangzhou University (Northern Jiangsu People's Hospital Affiliated to Yangzhou University), Yangzhou
| | - Hai-Yang Qiu
- Department of Rheumatology, Clinical Medical College of Yangzhou University (Northern Jiangsu People's Hospital Affiliated to Yangzhou University), Yangzhou
| |
Collapse
|
66
|
Singh AK, Kumar S, Vinayak M. Recent development in antihyperalgesic effect of phytochemicals: anti-inflammatory and neuro-modulatory actions. Inflamm Res 2018; 67:633-654. [PMID: 29767332 DOI: 10.1007/s00011-018-1156-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 05/04/2018] [Accepted: 05/08/2018] [Indexed: 02/08/2023] Open
Abstract
INTRODUCTION Pain is an unpleasant sensation triggered by noxious stimulation. It is one of the most prevalent conditions, limiting productivity and diminishing quality of life. Non steroidal anti inflammatory drugs (NSAIDs) are widely used as pain relievers in present day practice as pain is mostly initiated due to inflammation. However, due to potentially serious side effects, long term use of these antihyperalgesic drugs raises concern. Therefore there is a demand to search novel medicines with least side effects. Herbal products have been used for centuries to reduce pain and inflammation, and phytochemicals are known to cause fewer side effects. However, identification of active phytochemicals of herbal medicines and clear understanding of the molecular mechanism of their action is needed for clinical acceptance. MATERIALS AND METHODS In this review, we have briefly discussed the cellular and molecular changes during hyperalgesia via inflammatory mediators and neuro-modulatory action involved therein. The review includes 54 recently reported phytochemicals with antihyperalgesic action, as per the literature available with PubMed, Google Scholar and Scopus. CONCLUSION Compounds of high interest as potential antihyperalgesic agents are: curcumin, resveratrol, capsaicin, quercetin, eugenol, naringenin and epigallocatechin gallate (EGCG). Current knowledge about molecular targets of pain and their regulation by these phytochemicals is elaborated and the scope of further research is discussed.
Collapse
Affiliation(s)
- Ajeet Kumar Singh
- Department of Zoology, Biochemistry and Molecular Biology Laboratory, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.,Department of Zoology, CMP Degree College, University of Allahabad, Allahabad, 211002, India
| | - Sanjay Kumar
- Department of Zoology, Biochemistry and Molecular Biology Laboratory, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Manjula Vinayak
- Department of Zoology, Biochemistry and Molecular Biology Laboratory, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
67
|
Sarmiento Salinas FL, Santillán Benítez JG, Hernández Navarro MD, Mendieta Zerón H. NF-κB1/IKKε Gene Expression and Clinical Activity in Patients With Rheumatoid Arthritis. Lab Med 2018; 49:11-17. [PMID: 29069487 DOI: 10.1093/labmed/lmx033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Objective Rheumatoid arthritis (RA) is a systemic and autoimmune disorder whose primary characteristic is the chronic inflammation of joints. The objective of this study was to evaluate whether there was an association between nuclear factor kappa beta1/IKK epsilon (NF-κB1/IKKε) gene expression and clinical activity in RA. Methods Sixty patients with RA were included in the study: 30 with clinical activity and 30 with clinical remission. NF-κB1/IKKε gene expression was performed by real-time quantitative polymerase chain reaction through relative quantification with Taqman probes. A ROC curve for NF-κB1 and IKKε was also constructed. Results There were significant differences in NF-κB1 and IKKε gene expression (P ≤ .001 and P ≤ .029, respectively) between RA patients with clinical activity and clinical remission. The multivariate lineal general model showed that the use of nonsteroidal anti-inflammatory drugs influenced the NF-κB1 (P = .046) and IKKε (P = .005) expression. The ROC curves for the event "clinical activity" showed the greater area under the curve for NF-κB1 (0.827, 95% CI 0.717-0.937), P ≤ .001. Conclusion Although the use of NSAIDs influences the NF-κB1/IKKε pathway, the IKKε expression might be a useful laboratorial analysis to evaluate the RA clinical activity.
Collapse
Affiliation(s)
| | - Jonnathan Guadalupe Santillán Benítez
- Laboratory of Toxicology, Faculty of Chemistry, Autonomous University of the State of Mexico, Toluca, Mexico.,ISSEMyM Medical Center, Metepec, Mexico
| | | | - Hugo Mendieta Zerón
- Faculty of Medicine, Autonomous University of the State of Mexico, Toluca, Mexico.,Asociación Científica Latina A.C., Mexico
| |
Collapse
|
68
|
Xu XX, Bi JP, Ping L, Li P, Li F. A network pharmacology approach to determine the synergetic mechanisms of herb couple for treating rheumatic arthritis. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:967-979. [PMID: 29731604 PMCID: PMC5923250 DOI: 10.2147/dddt.s161904] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Purpose The purpose of this study was to investigate the therapeutic mechanism(s) of Clematis chinensis Osbeck/Notopterygium incisum K.C. Ting ex H.T (CN). Methods A network pharmacology approach integrating prediction of ingredients, target exploration, network construction, module partition and pathway analysis was used. Results This approach successfully helped to identify 12 active ingredients of CN, interacting with 13 key targets (Akt1, STAT3, TNFsf13, TP53, EPHB2, IL-10, IL-6, TNF, MAPK8, IL-8, RELA, ROS1 and STAT4). Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis indicated that CN-regulated pathways were mainly classified into signal transduction and immune system. Conclusion The present work may help to illustrate the mechanism(s) of action of CN, and it may provide a better understanding of antirheumatic effects.
Collapse
Affiliation(s)
- Xi-Xi Xu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Jian-Ping Bi
- Orthopedics Department, Shandong Provincial Traditional Chinese Medical Hospital, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Li Ping
- Center for Drug Safety Evaluation and Research, Zhejiang University, Hangzhou, People's Republic of China
| | - Ping Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Fei Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, People's Republic of China.,School of Pharmacy, Xinjiang Medical University, Urumqi, People's Republic of China
| |
Collapse
|
69
|
Pawlowska E, Szczepanska J, Wisniewski K, Tokarz P, Jaskólski DJ, Blasiak J. NF-κB-Mediated Inflammation in the Pathogenesis of Intracranial Aneurysm and Subarachnoid Hemorrhage. Does Autophagy Play a Role? Int J Mol Sci 2018; 19:E1245. [PMID: 29671828 PMCID: PMC5979412 DOI: 10.3390/ijms19041245] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 04/13/2018] [Accepted: 04/17/2018] [Indexed: 12/12/2022] Open
Abstract
The rupture of saccular intracranial aneurysms (IA) is the commonest cause of non-traumatic subarachnoid hemorrhage (SAH)—the most serious form of stroke with a high mortality rate. Aneurysm walls are usually characterized by an active inflammatory response, and NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) has been identified as the main transcription factor regulating the induction of inflammation-related genes in IA lesions. This transcription factor has also been related to IA rupture and resulting SAH. We and others have shown that autophagy interacts with inflammation in many diseases, but there is no information of such interplay in IA. Moreover, NF-κB, which is a pivotal factor controlling inflammation, is regulated by autophagy-related proteins, and autophagy is regulated by NF-κB signaling. It was also shown that autophagy mediates the normal functioning of vessels, so its disturbance can be associated with vessel-related disorders. Early brain injury, delayed brain injury, and associated cerebral vasospasm are among the most serious consequences of IA rupture and are associated with impaired function of the autophagy⁻lysosomal system. Further studies on the role of the interplay between autophagy and NF-κB-mediated inflammation in IA can help to better understand IA pathogenesis and to identify IA patients with an increased SAH risk.
Collapse
Affiliation(s)
- Elzbieta Pawlowska
- Department of Orthodontics, Medical University of Lodz, 92-216 Lodz, Poland.
| | - Joanna Szczepanska
- Department of Pediatric Dentistry, Medical University of Lodz, 92-216 Lodz, Poland.
| | - Karol Wisniewski
- Department of Neurosurgery and Neurooncology, Medical University of Lodz, Barlicki University Hospital, Kopcinskiego 22, 90-153 Lodz, Poland.
| | - Paulina Tokarz
- Department of Molecular Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland.
| | - Dariusz J Jaskólski
- Department of Neurosurgery and Neurooncology, Medical University of Lodz, Barlicki University Hospital, Kopcinskiego 22, 90-153 Lodz, Poland.
| | - Janusz Blasiak
- Department of Molecular Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland.
| |
Collapse
|
70
|
Targeting IκappaB kinases for cancer therapy. Semin Cancer Biol 2018; 56:12-24. [PMID: 29486318 DOI: 10.1016/j.semcancer.2018.02.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 02/18/2018] [Accepted: 02/21/2018] [Indexed: 01/05/2023]
Abstract
The inhibitory kappa B kinases (IKKs) and IKK related kinases are crucial regulators of the pro-inflammatory transcription factor, nuclear factor kappa B (NF-κB). The dysregulation in the activities of these kinases has been reported in several cancer types. These kinases are known to regulate survival, proliferation, invasion, angiogenesis, and metastasis of cancer cells. Thus, IKK and IKK related kinases have emerged as an attractive target for the development of cancer therapeutics. Several IKK inhibitors have been developed, few of which have advanced to the clinic. These inhibitors target IKK either directly or indirectly by modulating the activities of other signaling molecules. Some inhibitors suppress IKK activity by disrupting the protein-protein interaction in the IKK complex. The inhibition of IKK has also been shown to enhance the efficacy of conventional chemotherapeutic agents. Because IKK and NF-κB are the key components of innate immunity, suppressing IKK is associated with the risk of immune suppression. Furthermore, IKK inhibitors may hit other signaling molecules and thus may produce off-target effects. Recent studies suggest that multiple cytoplasmic and nuclear proteins distinct from NF-κB and inhibitory κB are also substrates of IKK. In this review, we discuss the utility of IKK inhibitors for cancer therapy. The limitations associated with the intervention of IKK are also discussed.
Collapse
|
71
|
Bao Y, Meng X, Liu F, Wang F, Yang J, Wang H, Xie G. Protective effects of osthole against inflammation induced by lipopolysaccharide in BV2 cells. Mol Med Rep 2018; 17:4561-4566. [DOI: 10.3892/mmr.2018.8447] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 12/01/2017] [Indexed: 11/05/2022] Open
Affiliation(s)
- Yuxin Bao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, P.R. China
| | - Xiaolin Meng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, P.R. China
| | - Fangning Liu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, P.R. China
| | - Fei Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, P.R. China
| | - Jinhui Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, P.R. China
| | - Haiyu Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, P.R. China
| | - Guanghong Xie
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, P.R. China
| |
Collapse
|
72
|
Interleukin-1β and tumor necrosis factor-α augment acidosis-induced rat articular chondrocyte apoptosis via nuclear factor-kappaB-dependent upregulation of ASIC1a channel. Biochim Biophys Acta Mol Basis Dis 2018; 1864:162-177. [DOI: 10.1016/j.bbadis.2017.10.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 09/09/2017] [Accepted: 10/02/2017] [Indexed: 02/07/2023]
|
73
|
Liu XZ, Fan J, Qi K, Liu SP, Xu WD, Gao Y, Gu XD, Li J, Bai CG, Shi YQ, Zhang LL, Zhao DB. Dishevelled2 promotes apoptosis and inhibits inflammatory cytokine secretion in rheumatoid arthritis fibroblast-like synoviocytes through crosstalk with the NF-κB pathway. Oncotarget 2017; 8:12649-12663. [PMID: 28187436 PMCID: PMC5355042 DOI: 10.18632/oncotarget.15172] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 01/24/2017] [Indexed: 12/13/2022] Open
Abstract
Dishevelled (Dvl) not only links the canonical Wnt and non-canonical Wnt pathways but can also crosstalk with other pathways. As there is no systematic study to date on Dvl in rheumatoid arthritis (RA), we explored the impact of Dvl2 on proliferation and inflammatory cytokine secretion in RA fibroblast-like synoviocytes (FLSs). Expression of Dvl2 in RA synovial tissue and RA-FLSs was measured. Dvl2 was overexpressed in collagen-induced arthritis rats and human RA-FLSs,. the apoptosis and secretion of inflammatory cytokines were observed. Genetic changes and corresponding mechanisms caused by overexpressing Dvl2 in RA-FLSs were assessed. Dvl2 was found to be overexpressed in RA synovial tissue and RA-FLSs. Overexpression of Dvl2 increased apoptosis and inhibited inflammatory cytokine secretion by RA-FLSs in vivo and in vitro, and Dvl2 inhibited expression of anti-apoptotic and inflammatory genes. One possible mechanism is that Dvl2 decreases the nuclear translocation of P65 and inhibits its ability to bind to the promoters of NF-κB target genes. Our findings reveal an underappreciated role of Dvl2 in regulating inflammation and RA-FLS apoptosis and provide insight into crosstalk between the Wnt and nuclear factor-κB (NF-κB) pathways.
Collapse
Affiliation(s)
- Xing Zhen Liu
- Department of Rheumatology and Immunology, Changhai Hospital, The Second Military Medical University, Shanghai, China.,Army Convalescence Area, Hangzhou Sanatorium of People's Liberation Army, Hangzhou, China
| | - Jie Fan
- Army Convalescence Area, Hangzhou Sanatorium of People's Liberation Army, Hangzhou, China
| | - Ke Qi
- Department of Joint Surgery, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Shu Peng Liu
- Experimental Center, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Wei Dong Xu
- Department of Joint Surgery, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Ying Gao
- Department of Rheumatology and Immunology, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Xiao Dan Gu
- Department of Rheumatology and Immunology, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Jia Li
- Department of Joint Surgery, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Chen Guang Bai
- Department of Pathology, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Ye Qing Shi
- Department of Rheumatology and Immunology, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Lan Ling Zhang
- Department of Rheumatology and Immunology, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Dong Bao Zhao
- Department of Rheumatology and Immunology, Changhai Hospital, The Second Military Medical University, Shanghai, China
| |
Collapse
|
74
|
Le Rossignol S, Ketheesan N, Haleagrahara N. Redox-sensitive transcription factors play a significant role in the development of rheumatoid arthritis. Int Rev Immunol 2017; 37:129-143. [PMID: 28898138 DOI: 10.1080/08830185.2017.1363198] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease which is associated with significant morbidity. Redox sensitive transcription factors including NF-κB, HIF, AP-1, and Nrf2 are intimately involved in the pathogenesis of RA. The treatment of this disease is limited by the elusive nature of the pathogenesis of RA. NF-κB is crucial for the maturation of immune cells as well as production of TNFα and MMPs, which escalate RA. HIF is essential for activation of inflammatory cells, angiogenesis and pannus formation in RA. AP-1 regulates cytokine and MMP production as well as synovial hyperplasia which are key processes in RA. Nrf2 is involved with chondrogenesis, osteoblastogenesis, prostaglandin secretion and ROS production in RA. Targeting two or more of these transcription factors may result in increased efficacy than either therapy in isolation. This review will highlight the control specific mediators on these transcription factors, the subsequent effect of these transcription factors once activated, and then mesh this with the pathogenesis of RA. The elucidation of key transcription factor regulation in the pathogenesis of RA may highlight the novel therapy interventions which may prove to have a greater efficacy than those therapies currently available.
Collapse
Affiliation(s)
- Scott Le Rossignol
- a College of Medicine and Dentistry , James Cook University Townsville , Queensland , Australia
| | - Natkunam Ketheesan
- b Biomedicine, College of Public Health, Medical and Veterinary Sciences , James Cook University , Townsville , Queensland , Australia.,c Australian Institute of Tropical Health and Medicine , James Cook University , Townsville , Queensland , Australia
| | - Nagaraja Haleagrahara
- b Biomedicine, College of Public Health, Medical and Veterinary Sciences , James Cook University , Townsville , Queensland , Australia.,c Australian Institute of Tropical Health and Medicine , James Cook University , Townsville , Queensland , Australia
| |
Collapse
|
75
|
The non-canonical NF-κB pathway in immunity and inflammation. NATURE REVIEWS. IMMUNOLOGY 2017. [PMID: 28580957 DOI: 10.1038/nri.2017.52)] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
The nuclear factor-κB (NF-κB) family of transcription factors is activated by canonical and non-canonical signalling pathways, which differ in both signalling components and biological functions. Recent studies have revealed important roles for the non-canonical NF-κB pathway in regulating different aspects of immune functions. Defects in non-canonical NF-κB signalling are associated with severe immune deficiencies, whereas dysregulated activation of this pathway contributes to the pathogenesis of various autoimmune and inflammatory diseases. Here we review the signalling mechanisms and the biological function of the non-canonical NF-κB pathway. We also discuss recent progress in elucidating the molecular mechanisms regulating non-canonical NF-κB pathway activation, which may provide new opportunities for therapeutic strategies.
Collapse
|
76
|
Abstract
The nuclear factor-κB (NF-κB) family of transcription factors is activated by canonical and non-canonical signalling pathways, which differ in both signalling components and biological functions. Recent studies have revealed important roles for the non-canonical NF-κB pathway in regulating different aspects of immune functions. Defects in non-canonical NF-κB signalling are associated with severe immune deficiencies, whereas dysregulated activation of this pathway contributes to the pathogenesis of various autoimmune and inflammatory diseases. Here we review the signalling mechanisms and the biological function of the non-canonical NF-κB pathway. We also discuss recent progress in elucidating the molecular mechanisms regulating non-canonical NF-κB pathway activation, which may provide new opportunities for therapeutic strategies.
Collapse
Affiliation(s)
- Shao-Cong Sun
- Department of Immunology, The University of Texas MD Anderson Cancer Center, MD Anderson Cancer Center UT Heath Graduate School of Biomedical Sciences, 7455 Fannin Street, Box 902, Houston, Texas 77030, USA
| |
Collapse
|
77
|
Wang Y, Li J, Huang Y, Dai X, Liu Y, Liu Z, Wang Y, Wang N, Zhang P. Tripartite motif-containing 28 bridges endothelial inflammation and angiogenic activity by retaining expression of TNFR-1 and -2 and VEGFR2 in endothelial cells. FASEB J 2017; 31:2026-2036. [PMID: 28159803 DOI: 10.1096/fj.201600988rr] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 01/17/2017] [Indexed: 11/11/2022]
Abstract
Angiogenesis and inflammation are regarded as important factors in the pathogenesis of chronic inflammation, cancer, and wound healing. Recent studies have supported prior evidence that common signaling pathways are involved in angiogenesis and inflammatory responses; however, key factors controlling both processes remain unclear. Although tripartite motif-containing (TRIM)-28 is known to have an immunosuppressive role in immune cells, its expression level and role in endothelial cells (ECs) are still unclear. In this study, we investigated the role of TRIM28 in inflammatory responses and angiogenic activity of ECs for the first time. We showed that TRIM28 is the most abundant TRIM family member and is localized in nuclei of ECs. Small interfering RNA-mediated knockdown of TRIM28 strikingly suppressed expression of TNF receptor (TNFR)-1 and -2, decreased TNF-α-induced phosphorylation of IKKα/β and IκBα and degradation of IκBα and nuclear translocation of p65, and suppressed basal level and TNF-α-induced expression of chemokines and adhesion molecules, including VCAM-1, IL-6, ICAM-1, E-selectin, and monocyte chemoattractant protein (MCP)-1. Unexpectedly, IL-8 was potentiated by TRIM28 knockdown in ECs in an NF-κB-inducing kinase-dependent manner. Meanwhile, knockdown of TRIM28 inhibited expression of VEGF receptor 2 and suppressed VEGF-induced proliferation and tube formation by ECs. Finally, knockdown of TRIM28 suppressed recruitment of ECs in vivo in a murine synthetic basement membrane model. In summary, we found that TRIM28 acts as a central factor in controlling endothelial inflammatory responses and angiogenic activities by retaining expression of TNFR-1 and -2 and VEGF receptor 2 in ECs.-Wang, Y., Li, J., Huang Y., Dai, X., Liu, Y., Liu, Z., Wang, Y., Wang, N., Zhang, P. Tripartite motif-containing 28 bridges endothelial inflammation and angiogenic activity by retaining expression of TNFR1 and -2 and VEGFR2 in endothelial cells.
Collapse
Affiliation(s)
- Yinfang Wang
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jinping Li
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yitong Huang
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiuqin Dai
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Youbin Liu
- Department of Cardiovascular Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zongjun Liu
- Department of Cardiovascular Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ying Wang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, Florida, USA; and
| | - Nanping Wang
- The Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Peng Zhang
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China;
- Department of Cardiovascular Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
78
|
Ravani A, Vincenzi F, Bortoluzzi A, Padovan M, Pasquini S, Gessi S, Merighi S, Borea PA, Govoni M, Varani K. Role and Function of A 2A and A₃ Adenosine Receptors in Patients with Ankylosing Spondylitis, Psoriatic Arthritis and Rheumatoid Arthritis. Int J Mol Sci 2017; 18:ijms18040697. [PMID: 28338619 PMCID: PMC5412283 DOI: 10.3390/ijms18040697] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 03/13/2017] [Accepted: 03/20/2017] [Indexed: 12/12/2022] Open
Abstract
Rheumatoid arthritis (RA), ankylosing spondylitis (AS) and psoriatic arthritis (PsA) are chronic inflammatory rheumatic diseases that affect joints, causing debilitating pain and disability. Adenosine receptors (ARs) play a key role in the mechanism of inflammation, and the activation of A2A and A₃AR subtypes is often associated with a reduction of the inflammatory status. The aim of this study was to investigate the involvement of ARs in patients suffering from early-RA (ERA), RA, AS and PsA. Messenger RNA (mRNA) analysis and saturation binding experiments indicated an upregulation of A2A and A₃ARs in lymphocytes obtained from patients when compared with healthy subjects. A2A and A₃AR agonists inhibited nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) activation and reduced inflammatory cytokines release, such as tumor necrosis factor-α (TNF-α), interleukin (IL)-1β and IL-6. Moreover, A2A and A₃AR activation mediated a reduction of metalloproteinases (MMP)-1 and MMP-3. The effect of the agonists was abrogated by selective antagonists demonstrating the direct involvement of these receptor subtypes. Taken together, these data confirmed the involvement of ARs in chronic autoimmune rheumatic diseases highlighting the possibility to exploit A2A and A₃ARs as therapeutic targets, with the aim to limit the inflammatory responses usually associated with RA, AS and PsA.
Collapse
MESH Headings
- Adenosine/analogs & derivatives
- Adenosine/chemistry
- Adenosine/metabolism
- Adenosine A2 Receptor Agonists/chemistry
- Adenosine A2 Receptor Agonists/metabolism
- Adenosine A2 Receptor Antagonists/chemistry
- Adenosine A2 Receptor Antagonists/metabolism
- Adenosine A3 Receptor Agonists/chemistry
- Adenosine A3 Receptor Agonists/metabolism
- Adenosine A3 Receptor Antagonists/chemistry
- Adenosine A3 Receptor Antagonists/metabolism
- Arthritis, Psoriatic/metabolism
- Arthritis, Psoriatic/pathology
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/pathology
- Case-Control Studies
- Cytokines/metabolism
- Female
- Humans
- Kinetics
- Lymphocytes/metabolism
- Male
- Matrix Metalloproteinase 1/metabolism
- Matrix Metalloproteinase 3/metabolism
- Middle Aged
- NF-kappa B/metabolism
- Phenethylamines/chemistry
- Phenethylamines/metabolism
- Pyrazoles/chemistry
- Pyrazoles/metabolism
- Pyrimidines/chemistry
- Pyrimidines/metabolism
- RNA, Messenger/metabolism
- Receptor, Adenosine A2A/genetics
- Receptor, Adenosine A2A/metabolism
- Receptor, Adenosine A3/genetics
- Receptor, Adenosine A3/metabolism
- Spondylitis, Ankylosing/metabolism
- Spondylitis, Ankylosing/pathology
Collapse
Affiliation(s)
- Annalisa Ravani
- Department of Medical Sciences, Pharmacology Unit, University of Ferrara, 44121 Ferrara, Italy.
| | - Fabrizio Vincenzi
- Department of Medical Sciences, Pharmacology Unit, University of Ferrara, 44121 Ferrara, Italy.
| | - Alessandra Bortoluzzi
- Department of Medical Sciences, Section of Rheumatology, University of Ferrara and Azienda Ospedaliero Universitaria Sant'Anna, 44124 Cona, Ferrara, Italy.
| | - Melissa Padovan
- Department of Medical Sciences, Section of Rheumatology, University of Ferrara and Azienda Ospedaliero Universitaria Sant'Anna, 44124 Cona, Ferrara, Italy.
| | - Silvia Pasquini
- Department of Medical Sciences, Pharmacology Unit, University of Ferrara, 44121 Ferrara, Italy.
| | - Stefania Gessi
- Department of Medical Sciences, Pharmacology Unit, University of Ferrara, 44121 Ferrara, Italy.
| | - Stefania Merighi
- Department of Medical Sciences, Pharmacology Unit, University of Ferrara, 44121 Ferrara, Italy.
| | - Pier Andrea Borea
- Department of Medical Sciences, Pharmacology Unit, University of Ferrara, 44121 Ferrara, Italy.
| | - Marcello Govoni
- Department of Medical Sciences, Section of Rheumatology, University of Ferrara and Azienda Ospedaliero Universitaria Sant'Anna, 44124 Cona, Ferrara, Italy.
| | - Katia Varani
- Department of Medical Sciences, Pharmacology Unit, University of Ferrara, 44121 Ferrara, Italy.
| |
Collapse
|
79
|
Arroul-Lammali A, Rahal F, Chetouane R, Djeraba Z, Medjeber O, Ladjouze-Rezig A, Touil-Boukoffa C. Ex vivo all-trans retinoic acid modulates NO production and regulates IL-6 effect during rheumatoid arthritis: a study in Algerian patients. Immunopharmacol Immunotoxicol 2017; 39:87-96. [PMID: 28211306 DOI: 10.1080/08923973.2017.1285919] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease. The pathophysiology of RA implicates several mediators such as nitric oxide (NO) and cytokines such as interleukin-6 (IL-6), which is deeply involved in the main characteristics of RA. Furthermore, all-trans retinoic acid (ATRA) is an active vitamin A derivative well-known to have diverse immunomodulatory actions. In our study, we investigated first, the ex vivo immunomodulatory potential of ATRA on NO pathway by peripheral blood mononuclear cells (PBMCs) from Algerian RA patients. Then, we assessed the possible regulatory effect of ATRA on NO production induced by IL-6. PBMCs isolated from active and inactive RA patients and healthy controls were cultured with different concentrations of IL-6 or/with ATRA. NO production was assessed using the Griess method. Inducible nitric oxide synthase expression and NF-κB activity were analyzed by immunofluorescence test. Our results revealed a high NO production during active RA. We noticed that while IL-6 induced a high NO production and iNOS expression, ATRA downregulated both. ATRA also inhibited nuclear NF-κB translocation. Interestingly, it seems that NO production mediated by IL-6 on PBMCs of RA patients is downregulated by ATRA. Taken together, our results highlight the immunomodulatory effect of ATRA on NO pathway in RA patients and its possible role in regulating IL-6-mediated NO production. All these findings suggest its potential therapeutic role during RA.
Collapse
Affiliation(s)
- Amina Arroul-Lammali
- a Laboratory of Cellular and Molecular Biology (LBCM), Cytokines and NO Synthases Team, Faculty of Biological Sciences , USTHB (University of Sciences and Technology) , Algiers , Algeria
| | - Fadia Rahal
- b Rheumatology Department , Ben aknoun hospital EHS , Algiers , Algeria
| | - Radia Chetouane
- b Rheumatology Department , Ben aknoun hospital EHS , Algiers , Algeria
| | - Zineb Djeraba
- a Laboratory of Cellular and Molecular Biology (LBCM), Cytokines and NO Synthases Team, Faculty of Biological Sciences , USTHB (University of Sciences and Technology) , Algiers , Algeria
| | - Oussama Medjeber
- a Laboratory of Cellular and Molecular Biology (LBCM), Cytokines and NO Synthases Team, Faculty of Biological Sciences , USTHB (University of Sciences and Technology) , Algiers , Algeria
| | | | - Chafia Touil-Boukoffa
- a Laboratory of Cellular and Molecular Biology (LBCM), Cytokines and NO Synthases Team, Faculty of Biological Sciences , USTHB (University of Sciences and Technology) , Algiers , Algeria
| |
Collapse
|
80
|
Pretorius E, Akeredolu OO, Soma P, Kell DB. Major involvement of bacterial components in rheumatoid arthritis and its accompanying oxidative stress, systemic inflammation and hypercoagulability. Exp Biol Med (Maywood) 2016; 242:355-373. [PMID: 27889698 PMCID: PMC5298544 DOI: 10.1177/1535370216681549] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
We review the evidence that infectious agents, including those that become dormant within the host, have a major role to play in much of the etiology of rheumatoid arthritis and the inflammation that is its hallmark. This occurs in particular because they can produce cross-reactive (auto-)antigens, as well as potent inflammagens such as lipopolysaccharide that can themselves catalyze further inflammagenesis, including via β-amyloid formation. A series of observables coexist in many chronic, inflammatory diseases as well as rheumatoid arthritis. They include iron dysregulation, hypercoagulability, anomalous morphologies of host erythrocytes, and microparticle formation. Iron dysregulation may be responsible for the periodic regrowth and resuscitation of the dormant bacteria, with concomitant inflammagen production. The present systems biology analysis benefits from the philosophical idea of "coherence," that reflects the principle that if a series of ostensibly unrelated findings are brought together into a self-consistent narrative, that narrative is thereby strengthened. As such, we provide a coherent and testable narrative for the major involvement of (often dormant) bacteria in rheumatoid arthritis.
Collapse
Affiliation(s)
- Etheresia Pretorius
- 1 Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia, Pretoria 0007, South Africa
| | - Oore-Ofe Akeredolu
- 1 Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia, Pretoria 0007, South Africa
| | - Prashilla Soma
- 1 Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia, Pretoria 0007, South Africa
| | - Douglas B Kell
- 2 School of Chemistry, The University of Manchester, Manchester, M13 9PL, UK.,3 The Manchester Institute of Biotechnology, The University of Manchester, Manchester, M1 7DN, UK.,4 Centre for Synthetic Biology of Fine and Speciality Chemicals, The University of Manchester, Manchester, M1 7DN, UK
| |
Collapse
|
81
|
Suppression of NF-κB activity via nanoparticle-based siRNA delivery alters early cartilage responses to injury. Proc Natl Acad Sci U S A 2016; 113:E6199-E6208. [PMID: 27681622 PMCID: PMC5068304 DOI: 10.1073/pnas.1608245113] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Osteoarthritis (OA) is a major cause of disability and morbidity in the aging population. Joint injury leads to cartilage damage, a known determinant for subsequent development of posttraumatic OA, which accounts for 12% of all OA. Understanding the early molecular and cellular responses postinjury may provide targets for therapeutic interventions that limit articular degeneration. Using a murine model of controlled knee joint impact injury that allows the examination of cartilage responses to injury at specific time points, we show that intraarticular delivery of a peptidic nanoparticle complexed to NF-κB siRNA significantly reduces early chondrocyte apoptosis and reactive synovitis. Our data suggest that NF-κB siRNA nanotherapy maintains cartilage homeostasis by enhancing AMPK signaling while suppressing mTORC1 and Wnt/β-catenin activity. These findings delineate an extensive crosstalk between NF-κB and signaling pathways that govern cartilage responses postinjury and suggest that delivery of NF-κB siRNA nanotherapy to attenuate early inflammation may limit the chronic consequences of joint injury. Therapeutic benefits of siRNA nanotherapy may also apply to primary OA in which NF-κB activation mediates chondrocyte catabolic responses. Additionally, a critical barrier to the successful development of OA treatment includes ineffective delivery of therapeutic agents to the resident chondrocytes in the avascular cartilage. Here, we show that the peptide-siRNA nanocomplexes are nonimmunogenic, are freely and deeply penetrant to human OA cartilage, and persist in chondrocyte lacunae for at least 2 wk. The peptide-siRNA platform thus provides a clinically relevant and promising approach to overcoming the obstacles of drug delivery to the highly inaccessible chondrocytes.
Collapse
|
82
|
Kell DB, Pretorius E. On the translocation of bacteria and their lipopolysaccharides between blood and peripheral locations in chronic, inflammatory diseases: the central roles of LPS and LPS-induced cell death. Integr Biol (Camb) 2016; 7:1339-77. [PMID: 26345428 DOI: 10.1039/c5ib00158g] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We have recently highlighted (and added to) the considerable evidence that blood can contain dormant bacteria. By definition, such bacteria may be resuscitated (and thus proliferate). This may occur under conditions that lead to or exacerbate chronic, inflammatory diseases that are normally considered to lack a microbial component. Bacterial cell wall components, such as the endotoxin lipopolysaccharide (LPS) of Gram-negative strains, are well known as potent inflammatory agents, but should normally be cleared. Thus, their continuing production and replenishment from dormant bacterial reservoirs provides an easy explanation for the continuing, low-grade inflammation (and inflammatory cytokine production) that is characteristic of many such diseases. Although experimental conditions and determinants have varied considerably between investigators, we summarise the evidence that in a great many circumstances LPS can play a central role in all of these processes, including in particular cell death processes that permit translocation between the gut, blood and other tissues. Such localised cell death processes might also contribute strongly to the specific diseases of interest. The bacterial requirement for free iron explains the strong co-existence in these diseases of iron dysregulation, LPS production, and inflammation. Overall this analysis provides an integrative picture, with significant predictive power, that is able to link these processes via the centrality of a dormant blood microbiome that can resuscitate and shed cell wall components.
Collapse
Affiliation(s)
- Douglas B Kell
- School of Chemistry and The Manchester Institute of Biotechnology, The University of Manchester, 131, Princess St, Manchester M1 7DN, Lancs, UK.
| | - Etheresia Pretorius
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia 0007, South Africa.
| |
Collapse
|
83
|
Wang F, Chen FF, Gao WB, Wang HY, Zhao NW, Xu M, Gao DY, Yu W, Yan XL, Zhao JN, Li XJ. Identification of citrullinated peptides in the synovial fluid of patients with rheumatoid arthritis using LC-MALDI-TOF/TOF. Clin Rheumatol 2016; 35:2185-94. [PMID: 27060082 PMCID: PMC4989008 DOI: 10.1007/s10067-016-3247-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 01/11/2016] [Accepted: 03/23/2016] [Indexed: 12/11/2022]
Abstract
The objective of the study is to investigate potential citrullinated autoantigens as targets of anti-citrullinated protein antibodies (ACPAs) response in synovial fluids (SFs) of patients with rheumatoid arthritis (RA). SFs from six RA patients and six osteoarthritis (OA) patients as controls were collected. The citrullinated proteins in SFs were extracted by immunoprecipitation with rabbit anti-citrulline antibodies. Matrix-assisted laser desorption/ionization time of flight mass spectrometry/time of flight mass spectrometry (MALDI-TOF/TOF) mass spectrometry was subsequently performed to discover a characteristic neutral loss to finally determine citrullinated autoantigens. A total of 182 citrullinated peptides and 200 citrullinated sites were identified in RA SFs, while 3 citrullinated peptides and 4 citrullinated sites were identified in OA SFs. The 182 citrullinated peptides from RA SFs and the 3 citrullinated peptides from OA SFs were derived from 83 and 3 autoantigens, respectively. Eighty-three autoantigens except protein-arginine deiminase type-2 (PADI2) and protein-arginine deiminase type-2 (PADI4) were over-citrullinated compared with controls, and the citrullinated sites of PADI2 and PADI4 were different in two groups. Interestingly, citrullinated histone H3.3 (H3F3A) was found in OA controls, but not in RA groups. The differential citrullinated proteins identified in RA SFs suggested potential autoantigens were targeted for ACPAs response and might contribute to the induction and perpetuation of complement activation and joint inflammation in RA.
Collapse
Affiliation(s)
- Fei Wang
- Institute of Clinical Laboratory Science, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, China
| | - Fang-Fang Chen
- Institute of Clinical Laboratory Science, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, China
| | - Wen-Bo Gao
- Institute of Clinical Laboratory Science, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, China
| | - Hai-Yong Wang
- Institute of Clinical Laboratory Science, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, China
| | - Ning-Wei Zhao
- Biomedical Research Laboratory, Shimadzu (China) Co., Ltd., Shanghai, 200052, China
| | - Min Xu
- Institute of Clinical Laboratory Science, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, China
| | - De-Yu Gao
- Institute of Clinical Laboratory Science, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, China
| | - Wei Yu
- Institute of Clinical Laboratory Science, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, China
| | - Xiao-Ling Yan
- Institute of Clinical Laboratory Science, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, China
| | - Jian-Ning Zhao
- Department of Osteology, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, China
| | - Xiao-Jun Li
- Institute of Clinical Laboratory Science, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, China. .,State Key Laboratory of Analytical Chemistry for Life Science, Department of Chemistry, Nanjing University, Nanjing, 210093, China.
| |
Collapse
|
84
|
Shih RH, Wang CY, Yang CM. NF-kappaB Signaling Pathways in Neurological Inflammation: A Mini Review. Front Mol Neurosci 2015; 8:77. [PMID: 26733801 PMCID: PMC4683208 DOI: 10.3389/fnmol.2015.00077] [Citation(s) in RCA: 565] [Impact Index Per Article: 62.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 11/30/2015] [Indexed: 12/14/2022] Open
Abstract
The NF-κB (nuclear factor κ-light-chain-enhancer of activated B cells) transcription factor family is a pleiotropic regulator of many cellular signaling pathways, providing a mechanism for the cells in response to a wide variety of stimuli linking to inflammation. The stimulated cells will be regulated by not only the canonical but also non-canonical NF-κB pathways. To initiate both of these pathways, IκB-degradation triggers NF-κB release and the nuclear translocated-heterodimer (or homodimer) can associate with the κB sites of promoter to regulate the gene transcriptions. NF-κB ubiquitously expresses in neurons and the constitutive NF-κB activation is associated with processing of neuronal information. NF-κB can regulate the transcription of genes such as chemokines, cytokines, proinflammatory enzymes, adhesion molecules, proinflammatory transcription factors, and other factors to modulate the neuronal survival. In neuronal insult, NF-κB constitutively active in neuron cell bodies can protect neurons against different injuries and regulate the neuronal inflammatory reactions. Besides neurons, NF-κB transcription factors are abundant in glial cells and cerebral blood vessels and the diverse functions of NF-κB also regulate the inflammatory reaction around the neuronal environment. NF-κB transcription factors are abundant in the brain and exhibit diverse functions. Several central nerve system (CNS) diseases are linked to NF-κB activated by inflammatory mediators. The RelA and c-Rel expression produce opposite effects on neuronal survival. Importantly, c-Rel expression in CNS plays a critical role in anti-apoptosis and reduces the age-related behaviors. Moreover, the different subunits of NF-κB dimer formation can modulate the neuroninflammation, neuronal protection, or neurotoxicity. The diverse functions of NF-κB depend on the subunits of the NF-κB dimer-formation which enable us to develop a therapeutic approach to neuroinflammation based on a new concept of inflammation as a strategic tool in neuronal cells. However, the detail role of NF-κB in neuroinflammation, remains to be clarified. In the present article, we provide an updated review of the current state of our knowledge about relationship between NF-κB and neuroinflammation.
Collapse
Affiliation(s)
- Ruey-Horng Shih
- Institute of Neuroscience, National Chengchi University Taipei, Taiwan
| | - Chen-Yu Wang
- Department of Physiology and Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung University Tao-Yuan, Taiwan
| | - Chuen-Mao Yang
- Department of Physiology and Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung University Tao-Yuan, Taiwan
| |
Collapse
|
85
|
Tas SW, Maracle CX, Balogh E, Szekanecz Z. Targeting of proangiogenic signalling pathways in chronic inflammation. Nat Rev Rheumatol 2015; 12:111-22. [PMID: 26633288 DOI: 10.1038/nrrheum.2015.164] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Angiogenesis is de novo capillary outgrowth from pre-existing blood vessels. This process not only is crucial for normal development, but also has an important role in supplying oxygen and nutrients to inflamed tissues, as well as in facilitating the migration of inflammatory cells to the synovium in rheumatoid arthritis, spondyloarthritis and other systemic autoimmune diseases. Neovascularization is dependent on the balance of proangiogenic and antiangiogenic mediators, including growth factors, cytokines, chemokines, cell adhesion molecules and matrix metalloproteinases. This Review describes the various intracellular signalling pathways that govern these angiogenic processes and discusses potential approaches to interfere with pathological angiogenesis, and thereby ameliorate inflammatory disease, by targeting these pathways.
Collapse
Affiliation(s)
- Sander W Tas
- Amsterdam Rheumatology &Immunology Centre, Department of Experimental Immunology, Academic Medical Centre and University of Amsterdam, EULAR &FOCIS (Federation of Clinical Immunology Societies) Centre of Excellence, Meibergdreef 9, F4-105, 1105 AZ Amsterdam, Netherlands
| | - Chrissta X Maracle
- Amsterdam Rheumatology &Immunology Centre, Department of Experimental Immunology, Academic Medical Centre and University of Amsterdam, EULAR &FOCIS (Federation of Clinical Immunology Societies) Centre of Excellence, Meibergdreef 9, F4-105, 1105 AZ Amsterdam, Netherlands
| | - Emese Balogh
- Department of Rheumatology, Institute of Medicine, University of Debrecen, Faculty of Medicine, Nagyerdei Str. 98, Debrecen 4032, Hungary
| | - Zoltán Szekanecz
- Department of Rheumatology, Institute of Medicine, University of Debrecen, Faculty of Medicine, Nagyerdei Str. 98, Debrecen 4032, Hungary
| |
Collapse
|
86
|
Maijer KI, Noort AR, de Hair MJH, van der Leij C, van Zoest KPM, Choi IY, Gerlag DM, Maas M, Tak PP, Tas SW. Nuclear Factor-κB-inducing Kinase Is Expressed in Synovial Endothelial Cells in Patients with Early Arthritis and Correlates with Markers of Inflammation: A Prospective Cohort Study. J Rheumatol 2015; 42:1573-81. [PMID: 26178280 DOI: 10.3899/jrheum.150245] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2015] [Indexed: 01/15/2023]
Abstract
OBJECTIVE The nuclear factor-κB (NF-κB) family of transcription factors is strongly involved in synovial inflammation. We have previously shown that NF-κB-inducing kinase (NIK) is a key regulator of inflammation-induced angiogenesis in rheumatoid arthritis (RA) synovial tissue (ST). Here, we investigated synovial NIK expression in patients with early arthritis and in autoantibody-positive individuals at risk of developing RA. METHODS ST biopsies were obtained by arthroscopy from 154 patients with early arthritis (duration < 1 yr) with various diagnoses and 54 IgM rheumatoid factor-positive and/or anticitrullinated protein antibodies-positive individuals without evidence of arthritis. ST was stained for NIK and endothelial cell (EC) markers. Additionally, measures of disease activity were collected and contrast-enhanced magnetic resonance imaging (MRI) was performed in a subset of these patients. RESULTS In patients with early arthritis, NIK was predominantly expressed in EC of small blood vessels. Further, NIK expression correlated with erythrocyte sedimentation rate (r 0.184, p = 0.024), C-reactive protein (r 0.194, p = 0.017), joint swelling (r 0.297, p < 0.001), synovial immune cell markers (lining r 0.585, p < 0.001; sublining macrophages r 0.728, p < 0.001; T cells r 0.733, p < 0.001; and B cells r 0.264, p = 0.040), MRI effusion (r 0.665, p < 0.001), MRI synovitis (r 0.632, p < 0.001), and MRI total score (r 0.569, p < 0.001). In 18.5% of autoantibody-positive individuals, ST NIK(+)EC were present, but this was not predictive of the development of arthritis. CONCLUSION NIK(+)EC are present in the earliest phase of synovial inflammation and may be indicative of high angiogenic activity in the inflamed ST. Therefore, NIK(+)EC may play an important role in the persistence of synovitis. Collectively, our data underscore the importance of angiogenesis in synovial inflammation and identify NIK as a potential therapeutic target in arthritis.
Collapse
Affiliation(s)
- Karen I Maijer
- From the Division of Clinical Immunology and Rheumatology, the Department of Experimental Immunology, and the Department of Radiology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands; GlaxoSmithKline, Stevenage; University of Cambridge, Cambridge, UK.K.I. Maijer, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; A.R. Noort, MSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; M.J. de Hair, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; C. van der Leij, MD, Department of Radiology, Academic Medical Center/University of Amsterdam; K.P. van Zoest, BSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; I.Y. Choi, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; D.M. Gerlag, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline; M. Maas, MD, PhD, Department of Radiology, Academic Medical Center/University of Amsterdam; P.P. Tak, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline, Stevenage, and University of Cambridge; S.W. Tas, MD, PhD, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam
| | - Ae Ri Noort
- From the Division of Clinical Immunology and Rheumatology, the Department of Experimental Immunology, and the Department of Radiology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands; GlaxoSmithKline, Stevenage; University of Cambridge, Cambridge, UK.K.I. Maijer, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; A.R. Noort, MSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; M.J. de Hair, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; C. van der Leij, MD, Department of Radiology, Academic Medical Center/University of Amsterdam; K.P. van Zoest, BSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; I.Y. Choi, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; D.M. Gerlag, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline; M. Maas, MD, PhD, Department of Radiology, Academic Medical Center/University of Amsterdam; P.P. Tak, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline, Stevenage, and University of Cambridge; S.W. Tas, MD, PhD, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam
| | - Maria J H de Hair
- From the Division of Clinical Immunology and Rheumatology, the Department of Experimental Immunology, and the Department of Radiology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands; GlaxoSmithKline, Stevenage; University of Cambridge, Cambridge, UK.K.I. Maijer, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; A.R. Noort, MSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; M.J. de Hair, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; C. van der Leij, MD, Department of Radiology, Academic Medical Center/University of Amsterdam; K.P. van Zoest, BSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; I.Y. Choi, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; D.M. Gerlag, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline; M. Maas, MD, PhD, Department of Radiology, Academic Medical Center/University of Amsterdam; P.P. Tak, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline, Stevenage, and University of Cambridge; S.W. Tas, MD, PhD, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam
| | - Christiaan van der Leij
- From the Division of Clinical Immunology and Rheumatology, the Department of Experimental Immunology, and the Department of Radiology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands; GlaxoSmithKline, Stevenage; University of Cambridge, Cambridge, UK.K.I. Maijer, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; A.R. Noort, MSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; M.J. de Hair, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; C. van der Leij, MD, Department of Radiology, Academic Medical Center/University of Amsterdam; K.P. van Zoest, BSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; I.Y. Choi, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; D.M. Gerlag, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline; M. Maas, MD, PhD, Department of Radiology, Academic Medical Center/University of Amsterdam; P.P. Tak, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline, Stevenage, and University of Cambridge; S.W. Tas, MD, PhD, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam
| | - Katinka P M van Zoest
- From the Division of Clinical Immunology and Rheumatology, the Department of Experimental Immunology, and the Department of Radiology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands; GlaxoSmithKline, Stevenage; University of Cambridge, Cambridge, UK.K.I. Maijer, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; A.R. Noort, MSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; M.J. de Hair, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; C. van der Leij, MD, Department of Radiology, Academic Medical Center/University of Amsterdam; K.P. van Zoest, BSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; I.Y. Choi, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; D.M. Gerlag, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline; M. Maas, MD, PhD, Department of Radiology, Academic Medical Center/University of Amsterdam; P.P. Tak, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline, Stevenage, and University of Cambridge; S.W. Tas, MD, PhD, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam
| | - Ivy Y Choi
- From the Division of Clinical Immunology and Rheumatology, the Department of Experimental Immunology, and the Department of Radiology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands; GlaxoSmithKline, Stevenage; University of Cambridge, Cambridge, UK.K.I. Maijer, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; A.R. Noort, MSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; M.J. de Hair, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; C. van der Leij, MD, Department of Radiology, Academic Medical Center/University of Amsterdam; K.P. van Zoest, BSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; I.Y. Choi, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; D.M. Gerlag, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline; M. Maas, MD, PhD, Department of Radiology, Academic Medical Center/University of Amsterdam; P.P. Tak, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline, Stevenage, and University of Cambridge; S.W. Tas, MD, PhD, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam
| | - Daniëlle M Gerlag
- From the Division of Clinical Immunology and Rheumatology, the Department of Experimental Immunology, and the Department of Radiology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands; GlaxoSmithKline, Stevenage; University of Cambridge, Cambridge, UK.K.I. Maijer, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; A.R. Noort, MSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; M.J. de Hair, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; C. van der Leij, MD, Department of Radiology, Academic Medical Center/University of Amsterdam; K.P. van Zoest, BSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; I.Y. Choi, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; D.M. Gerlag, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline; M. Maas, MD, PhD, Department of Radiology, Academic Medical Center/University of Amsterdam; P.P. Tak, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline, Stevenage, and University of Cambridge; S.W. Tas, MD, PhD, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam
| | - Mario Maas
- From the Division of Clinical Immunology and Rheumatology, the Department of Experimental Immunology, and the Department of Radiology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands; GlaxoSmithKline, Stevenage; University of Cambridge, Cambridge, UK.K.I. Maijer, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; A.R. Noort, MSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; M.J. de Hair, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; C. van der Leij, MD, Department of Radiology, Academic Medical Center/University of Amsterdam; K.P. van Zoest, BSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; I.Y. Choi, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; D.M. Gerlag, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline; M. Maas, MD, PhD, Department of Radiology, Academic Medical Center/University of Amsterdam; P.P. Tak, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline, Stevenage, and University of Cambridge; S.W. Tas, MD, PhD, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam
| | - Paul P Tak
- From the Division of Clinical Immunology and Rheumatology, the Department of Experimental Immunology, and the Department of Radiology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands; GlaxoSmithKline, Stevenage; University of Cambridge, Cambridge, UK.K.I. Maijer, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; A.R. Noort, MSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; M.J. de Hair, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; C. van der Leij, MD, Department of Radiology, Academic Medical Center/University of Amsterdam; K.P. van Zoest, BSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; I.Y. Choi, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; D.M. Gerlag, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline; M. Maas, MD, PhD, Department of Radiology, Academic Medical Center/University of Amsterdam; P.P. Tak, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline, Stevenage, and University of Cambridge; S.W. Tas, MD, PhD, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam
| | - Sander W Tas
- From the Division of Clinical Immunology and Rheumatology, the Department of Experimental Immunology, and the Department of Radiology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands; GlaxoSmithKline, Stevenage; University of Cambridge, Cambridge, UK.K.I. Maijer, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; A.R. Noort, MSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; M.J. de Hair, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; C. van der Leij, MD, Department of Radiology, Academic Medical Center/University of Amsterdam; K.P. van Zoest, BSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; I.Y. Choi, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; D.M. Gerlag, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline; M. Maas, MD, PhD, Department of Radiology, Academic Medical Center/University of Amsterdam; P.P. Tak, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline, Stevenage, and University of Cambridge; S.W. Tas, MD, PhD, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam.
| |
Collapse
|
87
|
van Delft MAM, Huitema LFA, Tas SW. The contribution of NF-κB signalling to immune regulation and tolerance. Eur J Clin Invest 2015; 45:529-39. [PMID: 25735405 DOI: 10.1111/eci.12430] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 03/02/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND Immune regulation is necessary to control inflammatory responses and to prevent autoimmune diseases. Therefore, mechanisms of central and peripheral tolerance have evolved to ensure that T cells recognize antigens as self- or non-self-antigens. The thymus is crucially important for central tolerance induction to self-antigens via negative selection of T cells. However, if T cells escape negative selection in the thymus and enter the periphery, peripheral mechanisms are active to warrant immune tolerance. Secondary lymphoid organs, as well as tolerogenic dendritic cells and regulatory T cells, play an important role in peripheral tolerance. In chronic inflammatory diseases, tertiary lymphoid organs are sometimes formed that may also be involved in the induction of peripheral tolerance. This review discusses the main processes that are involved in immune regulation and tolerance, and focuses on the contribution of NF-κB signalling to these processes. MATERIAL AND METHODS This narrative review is based on peer-reviewed publications listed on PubMed up to December 2014. The focus of our literature search was on studies investigating the role of (non)canonical NF-κB signalling in central and peripheral mechanisms of tolerance. Only studies published in English language were considered. RESULTS This review discusses the immune phenotype of mutant mice with defective (non)canonical NF-κB signalling, corroborated with human data, and emphasizes the contribution of the noncanonical NF-κB pathway to immune regulation and tolerance induction. CONCLUSIONS Noncanonical NF-κB signalling has an important immunoregulatory role in the immune system and contributes to both central and peripheral mechanisms of tolerance.
Collapse
Affiliation(s)
- Myrthe A M van Delft
- Amsterdam Rheumatology & immunology Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | |
Collapse
|