1
|
Zhou H, Huang W, Li J, Chen P, Shen L, Huang W, Mai K, Zou H, Shi X, Weng Y, Liu Y, Yang Z, Ou C. Oral probiotic extracellular vesicle therapy mitigates Influenza A Virus infection via blunting IL-17 signaling. Bioact Mater 2025; 45:401-416. [PMID: 39697241 PMCID: PMC11652895 DOI: 10.1016/j.bioactmat.2024.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/12/2024] [Accepted: 11/12/2024] [Indexed: 12/20/2024] Open
Abstract
The influenza A virus (IAV) damages intestinal mucosal tissues beyond the respiratory tract. Probiotics play a crucial role in maintaining the balance and stability of the intestinal microecosystem. Extracellular vesicles (EVs) derived from probiotics have emerged as potential mediators of host immune response and anti-inflammatory effect. However, the specific anti-inflammatory effects and underlying mechanisms of probiotics-derived EVs on IAV remain unclear. In the present study, we investigated the therapeutic efficacy of Lactobacillus reuteri EHA2-derived EVs (LrEVs) in a mouse model of IAV infection. Oral LrEVs were distributed in the liver, lungs, and gastrointestinal tract. In mice infected with IAV, oral LrEVs administration alleviated IAV-induced damages in the lungs and intestines, modified the microbiota compositions, and increased the levels of short-chain fatty acids in those organs. Mechanistically, LrEVs exerted their protective effects against IAV infection by blunting the pro-inflammatory IL-17 signaling. Furthermore, FISH analysis detected miR-4239, one of the most abundant miRNAs in LrEVs, in both lung and intestinal tissues. We confirmed that miR-4239 directly targets IL-17a. Our findings paved the ground for future application of LrEVs in influenza treatment and offered new mechanistic insights regarding the anti-inflammatory role of miR-4239.
Collapse
Affiliation(s)
- Hongxia Zhou
- Dongguan Institute of Respiratory and Critical Care Medicine, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, 523018, China
| | - Wenbo Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Jieting Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Peier Chen
- The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, 523018, China
| | - Lihan Shen
- Dongguan Institute of Respiratory and Critical Care Medicine, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, 523018, China
| | - Wenjing Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Kailin Mai
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Heyan Zou
- Dongguan Institute of Respiratory and Critical Care Medicine, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, 523018, China
| | - Xueqin Shi
- The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, 523018, China
| | - Yunceng Weng
- Becton Dickinson Medical Devices (Shanghai) Co., Ltd., Guangzhou, 510180, China
| | - Yuhua Liu
- Department of General Practice, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, 523018, China
| | - Zifeng Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
- Guangzhou National Laboratory, Guangzhou, 510000, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR, 519020, China
| | - Caiwen Ou
- The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, 523018, China
| |
Collapse
|
2
|
Wang Z, Sun X, Lin Y, Fu Y, Yi Z. Stealth in non-tuberculous mycobacteria: clever challengers to the immune system. Microbiol Res 2025; 292:128039. [PMID: 39752805 DOI: 10.1016/j.micres.2024.128039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 01/19/2025]
Abstract
Non-tuberculous Mycobacteria (NTM) are found extensively in various environments, yet most are non-pathogenic. Only a limited number of these organisms can cause various infections, including those affecting the lungs, skin, and central nervous system, particularly when the host's autoimmune function is compromised. Among these, Non-tuberculous Mycobacteria Pulmonary Diseases (NTM-PD) are the most prevalent. Currently, there is a lack of effective treatments and preventive measures for NTM infections. This article aims to deepen the comprehension of the pathogenic mechanisms linked to NTM and to formulate new intervention strategies by synthesizing current research and detailing the different tactics used by NTM to avoid elimination by the host's immune response. These intricate mechanisms not only affect the innate immune response but also successfully oppose the adaptive immune response, establishing persistent infections within the host. This includes effects on the functions of macrophages, neutrophils, dendritic cells, and T lymphocytes, as well as modulation of cytokine production. The article particularly emphasizes the survival strategies of NTM within macrophages, such as inhibiting phagosome maturation and acidification, resisting intracellular killing mechanisms, and interfering with autophagy and cell death pathways. This review aims to deepen the understanding of NTM's immune evasion mechanisms, thereby facilitating efforts to inhibit its proliferation and spread within the host, ultimately providing new methods and strategies for NTM-related treatments.
Collapse
Affiliation(s)
- Zhenghao Wang
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, China
| | - Xiurong Sun
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, China
| | - Yuli Lin
- School of Medical Laboratory, Shandong Second Medical University, Weifang 261053, China
| | - Yurong Fu
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, China.
| | - Zhengjun Yi
- School of Medical Laboratory, Shandong Second Medical University, Weifang 261053, China.
| |
Collapse
|
3
|
Mann J, Runge S, Schell C, Gräwe K, Thoulass G, Lao J, Ammann S, Grün S, König C, Berger SA, Hild B, Aichele P, Rosshart SP, Ehl S. The Microbiome Modifies Manifestations of Hemophagocytic Lymphohistiocytosis in Perforin-Deficient Mice. Eur J Immunol 2025; 55:e202451061. [PMID: 39548906 PMCID: PMC11739664 DOI: 10.1002/eji.202451061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 11/18/2024]
Abstract
Primary hemophagocytic lymphohistiocytosis (HLH) is a life-threatening hyperinflammatory syndrome caused by inborn errors of cytotoxicity. Patients with biallelic PRF1 null mutations (encoding perforin) usually develop excessive immune cell activation, hypercytokinemia, and life-threatening immunopathology in the first 6 months of life, often without an apparent infectious trigger. In contrast, perforin-deficient (PKO) mice only develop HLH after systemic infection with lymphocytic choriomeningitis virus (LCMV). We hypothesized that restricted microbe-immune cell interactions due to specific pathogen-free (SPF) housing might explain the need for this specific viral trigger in PKO mice. To investigate the influence of a "wild" microbiome in PKO mice, we fostered PKO newborns with Wildling microbiota ('PKO-Wildlings') and monitored them for signs of HLH. PKO-Wildlings survived long-term without spontaneous disease. Also, systemic infection with vaccinia virus did not reach the threshold of immune activation required to trigger HLH in PKO-Wildlings. Interestingly, after infection with LCMV, PKO-Wildlings developed an altered HLH pattern. This included lower IFN-γ serum levels along with improved IFN-γ-driven anemia, but more elevated levels of IL-17 and increased liver inflammation compared with PKO-SPF mice. Thus, wild microbiota alone is not sufficient to trigger HLH in PKO mice, but host-microbe interactions shape inflammatory cytokine patterns, thereby influencing manifestations of HLH immunopathology.
Collapse
Affiliation(s)
- Jasmin Mann
- Institute for ImmunodeficiencyCenter for Chronic Immunodeficiency (CCI), Medical Center‐ University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
- Faculty of BiologyUniversity of FreiburgFreiburgGermany
| | - Solveig Runge
- Faculty of BiologyUniversity of FreiburgFreiburgGermany
- Department of Medicine II, Medical Center‐ University of Freiburg, Faculty of MedicineUniversity of FreiburgFreiburgGermany
- Department of Microbiome Research, University Hospital ErlangenFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
| | - Christoph Schell
- Institute for Surgical Pathology, Medical Center‐ University of Freiburg, Faculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Katja Gräwe
- Institute for Surgical Pathology, Medical Center‐ University of Freiburg, Faculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Gudrun Thoulass
- Institute for ImmunodeficiencyCenter for Chronic Immunodeficiency (CCI), Medical Center‐ University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
- Faculty of BiologyUniversity of FreiburgFreiburgGermany
| | - Jessica Lao
- Institute for ImmunodeficiencyCenter for Chronic Immunodeficiency (CCI), Medical Center‐ University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
- Faculty of BiologyUniversity of FreiburgFreiburgGermany
| | - Sandra Ammann
- Institute for ImmunodeficiencyCenter for Chronic Immunodeficiency (CCI), Medical Center‐ University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Sarah Grün
- Institute for ImmunodeficiencyCenter for Chronic Immunodeficiency (CCI), Medical Center‐ University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
- Faculty of BiologyUniversity of FreiburgFreiburgGermany
| | - Christoph König
- Institute for ImmunodeficiencyCenter for Chronic Immunodeficiency (CCI), Medical Center‐ University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
- Faculty of BiologyUniversity of FreiburgFreiburgGermany
| | - Sarah A. Berger
- Institute for ImmunodeficiencyCenter for Chronic Immunodeficiency (CCI), Medical Center‐ University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
- Faculty of BiologyUniversity of FreiburgFreiburgGermany
| | - Benedikt Hild
- Department of Gastroenterology, Hepatology and Transplantation MedicineMedical Faculty University of Duisburg‐EssenEssenGermany
| | - Peter Aichele
- Institute for ImmunodeficiencyCenter for Chronic Immunodeficiency (CCI), Medical Center‐ University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Stephan P. Rosshart
- Department of Medicine II, Medical Center‐ University of Freiburg, Faculty of MedicineUniversity of FreiburgFreiburgGermany
- Department of Microbiome Research, University Hospital ErlangenFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
| | - Stephan Ehl
- Institute for ImmunodeficiencyCenter for Chronic Immunodeficiency (CCI), Medical Center‐ University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
| |
Collapse
|
4
|
Joveini S, Yarmohammadi F, Iranshahi M, Nikpoor AR, Askari VR, Attaranzadeh A, Etemad L, Taherzadeh Z. Distinct therapeutic effects of auraptene and umbelliprenin on TNF-α and IL-17 levels in a murine model of chronic inflammation. Heliyon 2024; 10:e40731. [PMID: 39687160 PMCID: PMC11648749 DOI: 10.1016/j.heliyon.2024.e40731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 11/23/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Objective To compare the anti-arthritic potential of orally administered auraptene (AUR) and umbellliprenin (UMB) in chronic inflammation by exploring the differential effect on regulating TNF-α and IL-17. Methods & materials Sixty male rats were divided into ten groups, and after confirming chronic inflammation, the treatment groups received AUR or UMB orally for 9 days. On day 16, histopathological changes were evaluated. Altered serum levels of the inflammatory cytokines TNF-α and IL-17 were examined as the underlying mechanisms. Results Administering AUR orally at 16 mM/kg caused a significant increase in body weight gain compared to the baseline (p < 0.05), while UMB at a dose of 64 mM/kg significantly reduced edema size (p < 0.01). TNF-α levels were significantly lower in all doses of AUR and UMB treatments compared to the arthritis control group (p < 0.05). Treatment with AUR at all relative doses resulted in a significant decrease in IL-17 levels compared to the arthritis control group (p < 0.05), whereas UMB treatment did not show a significant effect on IL-17 levels. Conclusion AUR and UMB regulate TNF-α and IL-17 differently; AUR inhibits both, showing broad therapeutic potential, while UMB specifically targets TNF-α, showing a specialized role.
Collapse
Affiliation(s)
- Saeid Joveini
- Department of Pharmacodynamy and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Yarmohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mehrdad Iranshahi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amin Reza Nikpoor
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Vahid Reza Askari
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Armin Attaranzadeh
- Department of Medical Genetics, Faculty of Medicines, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Etemad
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zhila Taherzadeh
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Targeted Drug Delivery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
5
|
Lin L, Sun C, Ye Y, Zhu P, Pan K, Chen L. Transcriptome analysis revealed that ischemic post-conditioning suppressed the expression of inflammatory genes in lung ischemia-reperfusion injury. Front Genet 2024; 15:1425420. [PMID: 39655220 PMCID: PMC11625726 DOI: 10.3389/fgene.2024.1425420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024] Open
Abstract
Introduction Ischemic post-conditioning (I-post C) is a recognized therapeutic strategy for lung ischemia/reperfusion injury (LIRI). However, the specific mechanisms underlying the lung protection conferred by I-post C remain unclear. This study aimed to investigate the protective mechanisms and potential molecular regulatory networks of I-post C on lung tissue. Methods Transcriptome analysis was performed on rat lung tissues obtained from Sham, ischemia-reperfusion (IR), and I-post C groups using RNA-seq to identify differentially expressed genes (DEGs). Subsequently, gene ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, and gene set enrichment analysis (GSEA) were conducted to elucidate significantly enriched pathways in the IR and I-post C groups. Additionally, protein-protein interaction (PPI) network analysis was carried out to examine associations among the DEGs. Pathological changes in lung tissues were assessed using hematoxylin-eosin (H&E) staining. The expression levels of CXCL1 and CXCL6 in the IR and I-post C groups were evaluated through immunofluorescence and Western blotting. Results Our results showed that I-post C significantly attenuated both pulmonary edema and inflammatory cell infiltration. Transcriptome analysis identified 38 DEGs in the I-post C group compared to the IR group, comprising 21 upregulated and 17 downregulated genes. Among these, seven inflammation-related DEGs exhibited co-expression patterns with the Sham and IR groups, with notable downregulation of Cxcl1 and Cxcl6. GO analysis primarily linked these DEGs to neutrophil activation, chemotaxis, cytokine activity, and CCR chemokine receptor binding. KEGG analysis revealed enriched pathways, including the IL-17, TNF, and NF-κB signaling pathways. GSEA indicated downregulation of neutrophil chemotaxis and the IL-17 signaling pathway, correlating with reduced expression of Cxcl1 and Cxcl6. Validation of Cxcl1 and Cxcl6 mRNA expression via immunofluorescence and Western blotting supported the RNA-seq findings. Furthermore, a PPI network was constructed to elucidate interactions among the 29 DEGs. Conclusions Through RNA-Seq analysis, we concluded that I-post C may reduce inflammation and suppress the IL-17 signaling pathway, thereby protecting against lung damage caused by LIRI, potentially involving neutrophil extracellular traps.
Collapse
Affiliation(s)
- Liangen Lin
- Department of Emergency, Wenzhou People’s Hospital, The Third Affiliated to Shanghai University, Wenzhou, Zhejiang, China
| | - Congcong Sun
- Department of Scientific Research Center, Wenzhou People’s Hospital, The Third Affiliated to Shanghai University, Wenzhou, Zhejiang, China
| | - Yuanwen Ye
- Department of Emergency, Wenzhou People’s Hospital, The Third Affiliated to Shanghai University, Wenzhou, Zhejiang, China
| | - Peng Zhu
- Department of Emergency, Wenzhou People’s Hospital, The Third Affiliated to Shanghai University, Wenzhou, Zhejiang, China
| | - Keyue Pan
- Department of Emergency, Wenzhou People’s Hospital, The Third Affiliated to Shanghai University, Wenzhou, Zhejiang, China
| | - Linglong Chen
- Department of Emergency, Wenzhou People’s Hospital, The Third Affiliated to Shanghai University, Wenzhou, Zhejiang, China
| |
Collapse
|
6
|
Li Y, Guo N, Zhao Y, Chen J, Zhao J, Bian J, Guo J, Yang C, Zhang X, Huang L. IL-17A activates JAK/STAT signaling to affect drug metabolizing enzymes and transporters in HepaRG cells. Mol Immunol 2024; 175:55-62. [PMID: 39305848 DOI: 10.1016/j.molimm.2024.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 08/13/2024] [Accepted: 09/15/2024] [Indexed: 11/11/2024]
Abstract
The founding family member, Interleukin (IL)-17A, is commonly known as IL-17 and has garnered increasingly attention for proinflammatory functions in autoimmune disorders. Although the effects of IL-17A on hepatic important drug-metabolizing enzymes and transporters (DMETs) expression still remain unclear, it is critical to ascertain owing to the well-established alterations of the drug disposition capacity of the liver occurring during immune imbalance. The present study was designed to explore the effects and mechanisms of IL-17A on DMETs mRNA and protein expression in HepaRG cells by real-time quantitative reverse transcription polymerase chain reaction and Western blot, respectively. It is discovered that IL-17A can inhibit most DMETs mRNA expression (drug-metabolizing enzymes of CYP1A2, CYP3A4, CYP2C9, CYP2C19, GSTA1 and UGT1A1 and transporters of NTCP, OCT1, OATP1B1, BCRP and MDR1) as well as the protein expression of CYP3A4 and CYP2C19, via the janus kinase 2 (JAK2)-signal transducer and activator of transcription 3 (STAT3) signaling pathway. Thus, abnormal regulation of DMETs in IL-17A-mediated immune disorders such as psoriasis may cause alterations in pharmacokinetic processes and may occasionally result in unexpected drug-drug interactions (DDIs) in clinical practice.
Collapse
Affiliation(s)
- Yuanyuan Li
- Department of Pharmacy, People's Hospital of Peking University, Beijing, China; School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Nan Guo
- Department of Pharmacy, People's Hospital of Peking University, Beijing, China; School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yinyu Zhao
- Department of Pharmacy, People's Hospital of Peking University, Beijing, China; Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jiali Chen
- Department of Pharmacy, People's Hospital of Peking University, Beijing, China; School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jinxia Zhao
- Department of Pharmacy, People's Hospital of Peking University, Beijing, China; Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jialu Bian
- Department of Pharmacy, People's Hospital of Peking University, Beijing, China; Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jing Guo
- Department of Pharmacy, People's Hospital of Peking University, Beijing, China
| | - Changqing Yang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaohong Zhang
- Department of Pharmacy, People's Hospital of Peking University, Beijing, China
| | - Lin Huang
- Department of Pharmacy, People's Hospital of Peking University, Beijing, China.
| |
Collapse
|
7
|
MacMahon Copas AN, McComish SF, Petrasca A, McCormack R, Ivers D, Stricker A, Fletcher JM, Caldwell MA. CD4 + T cell-associated cytokines induce a chronic pro-inflammatory phenotype in induced pluripotent stem cell-derived midbrain astrocytes. Glia 2024; 72:2142-2154. [PMID: 39056451 DOI: 10.1002/glia.24601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/05/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024]
Abstract
Astrocytes are mediators of homeostasis but contribute to neuroinflammation in Parkinson's disease (PD). Mounting evidence suggests involvement of peripheral immune cells in PD pathogenesis. Therefore, this study aimed to determine the potential role of peripheral immune secreted cytokines in modulating midbrain astrocyte reactivity. Human iPSC-derived midbrain astrocytes were exposed to 5% and 10% CD4+ T cell conditioned media (CD4CM) for 24 h, 72 h, and 7 days to assess chronic exposure. Additionally, astrocytes were exposed to the Th17 cell cytokine, IL-17A (10 ng/mL), alone and in combination with TNF-α (0.3 ng/mL) to assess potential synergistic effects of both cytokines at 24 h, 72 h, and 7 days. CD4CM induced acute and chronic alterations in midbrain astrocytes. Increased NFκB translocation to the nucleus, increased expression of the pro-inflammatory genes, IL-1β, CXCL10 at 24 h, C3, LCN2, IL-6 at 24 and 48 h, as well as an increase in their release of pro-inflammatory cytokines IL-6 and CXCL10 at both these time points were observed. A synergistic response to the combination of IL-17A and TNF-α on increasing inflammatory gene expression and cytokine release occurred. IL-17A and TNF-α increased intensity of S100β at 24 h, decreased nuclear area and increased circularity of astrocytes at 72 h. A synergistic effect on γH2AX intensity at 72 h and an increase in LDH release at 7 days was observed. Our results demonstrate that IL-17A and TNF-α act synergistically, enhancing midbrain astrocyte reactivity to a similar degree as CD4CM. This highlights the importance of the peripheral immune secreted cytokines in increasing the reactivity status of midbrain astrocytes, implicating their role in PD.
Collapse
Affiliation(s)
- Adina N MacMahon Copas
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College, Dublin, Ireland
| | - Sarah F McComish
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College, Dublin, Ireland
| | - Andreea Petrasca
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Rachel McCormack
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College, Dublin, Ireland
| | - Daniel Ivers
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College, Dublin, Ireland
| | - Anna Stricker
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College, Dublin, Ireland
| | - Jean M Fletcher
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Maeve A Caldwell
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College, Dublin, Ireland
| |
Collapse
|
8
|
Nakanishi T, Mieda K, Kuramoto H, Takegawa D. Effect of interleukin-17A on inflammatory mediator production in interleukin-1β-stimulated human dental pulp fibroblasts. Eur J Oral Sci 2024; 132:e13019. [PMID: 39302740 DOI: 10.1111/eos.13019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 08/26/2024] [Indexed: 09/22/2024]
Abstract
In response to pro-inflammatory cytokines such as interleukin (IL)-1β, dental pulp fibroblasts produce various inflammatory mediators, including IL-6, IL-8, CC chemokine ligand 20 (CCL20), and CXC chemokine ligand 10 (CXCL10), leading to the progression of pulpitis. IL-17/IL-17A (IL-17A) is a pro-inflammatory cytokine secreted by T helper (Th) 17 cells following their recruitment to inflamed sites; however, the roles of IL-17A during pulpitis remain unclear. The purpose of this study was to investigate the effect of IL-17A on IL-6, IL-8, CCL20 and CXCL10 production by human dental pulp fibroblasts (HDPFs) in vitro. IL-17A at a concentration of 100 ng/ml induced the production of 10 times more IL-8 and 4 times more CXCL10, but not IL-6 and CCL20, compared to controls. Co-stimulation of HDPFs with IL-17A and IL-1β synergistically enhanced the production of IL-6, CCL20, IL-8 and CXCL10. IL-1β increased expression of IL-17 receptor/IL-17RA (IL-17R) on HDPFs. Moreover, the cell signal pathways of p38 mitogen-activated protein kinase (MAPK) and nuclear factor-κB (NF-κB) were more potently activated by simultaneous stimulation with IL-17A and IL-1β. These findings suggest that IL-17A participates in the progression of dental pulp inflammation through the enhanced production of inflammatory mediators in HDPFs.
Collapse
Affiliation(s)
- Tadashi Nakanishi
- Department of Regenerative Dental Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Katsuhiro Mieda
- Department of Regenerative Dental Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Hitomi Kuramoto
- Department of Regenerative Dental Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Daisuke Takegawa
- Department of Regenerative Dental Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| |
Collapse
|
9
|
Li S, Wang Y, Xu N, Xie D. Association between C-reactive protein to albumin ratio and subclinical myocardial injury in the general population free from cardiovascular disease. J Cardiothorac Surg 2024; 19:487. [PMID: 39175007 PMCID: PMC11342680 DOI: 10.1186/s13019-024-02988-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/13/2024] [Indexed: 08/24/2024] Open
Abstract
OBJECTIVE The study aimed to examine the role of the C-reactive protein to albumin ratio (CAR) as an inflammatory biomarker in relation to subclinical myocardial injury (SC-MI), addressing the limited knowledge of their association. METHODS The study included 5,949 individuals without cardiovascular disease (CVD) from the National Health and Nutrition Examination Survey. SC-MI was identified through a Cardiac Infarction Injury Score (CIIS) of ≥ 10 units based on a 12-lead electrocardiogram. The study used multivariate logistic regression models, adjusted for potential confounders, to evaluate the relationship between CAR and SC-MI. Subgroup analyses were conducted to substantiate the results, and the non-linear correlation was assessed via restricted cubic spline (RCS) regression. RESULTS The RCS curve showed a significant positive correlation between CAR and SC-MI (P for nonlinear = 0.2496). When adjusted for all confounders, individuals in the highest tertile of CAR exhibited a higher likelihood of SC-MI compared to those in the lowest tertile, with an odds ratio (OR) of 1.21 (95% CI: 1.06-1.39, P for trend = 0.029). A 10-unit increment in CAR was linked to a 3.6% heightened risk of SC-MI [OR = 1.036 (95% CI: 1.006, 1.066)], with this association being more prominent among male adults, non-smokers, married individuals, those without diabetes mellitus, and those with no history of cancer. CONCLUSION The findings of this study suggest a positive correlation between CAR and SC-MI among the US adult population, indicating the potential of CAR in enhancing SC-MI prevention strategies in the general population.
Collapse
Affiliation(s)
- Shuiying Li
- Department of Cardiology, Ningbo Ninth Hospital, No. 68 Xiangbei Road, Jiangbei District, Ningbo, 315000, Zhejiang, China
| | - Yichen Wang
- Department of Hyperbaric Oxygen, Rehabilitation Hospital Affiliated to National Research Center for Rehabilitation Technical Aids, Beijing, 100000, China
| | - Na Xu
- Department of Cardiology, Ningbo Ninth Hospital, No. 68 Xiangbei Road, Jiangbei District, Ningbo, 315000, Zhejiang, China
| | - Daqi Xie
- Department of Cardiology, Ningbo Ninth Hospital, No. 68 Xiangbei Road, Jiangbei District, Ningbo, 315000, Zhejiang, China.
| |
Collapse
|
10
|
Abdelnabi MN, Hassan GS, Shoukry NH. Role of the type 3 cytokines IL-17 and IL-22 in modulating metabolic dysfunction-associated steatotic liver disease. Front Immunol 2024; 15:1437046. [PMID: 39156888 PMCID: PMC11327067 DOI: 10.3389/fimmu.2024.1437046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/12/2024] [Indexed: 08/20/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) comprises a spectrum of liver diseases that span simple steatosis, metabolic dysfunction-associated steatohepatitis (MASH) and fibrosis and may progress to cirrhosis and cancer. The pathogenesis of MASLD is multifactorial and is driven by environmental, genetic, metabolic and immune factors. This review will focus on the role of the type 3 cytokines IL-17 and IL-22 in MASLD pathogenesis and progression. IL-17 and IL-22 are produced by similar adaptive and innate immune cells such as Th17 and innate lymphoid cells, respectively. IL-17-related signaling is upregulated during MASLD resulting in increased chemokines and proinflammatory cytokines in the liver microenvironment, enhanced recruitment of myeloid cells and T cells leading to exacerbation of inflammation and liver disease progression. IL-17 may also act directly by activating hepatic stellate cells resulting in increased fibrosis. In contrast, IL-22 is a pleiotropic cytokine with a dominantly protective signature in MASLD and is currently being tested as a therapeutic strategy. IL-22 also exhibits beneficial metabolic effects and abrogates MASH-related inflammation and fibrosis development via inducing the production of anti-oxidants and anti-apoptotic factors. A sex-dependent effect has been attributed to both cytokines, most importantly to IL-22 in MASLD or related conditions. Altogether, IL-17 and IL-22 are key effectors in MASLD pathogenesis and progression. We will review the role of these two cytokines and cells that produce them in the development of MASLD, their interaction with host factors driving MASLD including sexual dimorphism, and their potential therapeutic benefits.
Collapse
Affiliation(s)
- Mohamed N. Abdelnabi
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Ghada S. Hassan
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Naglaa H. Shoukry
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de médecine, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
11
|
Ning Y, Dou X, Wang Z, Shi K, Wang Z, Ding C, Sang X, Zhong X, Shao M, Han X, Cao G. SIRT3: A potential therapeutic target for liver fibrosis. Pharmacol Ther 2024; 257:108639. [PMID: 38561088 DOI: 10.1016/j.pharmthera.2024.108639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 03/11/2024] [Accepted: 03/19/2024] [Indexed: 04/04/2024]
Abstract
Sirtuin3 (SIRT3) is a nicotinamide adenine dinucleotide (NAD+)-dependent protein deacetylase located in the mitochondria, which mainly regulates the acetylation of mitochondrial proteins. In addition, SIRT3 is involved in critical biological processes, including oxidative stress, inflammation, DNA damage, and apoptosis, all of which are closely related to the progression of liver disease. Liver fibrosis characterized by the deposition of extracellular matrix is a result of long termed or repeated liver damage, frequently accompanied by damaged hepatocytes, the recruitment of inflammatory cells, and the activation of hepatic stellate cells. Based on the functions and pharmacology of SIRT3, we will review its roles in liver fibrosis from three aspects: First, the main functions and pharmacological effects of SIRT3 were investigated based on its structure. Second, the roles of SIRT3 in major cells in the liver were summarized to reveal its mechanism in developing liver fibrosis. Last, drugs that regulate SIRT3 to prevent and treat liver fibrosis were discussed. In conclusion, exploring the pharmacological effects of SIRT3, especially in the liver, may be a potential strategy for treating liver fibrosis.
Collapse
Affiliation(s)
- Yan Ning
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xinyue Dou
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhichao Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Kao Shi
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zeping Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chuan Ding
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xianan Sang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiang Zhong
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Meiyu Shao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xin Han
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China; The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| |
Collapse
|
12
|
Robert M, Miossec P. Structural cell heterogeneity underlies the differential contribution of IL-17A, IL-17F and IL-23 to joint versus skin chronic inflammation. Autoimmun Rev 2024; 23:103529. [PMID: 38492906 DOI: 10.1016/j.autrev.2024.103529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 03/18/2024]
Abstract
The current therapeutic strategy used in immune-mediated inflammatory diseases (IMIDs) primarily targets immune cells or associated-pathways. However, recent evidence suggests that the microenvironment modulates immune cell development and responses. During inflammation, structural cells acquire a pathogenetic phenotype and the interactions with immune cells are often greatly modified. Understanding the importance of these tissue-specific interactions may allow to explain why some biologics are effective in some IMIDs but not in others. The differential effects of interleukin (IL)-17 A, IL-17F and IL-23 in joint versus skin inflammation depends on structural cell heterogeneity. In addition, the sometimes opposite effects of immune/structural cell interactions on the production of these cytokines illustrate the importance of these cells in chronic inflammation, using the examples of rheumatoid arthritis, psoriasis and spondyloarthritis. This review describes these concepts, shows their interests through clinical observations, and finally discusses strategies to optimize therapeutic strategies.
Collapse
Affiliation(s)
- Marie Robert
- Immunogenomics and Inflammation Research Unit, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France
| | - Pierre Miossec
- Immunogenomics and Inflammation Research Unit, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France.
| |
Collapse
|
13
|
Fumagalli S, Ricciardi G, Di Serio C, La Marca G, Pieraccini G, Franci Montorzi R, Santamaria E, Spanalatte G, Marchetti F, Corti G, Pinton L, Marchionni N. Inflammation, mitochondrial dysfunction and physical performance: a possible association in older patients with persistent atrial fibrillation-the results of a preliminary study. Aging Clin Exp Res 2023; 35:2831-2837. [PMID: 37733227 DOI: 10.1007/s40520-023-02558-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 09/01/2023] [Indexed: 09/22/2023]
Abstract
BACKGROUND Atrial fibrillation (AF) is associated with chronic inflammation, a hallmark of ageing process. The aim of this study was to determine interleukin-6 (IL-6)-associated variables, also exploring acylcarnitines, expression of mitochondrial abnormalities. METHODS We evaluated 22 controls and 50 patients with persistent AF. IL-6 and acylcarnitines were measured with ELISA kits and mass spectrometry techniques. RESULTS IL-6 concentration (mean: 3.9 ± 3.1 pg/mL) was lower in controls and increased in AF patients, especially with heart failure. The CHA2DS2-VASc, the MMSE and the SPPB scores were 3.8 ± 1.6, 28 ± 2 and 9.4 ± 2.1. Thirteen acylcanitines correlated with IL-6. At multivariable analysis, IL-6 was directly associated with C4-OH-a short-chain acylcarnitine, fibrinogen and alanine aminotransferase values, and with hyperuricemia. An inverse association existed with calcium concentration and SPPB score. CONCLUSIONS In older AF patients, IL-6 correlated with acylcarnitines and lower physical performance. Alterations in energy production, reduced physical function and inflammation could contribute to frailty development.
Collapse
Affiliation(s)
- Stefano Fumagalli
- Department of Experimental and Clinical Medicine, Geriatric Intensive Care Unit and Geriatric Arrhythmia Unit, University of Florence, Largo G. Brambilla 3, 50134, Florence, Italy.
| | - Giulia Ricciardi
- Department of Experimental and Clinical Medicine, Geriatric Intensive Care Unit and Geriatric Arrhythmia Unit, University of Florence, Largo G. Brambilla 3, 50134, Florence, Italy
| | - Claudia Di Serio
- Department of Experimental and Clinical Medicine, Geriatric Intensive Care Unit and Geriatric Arrhythmia Unit, University of Florence, Largo G. Brambilla 3, 50134, Florence, Italy
| | - Giancarlo La Marca
- Department of Experimental and Clinical Biomedical Sciences 'Mario Serio', Newborn Screening, Clinical Chemistry and Pharmacology Lab, Meyer Children's Hospital and University of Florence, Florence, Italy
| | - Giuseppe Pieraccini
- Department of Health Sciences, CISM Mass Spectrometry Centre, University of Florence, Florence, Italy
| | - Riccardo Franci Montorzi
- Department of Experimental and Clinical Medicine, Geriatric Intensive Care Unit and Geriatric Arrhythmia Unit, University of Florence, Largo G. Brambilla 3, 50134, Florence, Italy
| | - Emanuele Santamaria
- Department of Experimental and Clinical Medicine, Geriatric Intensive Care Unit and Geriatric Arrhythmia Unit, University of Florence, Largo G. Brambilla 3, 50134, Florence, Italy
| | - Giulia Spanalatte
- Department of Experimental and Clinical Medicine, Geriatric Intensive Care Unit and Geriatric Arrhythmia Unit, University of Florence, Largo G. Brambilla 3, 50134, Florence, Italy
| | - Francesca Marchetti
- Department of Experimental and Clinical Medicine, Geriatric Intensive Care Unit and Geriatric Arrhythmia Unit, University of Florence, Largo G. Brambilla 3, 50134, Florence, Italy
| | - Ginevra Corti
- Department of Experimental and Clinical Medicine, Geriatric Intensive Care Unit and Geriatric Arrhythmia Unit, University of Florence, Largo G. Brambilla 3, 50134, Florence, Italy
| | - Laura Pinton
- Department of Experimental and Clinical Medicine, Geriatric Intensive Care Unit and Geriatric Arrhythmia Unit, University of Florence, Largo G. Brambilla 3, 50134, Florence, Italy
| | - Niccolò Marchionni
- Department of Experimental and Clinical Medicine, Geriatric Intensive Care Unit and Geriatric Arrhythmia Unit, University of Florence, Largo G. Brambilla 3, 50134, Florence, Italy
| |
Collapse
|
14
|
Yano A, Yuki S, Shiraishi A, Hakozaki M, Kanno Y, Kimura KI, Uesugi S. Golden berry leaf extract containing withanolides suppresses TNF-α and IL-17 induced IL-6 expression in HeLa Cells. Biosci Biotechnol Biochem 2023; 87:972-980. [PMID: 37279446 DOI: 10.1093/bbb/zbad070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/29/2023] [Indexed: 06/08/2023]
Abstract
Inflammation, characterized by the overexpression of IL-6 in various tissues, has been reported as a symptom of coronavirus disease 2019. In this study, we established an experimental system for overexpression of IL-6 in HeLa cells stimulated by TNF-α and IL-17, along with identification of anti-inflammatory materials and components from local agricultural, forestry, and fishery resources. We constructed a library of extracts from natural sources, of which 111 samples were evaluated for their anti-inflammatory activities. The MeOH extract of Golden Berry (Physalis peruviana L) leaf was found to exhibit strong anti-inflammatory properties (IC50 = 4.97 µg/mL). Preparative chromatography identified two active constituents, 4β-hydroxywithanolide E (4β-HWE) (IC50 = 183 nM) and withanolide E (WE) (IC50 = 65.1 nM). Withanolides are known anti-inflammatory ingredients of Withania somnifera, an Ayurvedic herbal medicine. P. peruviana leaves containing 4β-HWE and WE should be considered as useful natural resources for anti-inflammatory products.
Collapse
Affiliation(s)
- Akira Yano
- Iwate Biotechnology Research Center, Kitakami, Iwate, Japan
| | - Sayaka Yuki
- Iwate Biotechnology Research Center, Kitakami, Iwate, Japan
| | | | | | - Yuko Kanno
- Iwate Biotechnology Research Center, Kitakami, Iwate, Japan
| | - Ken-Ichi Kimura
- Department of Biological Chemistry and Food Science, Faculty of Agriculture, Iwate University, Morioka, Japan
| | - Shota Uesugi
- Iwate Biotechnology Research Center, Kitakami, Iwate, Japan
| |
Collapse
|
15
|
Meyer-Arndt L, Kerkering J, Kuehl T, Infante AG, Paul F, Rosiewicz KS, Siffrin V, Alisch M. Inflammatory Cytokines Associated with Multiple Sclerosis Directly Induce Alterations of Neuronal Cytoarchitecture in Human Neurons. J Neuroimmune Pharmacol 2023; 18:145-159. [PMID: 36862362 PMCID: PMC10485132 DOI: 10.1007/s11481-023-10059-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 01/16/2023] [Indexed: 03/03/2023]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) coined by inflammation and neurodegeneration. The actual cause of the neurodegenerative component of the disease is however unclear. We investigated here the direct and differential effects of inflammatory mediators on human neurons. We used embryonic stem cell-derived (H9) human neuronal stem cells (hNSC) to generate neuronal cultures. Neurons were subsequently treated with tumour necrosis factor alpha (TNFα), interferon gamma (IFNγ), granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin 17A (IL-17A) and interleukin 10 (IL-10) separately or in combination. Immunofluorescence staining and quantitative polymerase chain reaction (qPCR) were used to assess cytokine receptor expression, cell integrity and transcriptomic changes upon treatment. H9-hNSC-derived neurons expressed cytokine receptors for IFNγ, TNFα, IL-10 and IL-17A. Neuronal exposure to these cytokines resulted in differential effects on neurite integrity parameters with a clear decrease for TNFα- and GM-CSF-treated neurons. The combinatorial treatment with IL-17A/IFNγ or IL-17A/TNFα induced a more pronounced effect on neurite integrity. Furthermore, combinatorial treatments with two cytokines induced several key signalling pathways, i.e. NFκB-, hedgehog and oxidative stress signalling, stronger than any of the cytokines alone. This work supports the idea of immune-neuronal crosstalk and the need to focus on the potential role of inflammatory cytokines on neuronal cytoarchitecture and function.
Collapse
Affiliation(s)
- Lil Meyer-Arndt
- Experimental and Clinical Research Center (ECRC), Charité - Universitätsmedizin Berlin und Max Delbrück Center for Molecular Medicine in the Helmholtz Association Berlin, Lindenberger Weg 80, 13125, Berlin, Germany
- Department of Neurology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humbolt-Universität Zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
- NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humbolt-Universität Zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Janis Kerkering
- Experimental and Clinical Research Center (ECRC), Charité - Universitätsmedizin Berlin und Max Delbrück Center for Molecular Medicine in the Helmholtz Association Berlin, Lindenberger Weg 80, 13125, Berlin, Germany
| | - Tess Kuehl
- Experimental and Clinical Research Center (ECRC), Charité - Universitätsmedizin Berlin und Max Delbrück Center for Molecular Medicine in the Helmholtz Association Berlin, Lindenberger Weg 80, 13125, Berlin, Germany
| | - Ana Gil Infante
- Experimental and Clinical Research Center (ECRC), Charité - Universitätsmedizin Berlin und Max Delbrück Center for Molecular Medicine in the Helmholtz Association Berlin, Lindenberger Weg 80, 13125, Berlin, Germany
| | - Friedemann Paul
- Experimental and Clinical Research Center (ECRC), Charité - Universitätsmedizin Berlin und Max Delbrück Center for Molecular Medicine in the Helmholtz Association Berlin, Lindenberger Weg 80, 13125, Berlin, Germany
- NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humbolt-Universität Zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Kamil Sebastian Rosiewicz
- Experimental and Clinical Research Center (ECRC), Charité - Universitätsmedizin Berlin und Max Delbrück Center for Molecular Medicine in the Helmholtz Association Berlin, Lindenberger Weg 80, 13125, Berlin, Germany
| | - Volker Siffrin
- Experimental and Clinical Research Center (ECRC), Charité - Universitätsmedizin Berlin und Max Delbrück Center for Molecular Medicine in the Helmholtz Association Berlin, Lindenberger Weg 80, 13125, Berlin, Germany.
- Department of Neurology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humbolt-Universität Zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany.
| | - Marlen Alisch
- Experimental and Clinical Research Center (ECRC), Charité - Universitätsmedizin Berlin und Max Delbrück Center for Molecular Medicine in the Helmholtz Association Berlin, Lindenberger Weg 80, 13125, Berlin, Germany
| |
Collapse
|
16
|
Elbadawy HM, Khattab A, El-Agamy DS, Eltahir HM, Alhaddad A, Aljohani FD, Almuzaini TM, Abouzied MM, Aldhafiri A. IL-6 at the center of cytokine storm: Circulating inflammation mediators as biomarkers in hospitalized COVID-19 patients. J Clin Lab Anal 2023; 37:e24881. [PMID: 37096731 DOI: 10.1002/jcla.24881] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/11/2023] [Accepted: 03/26/2023] [Indexed: 04/26/2023] Open
Abstract
INTRODUCTION The management of hospitalized COVID-19 patients depends largely on controlling the intensified inflammatory response known as the cytokine storm. Candidate inflammatory cytokines can serve as new biomarkers for the management of hospitalized COVID-19 patients. METHODS Patients (80) were recruited into three groups: room air (RA), oxygen (OX) and mechanical ventilator (MV). Blood analysis was performed for RBC, WBC, Hb, Platelets, serum albumin and creatinine, INR, PTT, and hematocrit. ELISA was used to quantify a panel of inflammatory mediators including GM-SCF, IFN-α, IFNγ, IL-1β, IL-1R, IL-2, IL-2Ra, IL-6, IL-8, IL-10, IL-12p70, IL-13, MCP-1, MIP-1a, and TNF-α. Correlations between laboratory results and the levels of circulating inflammation mediators were investigated. RESULTS Patients on MV had low RBC, Hb, albumin, and HCT and high WBC count, PTT, and INR when compared to RA and OX groups. A statistical positive correlation was found between WBC and the levels of IL-6 and MCP-1. RBCs correlated negatively with IL-6 and IL-10 and positively with IL-8. Higher TNF-α correlated with lower platelet counts while higher levels of IL-1Rα and IL-10 were associated with lower Hb levels. Increases in IFN-γ and TNF-α were indicative of compromised kidney functions as creatinine levels increased significantly. Most significant correlations were found between IL-6 and lab results, showing positive correlation with WBC and INR, and negative correlation with RBC, albumin, and HCT. CONCLUSIONS Having the most significant correlations, IL-6 high levels in mechanically ventilated patients were shown to affect laboratory results, and, therefore, is suggested as a severity biomarker of COVID-19.
Collapse
Affiliation(s)
- Hossein M Elbadawy
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
| | | | - Dina S El-Agamy
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Heba M Eltahir
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
| | - Aisha Alhaddad
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
| | | | | | - Mekky M Abouzied
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Ahmed Aldhafiri
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
| |
Collapse
|
17
|
Zeng S, Rosati E, Saggau C, Messner B, Chu H, Duan Y, Hartmann P, Wang Y, Ma S, Huang WJM, Lee J, Lee SM, Carvalho-Gontijo R, Zhang V, Hoffmann JP, Kolls JK, Raz E, Brenner DA, Kisseleva T, LeibundGut-Landmann S, Bacher P, Stärkel P, Schnabl B. Candida albicans-specific Th17 cell-mediated response contributes to alcohol-associated liver disease. Cell Host Microbe 2023; 31:389-404.e7. [PMID: 36893735 PMCID: PMC10039706 DOI: 10.1016/j.chom.2023.02.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 01/04/2023] [Accepted: 01/31/2023] [Indexed: 03/11/2023]
Abstract
Alcohol-associated liver disease is accompanied by intestinal mycobiome dysbiosis, yet the impacts on liver disease are unclear. We demonstrate that Candida albicans-specific T helper 17 (Th17) cells are increased in circulation and present in the liver of patients with alcohol-associated liver disease. Chronic ethanol administration in mice causes migration of Candida albicans (C. albicans)-reactive Th17 cells from the intestine to the liver. The antifungal agent nystatin decreased C. albicans-specific Th17 cells in the liver and reduced ethanol-induced liver disease in mice. Transgenic mice expressing T cell receptors (TCRs) reactive to Candida antigens developed more severe ethanol-induced liver disease than transgene-negative littermates. Adoptively transferring Candida-specific TCR transgenic T cells or polyclonal C. albicans-primed T cells exacerbated ethanol-induced liver disease in wild-type mice. Interleukin-17 (IL-17) receptor A signaling in Kupffer cells was required for the effects of polyclonal C. albicans-primed T cells. Our findings indicate that ethanol increases C. albicans-specific Th17 cells, which contribute to alcohol-associated liver disease.
Collapse
Affiliation(s)
- Suling Zeng
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA; Department of Medicine, VA San Diego Healthcare System, San Diego, CA, USA
| | - Elisa Rosati
- Institute of Immunology & Institute of Clinical Molecular Biology, Christian-Albrechts Universität zu Kiel and Universitätsklinik Schleswig-Holstein, Kiel, Germany
| | - Carina Saggau
- Institute of Immunology & Institute of Clinical Molecular Biology, Christian-Albrechts Universität zu Kiel and Universitätsklinik Schleswig-Holstein, Kiel, Germany
| | - Berith Messner
- Institute of Immunology & Institute of Clinical Molecular Biology, Christian-Albrechts Universität zu Kiel and Universitätsklinik Schleswig-Holstein, Kiel, Germany
| | - Huikuan Chu
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Yi Duan
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Phillipp Hartmann
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA; Division of Gastroenterology, Hepatology & Nutrition, Rady Children's Hospital San Diego, San Diego, CA, USA
| | - Yanhan Wang
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA; Department of Medicine, VA San Diego Healthcare System, San Diego, CA, USA
| | - Shengyun Ma
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Wendy Jia Men Huang
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jihyung Lee
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Sung Min Lee
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | | | - Vivian Zhang
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Joseph P Hoffmann
- Center for Translational Research in Infection and Inflammation, Department of Pediatrics and Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Jay K Kolls
- Center for Translational Research in Infection and Inflammation, Department of Pediatrics and Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Eyal Raz
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - David A Brenner
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Tatiana Kisseleva
- Department of Surgery, University of California, San Diego, La Jolla, CA, USA
| | - Salomé LeibundGut-Landmann
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland; Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Petra Bacher
- Institute of Immunology & Institute of Clinical Molecular Biology, Christian-Albrechts Universität zu Kiel and Universitätsklinik Schleswig-Holstein, Kiel, Germany
| | - Peter Stärkel
- St. Luc University Hospital, Université Catholique de Louvain, Brussels, Belgium
| | - Bernd Schnabl
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA; Department of Medicine, VA San Diego Healthcare System, San Diego, CA, USA.
| |
Collapse
|
18
|
Di-Iacovo N, Pieroni S, Piobbico D, Castelli M, Scopetti D, Ferracchiato S, Della-Fazia MA, Servillo G. Liver Regeneration and Immunity: A Tale to Tell. Int J Mol Sci 2023; 24:1176. [PMID: 36674692 PMCID: PMC9864482 DOI: 10.3390/ijms24021176] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/28/2022] [Accepted: 12/30/2022] [Indexed: 01/11/2023] Open
Abstract
The physiological importance of the liver is demonstrated by its unique and essential ability to regenerate following extensive injuries affecting its function. By regenerating, the liver reacts to hepatic damage and thus enables homeostasis to be restored. The aim of this review is to add new findings that integrate the regenerative pathway to the current knowledge. An optimal regeneration is achieved through the integration of two main pathways: IL-6/JAK/STAT3, which promotes hepatocyte proliferation, and PI3K/PDK1/Akt, which in turn enhances cell growth. Proliferation and cell growth are events that must be balanced during the three phases of the regenerative process: initiation, proliferation and termination. Achieving the correct liver/body weight ratio is ensured by several pathways as extracellular matrix signalling, apoptosis through caspase-3 activation, and molecules including transforming growth factor-beta, and cyclic adenosine monophosphate. The actors involved in the regenerative process are numerous and many of them are also pivotal players in both the immune and non-immune inflammatory process, that is observed in the early stages of hepatic regeneration. Balance of Th17/Treg is important in liver inflammatory process outcomes. Knowledge of liver regeneration will allow a more detailed characterisation of the molecular mechanisms that are crucial in the interplay between proliferation and inflammation.
Collapse
Affiliation(s)
- Nicola Di-Iacovo
- Department of Medicine and Surgery, University of Perugia, Piazzale L. Severi 1, 06129 Perugia, Italy
| | - Stefania Pieroni
- Department of Medicine and Surgery, University of Perugia, Piazzale L. Severi 1, 06129 Perugia, Italy
| | - Danilo Piobbico
- Department of Medicine and Surgery, University of Perugia, Piazzale L. Severi 1, 06129 Perugia, Italy
| | - Marilena Castelli
- Department of Medicine and Surgery, University of Perugia, Piazzale L. Severi 1, 06129 Perugia, Italy
| | - Damiano Scopetti
- Department of Medicine and Surgery, University of Perugia, Piazzale L. Severi 1, 06129 Perugia, Italy
| | - Simona Ferracchiato
- Department of Medicine and Surgery, University of Perugia, Piazzale L. Severi 1, 06129 Perugia, Italy
| | - Maria Agnese Della-Fazia
- Department of Medicine and Surgery, University of Perugia, Piazzale L. Severi 1, 06129 Perugia, Italy
| | - Giuseppe Servillo
- Department of Medicine and Surgery, University of Perugia, Piazzale L. Severi 1, 06129 Perugia, Italy
- Centro Universitario di Ricerca sulla Genomica Funzionale (C.U.R.Ge.F.), University of Perugia, 06123 Perugia, Italy
| |
Collapse
|
19
|
Hasan N, Hasani NAH, Omar E, Sham FR, Fuad SBSA, Karim MKA, Ibahim MJ. A single targeted gamma-ray irradiation induced an acute modulation of immune cells and related cytokines in EMT6 mouse-bearing tumour model. Cancer Biomark 2023; 38:61-75. [PMID: 37522193 DOI: 10.3233/cbm-220268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
BACKGROUND A complicated interplay between radiation doses, tumour microenvironment (TME), and host immune system is linked to the active participation of immune response. OBJECTIVE The effects of single targeted 2 Gy and 8 Gy gamma-ray irradiations on the immune cell population (lymphocytes, B-cells, T-cells, neutrophils, eosinophils, and macrophages) in EMT6 mouse-bearing tumour models was investigated. METHODS The effects of both irradiation doses in early (96 hours) and acute phase (5 to 11 days) post-irradiation on immune parameters were monitored in blood circulation and TME using flow cytometry. Simultaneously, selected cytokines related to immune cells within the TME were measured using multiplex ELISA. RESULTS A temporary reduction in systemic total white blood count (TWBC) resulted from an early phase (96 hours) of gamma-ray irradiation at 2 Gy and 8 Gy compared to sham control group. No difference was obtained in the acute phase. Neutrophils dominated among other immune cells in TME in sham control group. Eosinophils in TME was significantly increased after 8 Gy treatment in acute phase compared to sham control (p< 0.005). Furthermore, the increment of tumour necrosis (TNF)-α, eotaxin and interleukin (IL)-7 (p< 0.05) in both treatment groups and phases were associated with anti-tumour activities within TME by gamma-ray irradiation. CONCLUSION The temporary changes in immune cell populations within systemic circulation and TME induced by different doses of gamma-ray irradiation correlated with suppression of several pro-tumorigenic cytokines in mouse-bearing EMT6 tumour models.
Collapse
Affiliation(s)
- Nurhaslina Hasan
- Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Selangor, Malaysia
- Faculty of Dentistry, University Teknologi MARA, Sungai Buloh, Selangor, Malaysia
| | | | - Effat Omar
- Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Selangor, Malaysia
| | - Fatihah Ronny Sham
- Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Selangor, Malaysia
| | | | | | | |
Collapse
|
20
|
Altieri A, Piyadasa H, Hemshekhar M, Osawa N, Recksiedler B, Spicer V, Hiemstra PS, Halayko AJ, Mookherjee N. Combination of IL-17A/F and TNF-α uniquely alters the bronchial epithelial cell proteome to enhance proteins that augment neutrophil migration. J Inflamm (Lond) 2022; 19:26. [PMCID: PMC9749191 DOI: 10.1186/s12950-022-00323-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 12/05/2022] [Indexed: 12/16/2022] Open
Abstract
Background The heterodimer interleukin (IL)-17A/F is elevated in the lungs in chronic respiratory disease such as severe asthma, along with the pro-inflammatory cytokine tumor necrosis factor-α (TNF-α). Although IL-17A/F and TNF-α are known to functionally cooperate to exacerbate airway inflammation, proteins altered by their interaction in the lungs are not fully elucidated. Results We used Slow Off-rate Modified Aptamer-based proteomic array to identify proteins that are uniquely and/or synergistically enhanced by concurrent stimulation with IL-17A/F and TNF-α in human bronchial epithelial cells (HBEC). The abundance of 38 proteins was significantly enhanced by the combination of IL-17A/F and TNF-α, compared to either cytokine alone. Four out of seven proteins that were increased > 2-fold were those that promote neutrophil migration; host defence peptides (HDP; Lipocalin-2 (LCN-2) and Elafin) and chemokines (IL-8, GROα). We independently confirmed the synergistic increase of these four proteins by western blots and ELISA. We also functionally confirmed that factors secreted by HBEC stimulated with the combination of IL-17A/F and TNF-α uniquely enhances neutrophil migration. We further showed that PI3K and PKC pathways selectively control IL-17A/F + TNF-α-mediated synergistic production of HDPs LCN-2 and Elafin, but not chemokines IL-8 and GROα. Using a murine model of airway inflammation, we demonstrated enhancement of IL-17A/F, TNF-α, LCN-2 and neutrophil chemokine KC in the lungs, thus corroborating our findings in-vivo. Conclusion This study identifies proteins and signaling mediated by concurrent IL-17A/F and TNF-α exposure in the lungs, relevant to respiratory diseases characterized by chronic inflammation, especially neutrophilic airway inflammation such as severe asthma. Supplementary Information The online version contains supplementary material available at 10.1186/s12950-022-00323-w.
Collapse
Affiliation(s)
- Anthony Altieri
- grid.21613.370000 0004 1936 9609Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB Canada ,grid.21613.370000 0004 1936 9609Department of Immunology, University of Manitoba, Winnipeg, MB Canada
| | - Hadeesha Piyadasa
- grid.21613.370000 0004 1936 9609Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB Canada ,grid.21613.370000 0004 1936 9609Department of Immunology, University of Manitoba, Winnipeg, MB Canada ,grid.168010.e0000000419368956Department of Pathology, School of Medicine, Stanford University, Palo Alto, CA USA
| | - Mahadevappa Hemshekhar
- grid.21613.370000 0004 1936 9609Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB Canada
| | - Natasha Osawa
- grid.21613.370000 0004 1936 9609Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB Canada
| | - Breann Recksiedler
- grid.21613.370000 0004 1936 9609Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB Canada
| | - Victor Spicer
- grid.21613.370000 0004 1936 9609Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB Canada
| | - Pieter S Hiemstra
- grid.10419.3d0000000089452978Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Andrew J Halayko
- grid.21613.370000 0004 1936 9609Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB Canada ,grid.460198.20000 0004 4685 0561Biology of Breathing Group, The Children’s Hospital Research Institute of Manitoba, Winnipeg, MB Canada
| | - Neeloffer Mookherjee
- grid.21613.370000 0004 1936 9609Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB Canada ,grid.21613.370000 0004 1936 9609Department of Immunology, University of Manitoba, Winnipeg, MB Canada ,grid.460198.20000 0004 4685 0561Biology of Breathing Group, The Children’s Hospital Research Institute of Manitoba, Winnipeg, MB Canada
| |
Collapse
|
21
|
Xu T, Jiang Y, Hu X, Yang G, Chen Y, Zhang S, Zhang Q, Zheng L, Xie HQ, Xu L, Zhao B. Effects of the emerging contaminant 1,3,6,8-tetrabromocarbazole on the NF-κB and correlated mechanism in human hepatocellular carcinoma cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 247:114199. [PMID: 36274317 DOI: 10.1016/j.ecoenv.2022.114199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/05/2022] [Accepted: 10/15/2022] [Indexed: 06/16/2023]
Abstract
1,3,6,8-Tetrabromocarbazole (1368-BCZ) is identified as an emerging contaminant that exerts angiogenic effects. Multiple studies indicated there was a positive correlation between angiogenesis and nuclear factor kappa B (NF-κB) activation. While the role of NF-κB in inflammation and apoptosis has been well known, the potential biological effects of 1368-BCZ on NF-κB signaling and related mechanism remain unclear. We, therefore, explored the possible effects of 1368-BCZ on the NF-κB pathway at the gene and protein levels and confirmed that NF-κB activation by 1368-BCZ exposure caused an augmented phosphorylated protein level, induction of NF-κB response element (κBRE)-driven luciferase activity and upregulation of transcriptional level of downstream responsive genes. Although 1368-BCZ did not produce detectable changes in hepatic fibrosis in vivo, it obviously altered the apoptosis in human hepatocellular carcinoma (HepG2) cells. Furthermore, the induction of apoptosis was confirmed by the increased cleaved caspase-3 level. These data revealed the activating effects of 1368-BCZ on NF-κB and its involvement in the underlying mechanisms, providing additional information for toxicology studies of emerging contaminants and introducing a mechanism-based toxicological evaluation of emerging pollutants.
Collapse
Affiliation(s)
- Tong Xu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yu Jiang
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environment Factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Xiaoxu Hu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guanglei Yang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yangsheng Chen
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Songyan Zhang
- Engineering Laboratory of Shenzhen Natural Small Molecule Innovative Drugs, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Qian Zhang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Beijing 100730, China
| | - Liping Zheng
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Heidi Qunhui Xie
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Li Xu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Bin Zhao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| |
Collapse
|
22
|
Ahmad T, Ishaq M, Karpiniec S, Park A, Stringer D, Singh N, Ratanpaul V, Wolfswinkel K, Fitton H, Caruso V, Eri R. Oral Macrocystis pyrifera Fucoidan Administration Exhibits Anti-Inflammatory and Antioxidant Properties and Improves DSS-Induced Colitis in C57BL/6J Mice. Pharmaceutics 2022; 14:2383. [PMID: 36365201 PMCID: PMC9693024 DOI: 10.3390/pharmaceutics14112383] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/27/2022] [Accepted: 10/31/2022] [Indexed: 07/30/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a complex and multifactorial disorder characterised by relapsing and remitting inflammation of the intestinal tract. Oxidative stress (OS) is the result of an imbalance between production and accumulation of reactive oxygen species (ROS), which has been associated with inflammatory responses and implicated in the exacerbation of IBD. Fucoidan, a sulfated polysaccharide from brown seaweed, is a well-known anti-inflammatory agent and emerging evidence indicates that fucoidan extracts from Macrocystis pyrifera (MPF and DP-MPF) may also modulate oxidative stress. This study investigated the impact of fucoidan extracts, MPF and DP-MPF in a dextran sodium sulphate (DSS)-induced mouse model of acute colitis. 3% DSS was administered in C57BL/6J male mice over a period of 7 days, and MPF and DP-MPF were co-administered orally at a dose of 400 mg/kg body weight. Our results indicated that MPF and DP-MPF significantly prevented body weight loss, improved the disease activity index (DAI), restored colon lengths, reduced the wet colon weight, reduced spleen enlargement, and improved the overall histopathological score. Consistent with the reported anti-inflammatory functions, fucoidan extracts, MPF and DP-MPF significantly reduced the colonic levels of myeloperoxidase (MPO), nitric oxide (NO), malondialdehyde (MDA) and increased the levels of antioxidant enzymes, superoxide dismutase (SOD) and catalase (CAT). In addition, MPF and DP-MPF significantly inhibited levels of pro-inflammatory cytokines in colon-derived tissues. Collectively, our results indicate that MPF and DP-MPF exhibited anti-inflammatory and antioxidant effects representing a promising therapeutic strategy for the cure of IBD.
Collapse
Affiliation(s)
- Tauseef Ahmad
- College of Health and Medicine, University of Tasmania, Newnham, TAS 7248, Australia
| | - Muhammad Ishaq
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, TAS 7001, Australia
| | | | - Ahyoung Park
- Marinova Pty Ltd., Cambridge, TAS 7170, Australia
| | | | - Neeraj Singh
- College of Health and Medicine, University of Tasmania, Newnham, TAS 7248, Australia
| | - Vishal Ratanpaul
- School of Science, RMIT University, Bundoora West Campus, Plenty Road, Melbourne, VIC 3083, Australia
| | - Karen Wolfswinkel
- Department of Pathology, Launceston General Hospital (LGH), Launceston, TAS 7250, Australia
| | | | - Vanni Caruso
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, TAS 7001, Australia
- Istituto di Formazione e Ricerca in Scienze Algologiche (ISAL), Torre Pedrera, 47922 Rimini, Italy
| | - Rajaraman Eri
- College of Health and Medicine, University of Tasmania, Newnham, TAS 7248, Australia
- School of Science, RMIT University, Bundoora West Campus, Plenty Road, Melbourne, VIC 3083, Australia
| |
Collapse
|
23
|
Xiang Y, Zhang H, Xu Zhang Z, Yang Qu X, Xia Zhu F. Dihydrosanguinarine based RNA-seq approach couple with network pharmacology attenuates LPS-induced inflammation through TNF/IL-17/PI3K/AKT pathways in mice liver. Int Immunopharmacol 2022; 109:108779. [DOI: 10.1016/j.intimp.2022.108779] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 04/02/2022] [Accepted: 04/13/2022] [Indexed: 01/24/2023]
|
24
|
Batko B. Exploring the Diverse Immune and Genetic Landscape of Psoriatic Arthritis. J Clin Med 2021; 10:jcm10245926. [PMID: 34945224 PMCID: PMC8706996 DOI: 10.3390/jcm10245926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/12/2021] [Accepted: 12/14/2021] [Indexed: 11/16/2022] Open
Abstract
Psoriatic arthritis (PsA) is characterized by delays in diagnosis and modest effect of treatment in terms of joint response. An understanding of molecular pathomechanisms may aid in developing diagnostic and prognostic models. Genetic susceptibility (e.g., HLA class I genes, IL-23-related genes) can be responsible for the pattern of psoriatic manifestations and affinity for tissue involvement. Gene expression analysis indicates an inflammatory profile that is distinct for PsA, but disparate across tissues. This has clinical implications, as for example, dual blockade of IL-17A and IL-17F can lead to superior clinical effects if there is differential expression of IL-17 receptors in tissues. Structural and functional impairment of barrier tissue, including host-microbiome interactions, may be the source of immune activation. Interplay between different cell populations of innate and adaptive immunity is emerging, potentially providing a link between the transition of skin-to-joint disease. Th17 subsets, IL-17A, IL-17F and IL-23 are crucial in PsA pathogenesis, with both clinical and experimental evidence suggesting a differential molecular landscape in cutaneous and articular compartments.
Collapse
Affiliation(s)
- Bogdan Batko
- Department of Rheumatology and Immunology, Faculty of Medicine and Health Sciences, Andrzej Frycz Modrzewski University, 30-705 Krakow, Poland
| |
Collapse
|
25
|
Gender-Related Differences in BMP Expression and Adult Hippocampal Neurogenesis within Joint-Hippocampal Axis in a Rat Model of Rheumatoid Arthritis. Int J Mol Sci 2021; 22:ijms222212163. [PMID: 34830044 PMCID: PMC8620092 DOI: 10.3390/ijms222212163] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/05/2021] [Accepted: 11/07/2021] [Indexed: 11/16/2022] Open
Abstract
BMPs regulate synovial quiescence and adult neurogenesis in the hippocampus in non-stress conditions. However, changes in BMP expression that are induced by inflammation during rheumatoid arthritis (RA) have not yet been reported. Here, we show that signalling with synovial BMPs (BMP-4 and -7) mediates the effect of systemic inflammation on adult neurogenesis in the hippocampus during pristane-induced arthritis (PIA) in Dark Agouti (DA) rats, an animal model of RA. Moreover, we show gender differences in BMP expressions and their antagonists (Noggin and Gremlin) during PIA and their correlations with the clinical course and IL-17A and TNF-α levels in serum. Our results indicate gender differences in the clinical course, where male rats showed earlier onset and earlier recovery but a worse clinical course in the first two phases of the disease (onset and peak), which correlates with the initial increase of serum IL-17A level. The clinical course of the female rats worsened in remission. Their prolonged symptoms could be a reflection of an increased TNF-α level in serum during remission. Synovial inflammation was greater in females in PIA-remission with greater synovial BMP and antagonist expressions. More significant correlations between serum cytokines (IL-17A and TNF-α), and synovial BMPs and their antagonists were found in females than in males. On the other hand, males showed an increase in hippocampal BMP-4 expression during the acute phase, but both genders showed a decrease in antagonist expressions during PIA in general. Both genders showed a decrease in the number of Ki-67+ and SOX-2+ and DCX+ cells and in the ratio of DCX+ to Ki67+ cells in the dentate gyrus during PIA. However, in PIA remission, females showed a faster increase in the number of Ki67+, SOX-2+, and DCX+ cells and a faster increase in the DCX/Ki67 ratio than males. Both genders showed an increase of hippocampal BMP-7 expression during remission, although males constantly showed greater BMP-7 expression at all time points. Our data show that gender differences exist in the BMP expressions in the periphery-hippocampus axis and in the IL-17A and TNF-α levels in serum, which could imply differences in the mechanisms for the onset and progression of the disease, the clinical course severity, and adult neurogenesis with subsequent neurological complications between genders.
Collapse
|
26
|
Analysis of the early response to spinal cord injury identified a key role for mTORC1 signaling in the activation of neural stem progenitor cells. NPJ Regen Med 2021; 6:68. [PMID: 34686684 PMCID: PMC8536777 DOI: 10.1038/s41536-021-00179-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 09/30/2021] [Indexed: 02/07/2023] Open
Abstract
Xenopus laevis are able to regenerate the spinal cord during larvae stages through the activation of neural stem progenitor cells (NSPCs). Here we use high-resolution expression profiling to characterize the early transcriptome changes induced after spinal cord injury, aiming to identify the signals that trigger NSPC proliferation. The analysis delineates a pathway that starts with a rapid and transitory activation of immediate early genes, followed by migration processes and immune response genes, the pervasive increase of NSPC-specific ribosome biogenesis factors, and genes involved in stem cell proliferation. Western blot and immunofluorescence analysis showed that mTORC1 is rapidly and transiently activated after SCI, and its pharmacological inhibition impairs spinal cord regeneration and proliferation of NSPC through the downregulation of genes involved in the G1/S transition of cell cycle, with a strong effect on PCNA. We propose that the mTOR signaling pathway is a key player in the activation of NPSCs during the early steps of spinal cord regeneration.
Collapse
|
27
|
Paiva IA, Badolato-Corrêa J, Familiar-Macedo D, de-Oliveira-Pinto LM. Th17 Cells in Viral Infections-Friend or Foe? Cells 2021; 10:cells10051159. [PMID: 34064728 PMCID: PMC8151546 DOI: 10.3390/cells10051159] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/20/2021] [Accepted: 04/28/2021] [Indexed: 12/12/2022] Open
Abstract
Th17 cells are recognized as indispensable in inducing protective immunity against bacteria and fungi, as they promote the integrity of mucosal epithelial barriers. It is believed that Th17 cells also play a central role in the induction of autoimmune diseases. Recent advances have evaluated Th17 effector functions during viral infections, including their critical role in the production and induction of pro-inflammatory cytokines and in the recruitment and activation of other immune cells. Thus, Th17 is involved in the induction both of pathogenicity and immunoprotective mechanisms seen in the host's immune response against viruses. However, certain Th17 cells can also modulate immune responses, since they can secrete immunosuppressive factors, such as IL-10; these cells are called non-pathogenic Th17 cells. Here, we present a brief review of Th17 cells and highlight their involvement in some virus infections. We cover these notions by highlighting the role of Th17 cells in regulating the protective and pathogenic immune response in the context of viral infections. In addition, we will be describing myocarditis and multiple sclerosis as examples of immune diseases triggered by viral infections, in which we will discuss further the roles of Th17 cells in the induction of tissue damage.
Collapse
|
28
|
He T, Liu C, Li M, Wang M, Liu N, Zhang D, Han S, Li W, Chen S, Yuan R, Huang J. Integrating non-targeted metabolomics and toxicology networks to study the mechanism of Esculentoside A-induced hepatotoxicity in rats. J Biochem Mol Toxicol 2021; 35:1-15. [PMID: 33788351 DOI: 10.1002/jbt.22761] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 12/11/2020] [Accepted: 03/02/2021] [Indexed: 11/06/2022]
Abstract
Esculentoside A (EsA) is a kind of triterpenoid saponins from the root tuber of Phytolacca acinosa Roxb. It has extensive medicinal activity, such as antibacterial, anti-inflammatory, immune regulation, and cell proliferation inhibition. However, some researches suggested that EsA can cause hepatotoxicity, whose mechanism is not precise. To ensure the safety and reliability in the clinical use of Phytolacca acinosa Roxb., it is necessary to establish a rapid and accurate method to evaluate the toxicity, analyze and verify the toxicity mechanism of EsA. Therefore, this research explored the mechanism of hepatotoxicity induced by EsA in rats and analyzed endogenous metabolites' changes in rat plasma by combining network toxicology with non-targeted metabolomics. We obtained 58 critical targets of EsA induced hepatotoxicity in rats based on the strategy of network toxicology, including albumin, mitogen-activated protein kinase 1, Caspase-3, etc. Many important pathways were obtained by Kyoto Encyclopedia of Genes and Genomes enrichment analysis, such as HIF-1 signaling pathway, TNF signaling pathway, IL-17 signaling pathway, and other concerning pathways. Sixteen biomarkers, including 5-hydroxykynurenamine, N-acetylserotonin, palmitic acid, etc., were screened from rat plasma using Ultra-performance liquid chromatography coupled with quadrupole time of flight mass spectrometry (UPLC-Q-TOF/MS), mainly involve Glycerophospholipid metabolism, Tryptophan metabolism, and other metabolic pathways. Further analysis showed that EsA may induce liver injury by activating oxidative stress and energy metabolism disorders, triggering inflammation and apoptosis.
Collapse
Affiliation(s)
- Tao He
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing, China
| | - Chuanxin Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing, China
| | - Mengyu Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing, China
| | - Mingshuang Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing, China
| | - Ning Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing, China
| | - Dan Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing, China
| | - Shuang Han
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing, China
| | - Wenxian Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing, China
| | - Shilin Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing, China
| | - Ruijuan Yuan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing, China
| | - Jianmei Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing, China
| |
Collapse
|
29
|
Miossec P. Local and systemic effects of IL-17 in joint inflammation: a historical perspective from discovery to targeting. Cell Mol Immunol 2021; 18:860-865. [PMID: 33692481 PMCID: PMC7943939 DOI: 10.1038/s41423-021-00644-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/18/2021] [Indexed: 12/14/2022] Open
Abstract
The role of IL-17 in many inflammatory and autoimmune diseases is now well established, with three currently registered anti-IL-17-targeted therapies. This story has taken place over a period of 20 years and led to the demonstration that a T cell product could regulate, and often amplify, the inflammatory response. The first results described the contribution of IL-17 to local features in arthritis. Then, understanding was extended to its systemic effects, with a focus on cardiovascular aspects. This review provides a historical perspective of these discoveries focused on arthritis, which started in 1995, followed 10 years later by the description of Th17 cells. Today, IL-17 inhibitors for three chronic inflammatory diseases have been registered. More options are now being tested in ongoing and future clinical trials. Inhibitors of IL-17 family members and Th17 cells ranging from antibodies to small molecules are under active development. The identification of patients with IL-17-driven disease is a key target for the improved selection of patients expected to have a strongly positive response.
Collapse
Affiliation(s)
- Pierre Miossec
- Department of Clinical Immunology and Rheumatology and the Immunogenomics and Inflammation Research Unit, University of Lyon, Hôpital Edouard Herriot, Lyon, France.
| |
Collapse
|
30
|
The Mediation of miR-34a/miR-449c for Immune Cytokines in Acute Cold/Heat-Stressed Broiler Chicken. Animals (Basel) 2020; 10:ani10112168. [PMID: 33233727 PMCID: PMC7699918 DOI: 10.3390/ani10112168] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/14/2020] [Accepted: 11/17/2020] [Indexed: 12/21/2022] Open
Abstract
Simple Summary In the intensive and scale poultry industry, the level of heat stress (HS) directly affects the growth, development, and production performance of poultry. To alleviate the adverse effects of stress in broilers, microRNA (miRNA) was regarded as a potential regulator of immune cytokines. In this study, through the sequencing analysis of spleens after cold/heat stress, we found that 33 and 37 miRNA were differentially expressed in the heat stress group compared with the normal (NS) group and cold stress (CS) group, respectively. The differential miRNA were mainly involved in biological processes such as the cytokine–cytokine receptor interaction. To further understand the miRNA-mediated effect of heat stress on the immune level of chickens, we selected miR-34a and miR-449c as the research objects, predicted and verified that interleukin 2 (IL-2) and interleukin 12α (IL-12α) were the target genes of miR-34a and miR-449c. Coupled with the analysis of the expression of other cytokines, we found that miRNA could change the expression of immune cytokines directly or indirectly. This discovery provides a new insight into the mediation of miRNA for immune cytokines in acute cold/heat stressed broiler chicken. Abstract An increasing amount of evidence has revealed that microRNAs (miRNAs) participated in immune regulation and reaction to acute cold and heat stresses. As a new type of post-transcriptional regulatory factor, miRNA has received widespread attention; However, the specific mechanism used for this regulation still needs to be determined. In this study, thirty broilers at the same growth period were divided into three groups and treated with different temperature and humidity of CS (10–15 °C and 90% Relative Humidity (RH)), HS (39 °C and 90% RH), and NS (26 °C and 50–60% RH) respectively. After 6 h, splenic tissues were collected from all study groups. miRNA sequencing was performed to identify the differentially expressed miRNAs (DEMs) between HS, CS, and NS. We found 33, 37, and 7 DEMs in the HS-NS, HS-CS, CS-NS group. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis showed that DEMs were significantly enriched in cytokine–cytokine receptor interaction and functioned as the cellular responders to stress. We chose two miRNA, miR-34a and miR-449c, from the same family and differential expressed in HS-CS and HS-NS group, as the research objects to predict and verify the target genes. The dual-luciferase reporter assay and quantitative real-time PCR (qRT-PCR) confirmed that two cytokines, IL-2 and IL-12α, were the direct target genes of miR-34a and miR-449c. To further understand the mediation mechanism of miRNAs in acute cold/heat-stressed broiler chicken, a splenic cytokines profile was constructed. The results showed that IL-1β was strongly related to acute heat stress in broiler chicken, and from this we predicted that the increased expression of IL-1β might promote the expression of miR-34a, inducing the upregulation of interferon-γ (INF-γ) and IL-17. Our finds have laid a theoretical foundation for the breeding of poultry resistance and alleviation of the adverse effects of stress.
Collapse
|
31
|
Miossec P. [Proinflammatory cytokines and cardio-vascular risk: From myocardial infarction tocytokine storm of COVID-19]. BULLETIN DE L ACADEMIE NATIONALE DE MEDECINE 2020; 205:43-48. [PMID: 33199926 PMCID: PMC7657003 DOI: 10.1016/j.banm.2020.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 10/16/2020] [Indexed: 11/16/2022]
Abstract
The mechanisms of inflammatory diseases involve the key inflammatory cytokines IL-1, TNF, IL-6, and IL-17 which are now therapeutic targets with biotherapies. They contribute to the local manifestations of clinically different diseases. In addition to these local aspects, these cytokines have systemic effects from their action on the liver, muscle, adipose tissue and the cardiovascular system. All these diseases have in common an increase in cardiovascular risk. In the general population, the same concepts are applicable, as shown by the link between an even modest rise in CRP and cardiovascular risk. More recently, the cytokine storm of severe forms of COVID-19 has shown that synergistic interactions between cytokines first described in vitro are further amplified in the clinical picture with multiple and severe impairment of key organs. In these chronic and acute contexts, control of inflammation by targeting cytokines is a new vascular treatment option, with already important results for IL-1.
Collapse
Affiliation(s)
- P Miossec
- Unité d'immunologie clinique, département d'immunologie et de rhumatologie, et laboratoire immunogénomique et inflammation EA 4130, université de Lyon, Lyon, France
| |
Collapse
|
32
|
Miossec P. Synergy Between Cytokines and Risk Factors in the Cytokine Storm of COVID-19: Does Ongoing Use of Cytokine Inhibitors Have a Protective Effect? Arthritis Rheumatol 2020; 72:1963-1966. [PMID: 32725790 DOI: 10.1002/art.41458] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/08/2020] [Accepted: 07/15/2020] [Indexed: 12/18/2022]
|
33
|
Álvarez-Coiradas E, Munteanu CR, Díaz-Sáez L, Pazos A, Huber KVM, Loza MI, Domínguez E. Discovery of novel immunopharmacological ligands targeting the IL-17 inflammatory pathway. Int Immunopharmacol 2020; 89:107026. [PMID: 33045560 DOI: 10.1016/j.intimp.2020.107026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/02/2020] [Accepted: 09/16/2020] [Indexed: 01/25/2023]
Abstract
Interleukin 17 (IL-17) is a proinflammatory cytokine that acts as an immune checkpoint for several autoimmune diseases. Therapeutic neutralizing antibodies that target this cytokine have demonstrated clinical efficacy in psoriasis. However, biologics have limitations such as their high cost and their lack of oral bioavailability. Thus, it is necessary to expand the therapeutic options for this IL-17A/IL-17RA pathway, applying novel drug discovery methods to find effective small molecules. In this work, we combined biophysical and cell-based assays with structure-based docking to find novel ligands that target this pathway. First, a virtual screening of our chemical library of 60000 compounds was used to identify 67 potential ligands of IL-17A and IL-17RA. We developed a biophysical label-free binding assay to determine interactions with the extracellular domain of IL-17RA. Two molecules (CBG040591 and CBG060392) with quinazolinone and pyrrolidinedione chemical scaffolds, respectively, were confirmed as ligands of IL-17RA with micromolar affinity. The anti-inflammatory activity of these ligands as cytokine-release inhibitors was evaluated in human keratinocytes. Both ligands inhibited the release of chemokines mediated by IL-17A, with an IC50 of 20.9 ± 12.6 μM and 23.6 ± 11.8 μM for CCL20 and an IC50 of 26.7 ± 13.1 μM and 45.3 ± 13.0 μM for CXCL8. Hence, they blocked IL-17A proinflammatory activity, which is consistent with the inhibition of the signalling of the IL-17A receptor by ligand CBG060392. Therefore, we identified two novel immunopharmacological ligands targeting the IL-17A/IL-17RA pathway with antiinflammatory efficacy that can be promising tools for a drug discovery program for psoriasis.
Collapse
Affiliation(s)
- Elia Álvarez-Coiradas
- Biofarma Research Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Avenida de Barcelona s/n, 15782 Santiago de Compostela, Spain
| | - Cristian R Munteanu
- RNASA-IMEDIR, Computer Science Faculty, CITIC, Universidade da Coruña, A Coruña, 15007, Spain; Biomedical Research Institute of A Coruña (INIBIC), University Hospital Complex of A Coruña (CHUAC), A Coruña 15006, Spain
| | - Laura Díaz-Sáez
- Structural Genomics Consortium & Target Discovery Institute, University of Oxford, Nuffield Department of Medicine, Old Road Campus, Oxford OX3 7DQ & OX3 7FZ, UK
| | - Alejandro Pazos
- RNASA-IMEDIR, Computer Science Faculty, CITIC, Universidade da Coruña, A Coruña, 15007, Spain; Biomedical Research Institute of A Coruña (INIBIC), University Hospital Complex of A Coruña (CHUAC), A Coruña 15006, Spain
| | - Kilian V M Huber
- Structural Genomics Consortium & Target Discovery Institute, University of Oxford, Nuffield Department of Medicine, Old Road Campus, Oxford OX3 7DQ & OX3 7FZ, UK
| | - María Isabel Loza
- Biofarma Research Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Avenida de Barcelona s/n, 15782 Santiago de Compostela, Spain.
| | - Eduardo Domínguez
- Biofarma Research Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Avenida de Barcelona s/n, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
34
|
Najjar S, Najjar A, Chong DJ, Pramanik BK, Kirsch C, Kuzniecky RI, Pacia SV, Azhar S. Central nervous system complications associated with SARS-CoV-2 infection: integrative concepts of pathophysiology and case reports. J Neuroinflammation 2020; 17:231. [PMID: 32758257 PMCID: PMC7406702 DOI: 10.1186/s12974-020-01896-0] [Citation(s) in RCA: 168] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a highly infectious pandemic caused by a novel coronavirus called severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). It frequently presents with unremitting fever, hypoxemic respiratory failure, and systemic complications (e.g., gastrointestinal, renal, cardiac, and hepatic involvement), encephalopathy, and thrombotic events. The respiratory symptoms are similar to those accompanying other genetically related beta-coronaviruses (CoVs) such as severe acute respiratory syndrome CoV (SARS-CoV) and Middle East Respiratory Syndrome CoV (MERS-CoV). Hypoxemic respiratory symptoms can rapidly progress to Acute Respiratory Distress Syndrome (ARDS) and secondary hemophagocytic lymphohistiocytosis, leading to multi-organ system dysfunction syndrome. Severe cases are typically associated with aberrant and excessive inflammatory responses. These include significant systemic upregulation of cytokines, chemokines, and pro-inflammatory mediators, associated with increased acute-phase proteins (APPs) production such as hyperferritinemia and elevated C-reactive protein (CRP), as well as lymphocytopenia. The neurological complications of SARS-CoV-2 infection are high among those with severe and critical illnesses. This review highlights the central nervous system (CNS) complications associated with COVID-19 attributed to primary CNS involvement due to rare direct neuroinvasion and more commonly secondary CNS sequelae due to exuberant systemic innate-mediated hyper-inflammation. It also provides a theoretical integration of clinical and experimental data to elucidate the pathogenesis of these disorders. Specifically, how systemic hyper-inflammation provoked by maladaptive innate immunity may impair neurovascular endothelial function, disrupt BBB, activate CNS innate immune signaling pathways, and induce para-infectious autoimmunity, potentially contributing to the CNS complications associated with SARS-CoV-2 infection. Direct viral infection of the brain parenchyma causing encephalitis, possibly with concurrent neurovascular endotheliitis and CNS renin angiotensin system (RAS) dysregulation, is also reviewed.
Collapse
Affiliation(s)
- Souhel Najjar
- Department of Neurology, Zucker School of Medicine at Hofstra/Northwell, Lenox Hill Hospital, New York, NY, USA.
- Department of Neurology, Zucker School of Medicine at Hofstra/Northwell, North Shore University Hospital, Manhasset, NY, USA.
| | - Amanda Najjar
- Department of Neurology, Zucker School of Medicine at Hofstra/Northwell, Lenox Hill Hospital, New York, NY, USA
- Ferkauf Graduate School of Psychology, Yeshiva University, Bronx, NY, USA
| | - Derek J Chong
- Department of Neurology, Zucker School of Medicine at Hofstra/Northwell, Lenox Hill Hospital, New York, NY, USA
| | - Bidyut K Pramanik
- Department of Radiology, Zucker School of Medicine at Hofstra/Northwell, Lenox Hill Hospital, New York, NY, USA
| | - Claudia Kirsch
- Department of Radiology, Zucker School of Medicine at Hofstra/Northwell, North Shore University Hospital, Manhasset, NY, USA
| | - Ruben I Kuzniecky
- Department of Neurology, Zucker School of Medicine at Hofstra/Northwell, Lenox Hill Hospital, New York, NY, USA
| | - Steven V Pacia
- Department of Neurology, Zucker School of Medicine at Hofstra/Northwell, Lenox Hill Hospital, New York, NY, USA
- Department of Neurology, Zucker School of Medicine at Hofstra/Northwell, North Shore University Hospital, Manhasset, NY, USA
| | - Salman Azhar
- Department of Neurology, Zucker School of Medicine at Hofstra/Northwell, Lenox Hill Hospital, New York, NY, USA
| |
Collapse
|
35
|
Erikson K, Tuominen H, Vakkala M, Liisanantti JH, Karttunen T, Syrjälä H, Ala-Kokko TI. Brain tight junction protein expression in sepsis in an autopsy series. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2020; 24:385. [PMID: 32600371 PMCID: PMC7325252 DOI: 10.1186/s13054-020-03101-3] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 06/17/2020] [Indexed: 12/28/2022]
Abstract
Background Neuroinflammation often develops in sepsis along with increasing permeability of the blood-brain barrier (BBB), which leads to septic encephalopathy. The barrier is formed by tight junction structures between the cerebral endothelial cells. We investigated the expression of tight junction proteins related to endothelial permeability in brain autopsy specimens in critically ill patients deceased with sepsis and analyzed the relationship of BBB damage with measures of systemic inflammation and systemic organ dysfunction. Methods The case series included all (385) adult patients deceased due to sepsis in the years 2007–2015 with available brain specimens taken at autopsy. Specimens were categorized according to anatomical location (cerebrum, cerebellum). The immunohistochemical stainings were performed for occludin, ZO-1, and claudin. Patients were categorized as having BBB damage if there was no expression of occludin in the endothelium of cerebral microvessels. Results Brain tissue samples were available in 47 autopsies, of which 38% (18/47) had no expression of occludin in the endothelium of cerebral microvessels, 34% (16/47) developed multiple organ failure before death, and 74.5% (35/47) had septic shock. The deceased with BBB damage had higher maximum SOFA scores (16 vs. 14, p = 0.04) and more often had procalcitonin levels above 10 μg/L (56% vs. 28%, p = 0.045) during their ICU stay. BBB damage in the cerebellum was more common in cases with C-reactive protein (CRP) above 100 mg/L as compared with CRP less than 100 (69% vs. 25%, p = 0.025). Conclusions In fatal sepsis, damaged BBB defined as a loss of cerebral endothelial expression of occludin is related with severe organ dysfunction and systemic inflammation.
Collapse
Affiliation(s)
- Kristo Erikson
- Division of Intensive Care Medicine, Department of Anesthesiology, Research Group of Surgery, Anesthesiology and Intensive Care Medicine, Oulu University Hospital, Medical Research Center Oulu, University of Oulu, Oulu, Finland. .,Department of Anesthesiology, Intensive Care Center, North Estonia Medical Centre, Tallinn, Estonia.
| | - Hannu Tuominen
- Department of Anesthesiology, Intensive Care Center, North Estonia Medical Centre, Tallinn, Estonia
| | - Merja Vakkala
- Division of Intensive Care Medicine, Department of Anesthesiology, Research Group of Surgery, Anesthesiology and Intensive Care Medicine, Oulu University Hospital, Medical Research Center Oulu, University of Oulu, Oulu, Finland
| | - Janne Henrik Liisanantti
- Division of Intensive Care Medicine, Department of Anesthesiology, Research Group of Surgery, Anesthesiology and Intensive Care Medicine, Oulu University Hospital, Medical Research Center Oulu, University of Oulu, Oulu, Finland
| | - Tuomo Karttunen
- Department of Anesthesiology, Intensive Care Center, North Estonia Medical Centre, Tallinn, Estonia
| | - Hannu Syrjälä
- Department of Pathology and Department of Infection Control, Oulu University Hospital, Medical Research Center Oulu, University of Oulu, Oulu, Finland
| | - Tero Ilmari Ala-Kokko
- Division of Intensive Care Medicine, Department of Anesthesiology, Research Group of Surgery, Anesthesiology and Intensive Care Medicine, Oulu University Hospital, Medical Research Center Oulu, University of Oulu, Oulu, Finland
| |
Collapse
|
36
|
Miossec P. Understanding the cytokine storm during COVID-19: Contribution of preexisting chronic inflammation. Eur J Rheumatol 2020; 7:S97-S98. [PMID: 32412405 DOI: 10.5152/eurjrheum.2020.2062] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 02/01/2023] Open
Affiliation(s)
- Pierre Miossec
- Departments of Immunology and Rheumatology, University of Lyon, Lyon, France
| |
Collapse
|
37
|
Li J, Zeng M, Yan K, Yang Y, Li H, Xu X. IL-17 promotes hepatocellular carcinoma through inhibiting apoptosis induced by IFN-γ. Biochem Biophys Res Commun 2020; 522:525-531. [DOI: 10.1016/j.bbrc.2019.11.134] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 11/20/2019] [Indexed: 02/06/2023]
|
38
|
Hathaway-Schrader JD, Poulides NA, Carson MD, Kirkpatrick JE, Warner AJ, Swanson BA, Taylor EV, Chew ME, Reddy SV, Liu B, Westwater C, Novince CM. Specific Commensal Bacterium Critically Regulates Gut Microbiota Osteoimmunomodulatory Actions During Normal Postpubertal Skeletal Growth and Maturation. JBMR Plus 2020; 4:e10338. [PMID: 32161843 PMCID: PMC7059828 DOI: 10.1002/jbm4.10338] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 01/06/2020] [Accepted: 01/07/2020] [Indexed: 12/19/2022] Open
Abstract
The commensal gut microbiota critically regulates immunomodulatory processes that influence normal skeletal growth and maturation. However, the influence of specific microbes on commensal gut microbiota osteoimmunoregulatory actions is unknown. We have shown previously that the commensal gut microbiota enhances TH17/IL17A immune response effects in marrow and liver that have procatabolic/antianabolic actions in the skeleton. Segmented filamentous bacteria (SFB), a specific commensal gut bacterium within phylum Firmicutes, potently induces TH17/IL17A‐mediated immunity. The study purpose was to delineate the influence of SFB on commensal gut microbiota immunomodulatory actions regulating normal postpubertal skeletal development. Two murine models were utilized: SFB‐monoassociated mice versus germ‐free (GF) mice and specific‐pathogen‐free (SPF) mice +/− SFB. SFB colonization was validated by 16S rDNA analysis, and SFB‐induced TH17/IL17A immunity was confirmed by upregulation of Il17a in ileum and IL17A in serum. SFB‐colonized mice had an osteopenic trabecular bone phenotype, which was attributed to SFB actions suppressing osteoblastogenesis and enhancing osteoclastogenesis. Intriguingly, SFB‐colonized mice had increased expression of proinflammatory chemokines and acute‐phase reactants in the liver. Lipocalin‐2 (LCN2), an acute‐phase reactant and antimicrobial peptide, was substantially elevated in the liver and serum of SFB‐colonized mice, which supports the notion that SFB regulation of commensal gut microbiota osteoimmunomodulatory actions are mediated in part through a gut–liver–bone axis. Proinflammatory TH17 and TH1 cells were increased in liver‐draining lymph nodes of SFB‐colonized mice, which further substantiates that SFB osteoimmune‐response effects may be mediated through the liver. SFB‐induction of Il17a in the gut and Lcn2 in the liver resulted in increased circulating levels of IL17A and LCN2. Recognizing that IL17A and LCN2 support osteoclastogenesis/suppress osteoblastogenesis, SFB actions impairing postpubertal skeletal development appear to be mediated through immunomodulatory effects in both the gut and liver. This research reveals that specific microbes critically impact commensal gut microbiota immunomodulatory actions regulating normal postpubertal skeletal growth and maturation. © 2020 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Jessica D Hathaway-Schrader
- Department of Oral Health Sciences College of Dental Medicine, Medical University of South Carolina Charleston SC USA.,Department of Pediatrics-Division of Endocrinology College of Medicine, Medical University of South Carolina Charleston SC USA
| | - Nicole A Poulides
- Department of Oral Health Sciences College of Dental Medicine, Medical University of South Carolina Charleston SC USA.,Department of Pediatrics-Division of Endocrinology College of Medicine, Medical University of South Carolina Charleston SC USA
| | - Matthew D Carson
- Department of Oral Health Sciences College of Dental Medicine, Medical University of South Carolina Charleston SC USA.,Department of Pediatrics-Division of Endocrinology College of Medicine, Medical University of South Carolina Charleston SC USA
| | - Joy E Kirkpatrick
- Department of Oral Health Sciences College of Dental Medicine, Medical University of South Carolina Charleston SC USA.,Department of Drug Discovery & Biomedical Sciences College of Pharmacy, Medical University of South Carolina Charleston SC USA
| | - Amy J Warner
- Department of Oral Health Sciences College of Dental Medicine, Medical University of South Carolina Charleston SC USA.,Department of Pediatrics-Division of Endocrinology College of Medicine, Medical University of South Carolina Charleston SC USA
| | - Brooks A Swanson
- Department of Oral Health Sciences College of Dental Medicine, Medical University of South Carolina Charleston SC USA.,Department of Pediatrics-Division of Endocrinology College of Medicine, Medical University of South Carolina Charleston SC USA
| | - Eliza V Taylor
- Department of Oral Health Sciences College of Dental Medicine, Medical University of South Carolina Charleston SC USA
| | - Michael E Chew
- Department of Oral Health Sciences College of Dental Medicine, Medical University of South Carolina Charleston SC USA
| | - Sakamuri V Reddy
- Department of Pediatrics-Division of Endocrinology College of Medicine, Medical University of South Carolina Charleston SC USA
| | - Bei Liu
- Department of Microbiology and Immunology College of Medicine, Medical University of South Carolina Charleston SC USA
| | - Caroline Westwater
- Department of Oral Health Sciences College of Dental Medicine, Medical University of South Carolina Charleston SC USA.,Department of Microbiology and Immunology College of Medicine, Medical University of South Carolina Charleston SC USA
| | - Chad M Novince
- Department of Oral Health Sciences College of Dental Medicine, Medical University of South Carolina Charleston SC USA.,Department of Pediatrics-Division of Endocrinology College of Medicine, Medical University of South Carolina Charleston SC USA
| |
Collapse
|
39
|
Noack M, Beringer A, Miossec P. Additive or Synergistic Interactions Between IL-17A or IL-17F and TNF or IL-1β Depend on the Cell Type. Front Immunol 2019; 10:1726. [PMID: 31396230 PMCID: PMC6664074 DOI: 10.3389/fimmu.2019.01726] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/09/2019] [Indexed: 01/15/2023] Open
Abstract
Background: IL-17A has effects on several cell types and is a therapeutic target in several inflammatory diseases. IL-17F shares 50% homology and biological activities with IL-17A. It is now of interest to target both cytokines. The objective was to compare the IL-17A and IL-17F effect on cytokine production by RA synoviocytes, and to extend to other cells. Methods: Cells (RA synoviocytes, psoriasis skin fibroblasts, endothelial cells, myoblasts, and hepatocytes) were cultured in the presence or not of: IL-17A, IL-17F, TNF, IL-1β alone or their combinations, IL-17A/TNF, IL-17A/IL-1β, IL-17A/TNF/IL-1β, IL-17F/TNF, IL-17F/IL-1β, and IL-17F/TNF/IL-1β. All experiments were performed in parallel to reduce variability. After 48 h, supernatants were recovered and IL-6 and IL-8 levels were measured by ELISA. Results: IL-17A and IL-17F alone increased significantly IL-6 and IL-8 productions by synoviocytes, with a stronger effect for IL-17A. For IL-6 production, TNF or IL-1β alone had the largest effect on myoblasts (5-fold increase), while for IL-8 production, it was on skin fibroblasts (5-fold increase). The IL-17A/TNF synergistic increase was observed on all cells for IL-6; and for IL-8, except for endothelial cells. For IL-17F/TNF, except with endothelial cells, a synergistic effect was also observed, but less powerful than with IL-17A/TNF. IL-17A/IL-1β or IL-17F/IL-1β effect was cell-type dependent, with an additive effect for synoviocytes (1.6 and 2-fold increase, respectively for IL-6, and 1.8 and 2-fold increase, respectively for IL-8) and a synergistic effect for hepatocytes (3.8 and 4.2-fold increase, respectively for IL-6, and 6 and 2-fold increase, respectively for IL-8). The three-cytokine combination induced an additive effect for synoviocytes and a synergistic effect for skin fibroblasts. Conclusion: IL-17A and IL-17F acted similarly by inducing pro-inflammatory cytokine secretion, with a stronger response intensity with IL-17A. Their activities were potentiated by the combination with TNF and IL-1β, with an effect dependent on the cell type.
Collapse
Affiliation(s)
- Mélissa Noack
- Immunogenomics and Inflammation Research Unit, EA 4130, Edouard Herriot Hospital, Hospices Civils de Lyon and University Claude Bernard Lyon 1, Lyon, France
| | - Audrey Beringer
- Immunogenomics and Inflammation Research Unit, EA 4130, Edouard Herriot Hospital, Hospices Civils de Lyon and University Claude Bernard Lyon 1, Lyon, France
| | - Pierre Miossec
- Immunogenomics and Inflammation Research Unit, EA 4130, Edouard Herriot Hospital, Hospices Civils de Lyon and University Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
40
|
Beringer A, Miossec P. Systemic effects of IL-17 in inflammatory arthritis. Nat Rev Rheumatol 2019; 15:491-501. [PMID: 31227819 DOI: 10.1038/s41584-019-0243-5] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2019] [Indexed: 12/20/2022]
Abstract
Inflammatory arthritis occurs in many diseases and is characterized by joint inflammation and damage. However, the inflammatory state in arthritis is commonly associated with systemic manifestations, which are generally linked to a poor prognosis. The pro-inflammatory cytokine IL-17 functions within a complex network of cytokines and contributes to the pathogenesis of various inflammatory diseases. Three IL-17 inhibitors have already been approved for the treatment of psoriasis, psoriatic arthritis, and ankylosing spondylitis. After a brief description of IL-17 and its local effects on joints, this Review focuses on the systemic effects of IL-17 in inflammatory arthritis. Increased circulating concentrations of bioactive IL-17 mediate changes in blood vessels, liver and cardiac and skeletal muscles. The effects of IL-17 on vascular and cardiac cells might contribute to the increased risk of cardiovascular events that occurs in all patients with inflammatory disorders. In the liver, IL-17 contributes to the high circulating concentrations of acute-phase proteins, such as C-reactive protein, and the appearance of liver lesions. In skeletal muscle, IL-17 contributes to muscle contractibility defects and weakness. Thus, targeting IL-17 might have beneficial effects at both local and systemic levels, and could also be proposed for the treatment of a wider range of inflammatory diseases.
Collapse
Affiliation(s)
- Audrey Beringer
- Immunogenomics and Inflammation Research Unit, EA 4130, University of Lyon, Hospices Civils de Lyon, Lyon, France
| | - Pierre Miossec
- Immunogenomics and Inflammation Research Unit, EA 4130, University of Lyon, Hospices Civils de Lyon, Lyon, France.
| |
Collapse
|
41
|
Two phase kinetics of the inflammatory response from hepatocyte-peripheral blood mononuclear cell interactions. Sci Rep 2019; 9:8378. [PMID: 31182764 PMCID: PMC6557861 DOI: 10.1038/s41598-019-44840-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 04/25/2019] [Indexed: 12/19/2022] Open
Abstract
Active liver diseases are characterized by an infiltration of inflammatory immune cells, which interact locally with hepatocytes. Co-cultures between non- and -activated human peripheral blood mononuclear cells (PBMCs) and human hepatoma HepaRG cells were used to determine the role of these cell interactions in the inflammatory response. At the early stage, PBMC-HepaRG cell interactions increased mRNA expression and/or secretion of IL-6, IL-8, CCL-20 and MCP-1, in part through direct cell contact and the induction was higher in PHA-activated conditions. The pro-inflammatory cytokines IL-17 and/or TNFα contributed to the increase of IL-6 and IL-8 secretion. HepaRG cells modulated T cell polarization by increasing Th1 cell transcription factor expression and by reducing CD3+ CD4+ IL-17+ cell frequency when PBMCs were activated with PHA. At a later stage, the presence of HepaRG cells inhibited PHA-induced HLA-DR expression on PBMCs, and PBMC proliferation. In contrast, the presence of skin fibroblasts had no effect of PBMC proliferation induced by PHA. After a first pro-inflammatory phase, PBMC-HepaRG cell interactions may down-regulate the immune response. The PBMC-hepatocyte interactions can thus participate first to the initiation of hepatitis and later to the maintenance of immune tolerance in liver, possibly contributing to chronicity.
Collapse
|
42
|
Beringer A, Miossec P. IL-17 and TNF-α co-operation contributes to the proinflammatory response of hepatic stellate cells. Clin Exp Immunol 2019; 198:111-120. [PMID: 31102558 DOI: 10.1111/cei.13316] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2019] [Indexed: 12/20/2022] Open
Abstract
Hepatic stellate cells (HSCs) have a central role in liver inflammation and fibrosis by producing inflammatory and fibrotic mediators. Their activation is regulated through direct cell-cell interactions, but also through systemic and local effects of soluble factors such as cytokines. The effects of the proinflammatory cytokines interleukin (IL)-17 and tumor necrosis factor (TNF)-α and cell interactions with hepatocytes on HSC activation were assessed. Human HSC and HepaRG cells were exposed to IL-17 and/or TNF-α. IL-17 and TNF-α contribution from immune cells was determined in a co-culture model with phytohemagglutinin (PHA)-activated peripheral blood mononuclear cells (PBMC), HSC and/or hepatocytes. IL-17 enhanced TNF-α effects on the induction of IL-6, IL-1β, and the chemokine IL-8, chemokine (C-C motif) ligand 20 (CCL20) and monocyte chemoattractant protein-1 (MCP-1) expression/secretion in isolated HSC cultures. HSC-hepatocyte interactions did not enhance IL-6, IL-8 and CCL20 production compared to hepatocyte alone. However, HSC-hepatocyte interactions increased C-reactive protein expression. IL-17 and/or TNF-α had no direct profibrotic effects on collagen 1 α1, tissue inhibitor of matrix metalloproteinase (TIMP) and matrix metalloproteinase (MMP) 2 gene expression, whereas mRNA levels of MMP3, an enzyme involved in matrix destruction, were up-regulated in HSCs. The use of specific inhibitors of IL-17 and TNF-α indicated their contribution to the strong increase of IL-6 and IL-8 production induced by PBMC, HSC and/or hepatocyte interactions. As chronic liver inflammation leads to liver fibrosis, IL-17 and/or TNF-α neutralization can be of interest to control liver inflammation and therefore its effects on fibrosis.
Collapse
Affiliation(s)
- A Beringer
- Immunogenomics and Inflammation research Unit EA 4130, University of Lyon, Lyon, France.,Department of Clinical Immunology and Rheumatology, Hospices Civils de Lyon, Lyon, France
| | - P Miossec
- Immunogenomics and Inflammation research Unit EA 4130, University of Lyon, Lyon, France.,Department of Clinical Immunology and Rheumatology, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
43
|
Wu Y, Liu FF, Xu Y, Wang JJ, Samadli S, Wu YF, Liu HH, Chen WX, Luo HH, Zhang DD, Wei W, Hu P. Interleukin-6 is prone to be a candidate biomarker for predicting incomplete and IVIG nonresponsive Kawasaki disease rather than coronary artery aneurysm. Clin Exp Med 2019; 19:173-181. [PMID: 30617865 DOI: 10.1007/s10238-018-00544-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 12/31/2018] [Indexed: 12/18/2022]
Abstract
Kawasaki disease (KD) is an acute, systemic vasculitis and occurs mainly in childhood. Interleukin-6 (IL-6) is a pleiotropic cytokine synthesized predominantly by neutrophils and monocytes/macrophages and plays an important role in systemic inflammatory disease. However, a little information is currently available on the relationship of serum IL-6 with conventional inflammatory mediators, clinical classification, IVIG response and coronary artery aneurysm (CAA). 165 Chinese children with KD were enrolled and divided into six subgroups, including complete KD, incomplete KD, IVIG-responsive KD, IVIG-nonresponsive KD, coronary artery noninvolvement KD and coronary artery involvement KD. Blood samples were collected from all subjects within 24-h pre- and 48-h post-IVIG therapy, respectively. Serum IL-6 and conventional inflammatory mediators were detected. (1) Serum IL-6 markedly increased in the acute phase of KD, whereas declined to normal after IVIG therapy; it was positively correlated with C-reactive protein and erythrocyte sedimentation rate. (2) Serum IL-6 was significantly elevated in patients with incomplete KD when compared with their complete counterparts. The area under receiver operating characteristic curve (AUC) value for serum IL-6 in prediction of incomplete KD was 0.596, and the estimated sensitivity and specificity were 77.80% and 54.40% with a cutoff of IL-6 > 13.25 pg/ml, respectively. (3) Serum IL-6 was significantly elevated in patients with IVIG-nonresponsive KD when compared with their IVIG-responsive counterparts; the AUC value for serum IL-6 in prediction of IVIG-nonresponsive KD was 0.580, and the estimated sensitivity and specificity were 60.00% and 66.30% with a cutoff of IL-6 > 26.40 pg/ml, respectively. (4) No significant differences in IL-6 were found between KD patients with and without CAA. IL-6 is prone to be a candidate biomarker for predicting incomplete and IVIG nonresponsive KD rather than CAA.
Collapse
Affiliation(s)
- Yue Wu
- Department of Pediatrics, the First Affiliated Hospital of Anhui Medical University, No. 218 Ji-Xi Road, Hefei, 230022, People's Republic of China
| | - Fei Fei Liu
- Department of Pediatrics, the First Affiliated Hospital of Anhui Medical University, No. 218 Ji-Xi Road, Hefei, 230022, People's Republic of China
| | - Yao Xu
- Department of Pediatrics, the First Affiliated Hospital of Anhui Medical University, No. 218 Ji-Xi Road, Hefei, 230022, People's Republic of China
| | - Jing Jing Wang
- Department of Pediatrics, the First Affiliated Hospital of Anhui Medical University, No. 218 Ji-Xi Road, Hefei, 230022, People's Republic of China
| | - Sama Samadli
- Department of Pediatrics, the First Affiliated Hospital of Anhui Medical University, No. 218 Ji-Xi Road, Hefei, 230022, People's Republic of China
| | - Yang Fang Wu
- Department of Pediatrics, the First Affiliated Hospital of Anhui Medical University, No. 218 Ji-Xi Road, Hefei, 230022, People's Republic of China
| | - Hui Hui Liu
- Department of Pediatrics, the First Affiliated Hospital of Anhui Medical University, No. 218 Ji-Xi Road, Hefei, 230022, People's Republic of China
| | - Wei Xia Chen
- Department of Pediatrics, the First Affiliated Hospital of Anhui Medical University, No. 218 Ji-Xi Road, Hefei, 230022, People's Republic of China
| | - Huang Huang Luo
- Department of Pediatrics, the First Affiliated Hospital of Anhui Medical University, No. 218 Ji-Xi Road, Hefei, 230022, People's Republic of China
| | - Dong Dong Zhang
- Department of Pediatrics, the First Affiliated Hospital of Anhui Medical University, No. 218 Ji-Xi Road, Hefei, 230022, People's Republic of China
| | - Wei Wei
- Department of Pediatrics, the First Affiliated Hospital of Anhui Medical University, No. 218 Ji-Xi Road, Hefei, 230022, People's Republic of China
| | - Peng Hu
- Department of Pediatrics, the First Affiliated Hospital of Anhui Medical University, No. 218 Ji-Xi Road, Hefei, 230022, People's Republic of China.
| |
Collapse
|
44
|
Zhao Y, Dang Z, Wei R, Gui W, Zhang Y, Chong S. The effects of CCR7 and related signaling pathways on Leishmania major -infected human dendritic cells. J Cell Physiol 2018; 234:13145-13156. [PMID: 30584667 DOI: 10.1002/jcp.27985] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 11/21/2018] [Indexed: 01/09/2023]
Abstract
OBJECTIVES In our research, we aimed to investigate the roles of CC-chemokine receptor 7 (CCR7) and relevant signaling pathways in Leishmania major-infected human dendritic cells (DCs). METHODS Differentially expressed genes (DEGs) in L. major-infected human DCs were selected out and visualized using R program. Kyoto Encyclopedia of Genes and Genomes pathway analysis was conducted for investigation of significantly enriched signaling pathways and Gene Ontology enrichment analysis was carried out for the unveiling of enriched Molecular Functions and Biological Processes in L. major-infected human DCs. Besides, Hub gene was screened out using weighted gene coexpression network analysis and Cytoscape. In addition, enzyme-linked immunosorbent assay and real-time quantitative polymerase chain reaction were used for detection of relative expression of CCR7, interleukin-12 (IL-12), and interferon-γ (IFN-γ) in L. major-infected human DCs and western blot analysis was used for detection of relative expression of CCR7 and other proteins in JAK-STAT signaling pathway in L. major-infected human DCs. RESULTS CCR7 was upregulated and both chemokine and JAK-STAT signaling pathway were activated in L. major-infected human DCs. During the L. major infection, total number of L. major-infected human DCs were increased, as well as the relative expression levels of CCR7, IL-12, and IFN-γ and proteins in the JAK-STAT signaling pathway. Overexpression of CCR7 not only increased expression levels of IL-12 and IFN-γ but also activated the JAK-STAT signaling pathway to affect the leishmaniasis progression. CONCLUSION L. major infection-induced activation of CCR7, as well as JAK2 and STAT1, might well upregulate the expression of BAX yet suppress the expression of both Bcl2 and c-Jun to affect leishmaniasis progression.
Collapse
Affiliation(s)
- Yumin Zhao
- Department of Parasitology, Guilin Medical University, Guilin, China
| | - Zhisheng Dang
- Key Laboratory on Biology of Parasite and Vector, Parasitic Disease Control Center of China Center for Disease Control and Prevention, Ministry of Health, Shanghai, China
| | - Riming Wei
- Collge of Biotechnology, Guilin Medical University, Guilin, China
| | - Weifeng Gui
- Department of Parasitology, Guilin Medical University, Guilin, China
| | - Yishu Zhang
- Department of Parasitology, Guilin Medical University, Guilin, China
| | - Shigui Chong
- School of Nursing, Guilin Medical University, Guilin, China
| |
Collapse
|
45
|
Maione F, Iqbal AJ, Raucci F, Letek M, Bauer M, D'Acquisto F. Repetitive Exposure of IL-17 Into the Murine Air Pouch Favors the Recruitment of Inflammatory Monocytes and the Release of IL-16 and TREM-1 in the Inflammatory Fluids. Front Immunol 2018; 9:2752. [PMID: 30555461 PMCID: PMC6284009 DOI: 10.3389/fimmu.2018.02752] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 11/08/2018] [Indexed: 12/21/2022] Open
Abstract
The infiltration of Th17 cells in tissues and organs during the development of many autoimmune diseases is considered a key step toward the establishment of chronic inflammation. Indeed, the localized and prolonged release of IL-17 in specific tissues has been associated with an increased severity of the inflammatory response that remains sustained over time. The cellular and molecular mechanisms behind these effects are far from being clear. In this study we investigated the effects of two repetitive administration of recombinant IL-17 into the murine air pouch to simulate a scenario where IL-17 is released over time in a pre-inflamed tissue. Consistent with our previous observations, mice receiving a single dose of IL-17 showed a transitory influx of neutrophils into the air pouch that peaked at 24 h and declined at 48 h. Conversely, mice receiving a double dose of the cytokine—one at time 0 and the second after 24 h—showed a more dramatic inflammatory response with almost 2-fold increase in the number of infiltrated leukocytes and significant higher levels of TNF-α and IL-6 in the inflammatory fluids. Further analysis of the exacerbated inflammatory response of double-injected IL-17 mice showed a unique cellular and biochemical profile with inflammatory monocytes as the second main population emigrating to the pouch and IL-16 and TREM-1 as the most upregulated cytokines found in the inflammatory fluids. Most interestingly, mice receiving a double injection of IL-1β did not show any change in the cellular or biochemical inflammatory response compared to those receiving a single injection or just vehicle. Collectively these results shed some light on the function of IL-17 as pro-inflammatory cytokine and provide possible novel ways to target therapeutically the pathogenic effects of IL-17 in autoimmune conditions.
Collapse
Affiliation(s)
- Francesco Maione
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy.,William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Asif Jilani Iqbal
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Federica Raucci
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Michal Letek
- Health Science Research Centre, Department of Life Science, University of Roehampton, London, United Kingdom
| | - Martina Bauer
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Fulvio D'Acquisto
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Health Science Research Centre, Department of Life Science, University of Roehampton, London, United Kingdom
| |
Collapse
|