1
|
Scott EN, Ye C, Yano H, Lipatova Z, Brunazzi E, Vignali KM, Workman CJ, Vignali DA. Ebi3 Binding to IFN-γ and IL-10 Limits Their Function. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1115-1124. [PMID: 39240167 PMCID: PMC11458358 DOI: 10.4049/jimmunol.2400236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/19/2024] [Indexed: 09/07/2024]
Abstract
EBV-induced gene 3 (Ebi3) is a β subunit within the IL-12 cytokine family that canonically binds to α subunits p19, p28, or p35 to form the heterodimeric cytokines IL-39, IL-27, and IL-35, respectively. In the last decade, the binding partners for Ebi3 have continued to expand to include IL-6 and the other IL-12 family β subunit p40, revealing the possibility that Ebi3 may be able to bind to other cytokines and have distinct functions. We first explored this possibility utilizing an in vivo mouse model of regulatory T cell-restricted deletions of the subunits composing the cytokine IL-35, p35, and Ebi3, and we observed a differential impact on CD8+ T cell inhibitory receptor expression despite comparable reduction in tumor growth. We then screened the ability of Ebi3 to bind to different cytokines with varying structural resemblance to the IL-12 family α subunits. These in vitro screens revealed extracellular binding of Ebi3 to both IFN-γ and IL-10. Ebi3 bound to IFN-γ and IL-10 abrogated signal transduction and downstream functions of both cytokines. Lastly, we validated that extracellular complex formation after mixing native proteins resulted in loss of function. These data suggest that secreted partnerless Ebi3 may bind to cytokines within the extracellular microenvironment and act as a cytokine sink, further expanding the potential immunological impact of Ebi3.
Collapse
Affiliation(s)
- Ellen N. Scott
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Program in Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA
| | - Cheng Ye
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA
- Present address: Neurophth Therapeutics, Minhang District, Shanghai, China
| | - Hiroshi Yano
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Program in Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA
- Present address and affiliation: Jill Roberts Institute for Research in Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY
- Friedman Center for Nutrition and Inflammation, Weill Cornell Medicine, Cornell University, New York, NY
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY
| | - Zhanna Lipatova
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA
| | - Erin Brunazzi
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA
| | - Kate M. Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA
| | - Creg J. Workman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA
| | - Dario A.A. Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA
| |
Collapse
|
2
|
Lopalco G, Cito A, Venerito V, Iannone F, Proft F. The management of axial spondyloarthritis with cutting-edge therapies: advancements and innovations. Expert Opin Biol Ther 2024; 24:835-853. [PMID: 39109494 DOI: 10.1080/14712598.2024.2389987] [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: 05/28/2024] [Accepted: 08/05/2024] [Indexed: 08/28/2024]
Abstract
INTRODUCTION Axial involvement in spondyloarthritis has significantly evolved from the original 1984 New York criteria for ankylosing spondylitis, leading to an improved understanding of axial spondyloarthritis (axSpA) as a disease continuum encompassing non- radiographic axSpA (nr-axSpA) and radiographic axSpA (r-axSpA). A clear definition for early axSpA has been established, underscoring the need for early intervention with biological and targeted synthetic drugs to mitigate pain, reduce functional impairment, and prevent radiographic progression. AREAS COVERED This review explores therapeutic strategies in axSpA management, focusing on biological and targeted synthetic therapies and recent advancements. Biologics targeting TNFα or IL-17 and targeted synthetic disease-modifying antirheumatic drugs (DMARDs) are primary treatment options. These therapies significantly impact clinical outcomes, radiographic progression, and patient-reported functional improvement. EXPERT OPINION AxSpA treatment has evolved significantly, offering various therapeutic options. Biological DMARDs, particularly TNFα inhibitors, have transformed treatment, significantly enhancing patient outcomes. However, challenges persist for patients unresponsive or intolerant to existing therapies. Emerging therapeutic targets promise to address these challenges. Comprehensive management strategies and personalized approaches, considering extra-articular manifestations and individual patient factors, are crucial for achieving optimal outcomes in axSpA management.
Collapse
Affiliation(s)
- Giuseppe Lopalco
- Department of Precision Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro, Bari, Italy
| | - Andrea Cito
- Department of Precision Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro, Bari, Italy
| | - Vincenzo Venerito
- Department of Precision Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro, Bari, Italy
| | - Florenzo Iannone
- Department of Precision Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro, Bari, Italy
| | - Fabian Proft
- Department of Gastroenterology, Infectiology and Rheumatology (including Nutrition Medicine), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
3
|
Ferrucci V, Miceli M, Pagliuca C, Bianco O, Castaldo L, Izzo L, Cozzolino M, Zannella C, Oglio F, Polcaro A, Randazzo A, Colicchio R, Galdiero M, Berni Canani R, Salvatore P, Zollo M. Modulation of innate immunity related genes resulting in prophylactic antimicrobial and antiviral properties. J Transl Med 2024; 22:574. [PMID: 38886736 PMCID: PMC11184722 DOI: 10.1186/s12967-024-05378-2] [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: 03/14/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND The innate immunity acts during the early phases of infection and its failure in response to a multilayer network of co-infections is cause of immune system dysregulation. Epidemiological SARS-CoV-2 infections data, show that Influenza Virus (FLU-A-B-C) and Respiratory Syncytial Virus (RSV) are co-habiting those respiratory traits. These viruses, especially in children (mostly affected by 'multi-system inflammatory syndrome in children' [MIS-C] and the winter pandemic FLU), in the aged population, and in 'fragile' patients are causing alteration in immune response. Then, bacterial and fungal pathogens are also co-habiting the upper respiratory traits (e.g., Staphylococcus aureus and Candida albicans), thus contributing to morbidity in those COVID-19 affected patients. METHODS Liquid chromatography coupled with high-resolution mass spectrometry using the quadrupole orbital ion trap analyser (i.e., UHPLC-Q-Orbitrap HRMS) was adopted to measure the polyphenols content of a new nutraceutical formula (Solution-3). Viral infections with SARS-CoV-2 (EG.5), FLU-A and RSV-A viruses (as performed in BLS3 authorised laboratory) and real time RT-PCR (qPCR) assay were used to test the antiviral action of the nutraceutical formula. Dilution susceptibility tests have been used to estimate the minimum inhibitory and bactericidal concentration (MIC and MBC, respectively) of Solution-3 on a variety of microorganisms belonging to Gram positive/ negative bacteria and fungi. Transcriptomic data analyses and functional genomics (i.e., RNAseq and data mining), coupled to qPCR and ELISA assays have been used to investigate the mechanisms of action of the nutraceutical formula on those processes involved in innate immune response. RESULTS Here, we have tested the combination of natural products containing higher amounts of polyphenols (i.e., propolis, Verbascum thapsus L., and Thymus vulgaris L.), together with the inorganic long chain polyphosphates 'polyPs' with antiviral, antibacterial, and antifungal behaviours, against SARS-CoV-2, FLU-A, RSV-A, Gram positive/ negative bacteria and fungi (i.e., Candida albicans). These components synergistically exert an immunomodulatory action by enhancing those processes involved in innate immune response (e.g., cytokines: IFNγ, TNFα, IL-10, IL-6/12; chemokines: CXCL1; antimicrobial peptides: HBD-2, LL-37; complement system: C3). CONCLUSION The prophylactic antimicrobial success of this nutraceutical formula against SARS-CoV-2, FLU-A and RSV-A viruses, together with the common bacteria and fungi co-infections as present in human oral cavity, is expected to be valuable.
Collapse
Affiliation(s)
- Veronica Ferrucci
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples 'Federico II', Via Sergio Pansini 5, 80131, Naples, Italy.
- CEINGE Biotecnologie Avanzate 'Franco Salvatore', Via Gaetano Salvatore 486, 80145, Naples, Italy.
- Elysium Cell Bio Ita, Via Gaetano Salvatore 486, 80145, Naples, Italy.
| | - Marco Miceli
- CEINGE Biotecnologie Avanzate 'Franco Salvatore', Via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Chiara Pagliuca
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples 'Federico II', Via Sergio Pansini 5, 80131, Naples, Italy
| | - Orazio Bianco
- CEINGE Biotecnologie Avanzate 'Franco Salvatore', Via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Luigi Castaldo
- Department of Pharmacy, University of Naples 'Federico II', Via Domenico Montesano 49, 80131, Naples, Italy
| | - Luana Izzo
- Department of Pharmacy, University of Naples 'Federico II', Via Domenico Montesano 49, 80131, Naples, Italy
| | - Marica Cozzolino
- CEINGE Biotecnologie Avanzate 'Franco Salvatore', Via Gaetano Salvatore 486, 80145, Naples, Italy
- Dipartimento Di Scienze Mediche Traslazionali, University of Naples Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Carla Zannella
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Franca Oglio
- CEINGE Biotecnologie Avanzate 'Franco Salvatore', Via Gaetano Salvatore 486, 80145, Naples, Italy
- Dipartimento Di Scienze Mediche Traslazionali, University of Naples Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Antonio Polcaro
- Polcaro Fitopreparazioni S.R.L, Via Sant Agnello, 9 D; 80030, Roccarainola, Naples, Italy
| | - Antonio Randazzo
- Department of Pharmacy, University of Naples 'Federico II', Via Domenico Montesano 49, 80131, Naples, Italy
| | - Roberta Colicchio
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples 'Federico II', Via Sergio Pansini 5, 80131, Naples, Italy
| | - Massimiliano Galdiero
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
- UOC of Virology and Microbiology, University Hospital of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Roberto Berni Canani
- CEINGE Biotecnologie Avanzate 'Franco Salvatore', Via Gaetano Salvatore 486, 80145, Naples, Italy
- Dipartimento Di Scienze Mediche Traslazionali, University of Naples Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Paola Salvatore
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples 'Federico II', Via Sergio Pansini 5, 80131, Naples, Italy
- CEINGE Biotecnologie Avanzate 'Franco Salvatore', Via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Massimo Zollo
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples 'Federico II', Via Sergio Pansini 5, 80131, Naples, Italy.
- CEINGE Biotecnologie Avanzate 'Franco Salvatore', Via Gaetano Salvatore 486, 80145, Naples, Italy.
- Elysium Cell Bio Ita, Via Gaetano Salvatore 486, 80145, Naples, Italy.
- DAI Medicina di Laboratorio e Trasfusionale, University of Naples Federico II, Via Sergio Pansini 5, 80131, Naples, Italy.
| |
Collapse
|
4
|
Chen Z, Balachandran YL, Chong WP, Chan KWY. Roles of Cytokines in Alzheimer's Disease. Int J Mol Sci 2024; 25:5803. [PMID: 38891990 PMCID: PMC11171747 DOI: 10.3390/ijms25115803] [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: 04/16/2024] [Revised: 05/18/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
The neuroimmune system is a collection of immune cells, cytokines, and the glymphatic system that plays a pivotal role in the pathogenesis and progression of Alzheimer's disease (AD). Of particular focus are cytokines, a group of immune signaling molecules that facilitate communication among immune cells and contribute to inflammation in AD. Extensive research has shown that the dysregulated secretion of certain cytokines (IL-1β, IL-17, IL-12, IL-23, IL-6, and TNF-α) promotes neuroinflammation and exacerbates neuronal damage in AD. However, anti-inflammatory cytokines (IL-2, IL-3, IL-33, and IL-35) are also secreted during AD onset and progression, thereby preventing neuroinflammation. This review summarizes the involvement of pro- and anti-inflammatory cytokines in AD pathology and discusses their therapeutic potential.
Collapse
Affiliation(s)
- Zilin Chen
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; (Z.C.); (Y.L.B.)
| | - Yekkuni L. Balachandran
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; (Z.C.); (Y.L.B.)
| | - Wai Po Chong
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen 518057, China
| | - Kannie W. Y. Chan
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; (Z.C.); (Y.L.B.)
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong, China
- Shenzhen Research Institute, City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, China
- Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong, China
| |
Collapse
|
5
|
Fonseca-Camarillo G, Furuzawa-Carballeda J, Barreto-Zúñiga R, Martínez-Benítez B, Yamamoto-Furusho JK. Increased synthesis and intestinal expression of IL-39 in patients with inflammatory bowel disease. Immunol Res 2024; 72:284-292. [PMID: 37968544 DOI: 10.1007/s12026-023-09432-x] [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: 06/26/2023] [Accepted: 10/26/2023] [Indexed: 11/17/2023]
Abstract
IL-39 (Interleukin-39) is a heterodimeric cytokine composed of IL-23p19 and EBI3 (Epstein-Barr virus-induced gene 3) subunits. Despite the evidence that correlates the role of IL-39 in regulating inflammation, its expression in the intestinal microenvironment of IBD (inflammatory bowel disease) patients is still unknown. Thus, this work was focused on characterizing relative mRNA (messenger RNA) IL-39 expression and intestinal synthesis in IBD patients. This study includes 37 patients diagnosed with ulcerative colitis (UC), 15 with Chron's disease (CD), and 22 controls. Gene expression of IL-39 subunits (IL-23p19/EBI3) was measured by RT-PCR (real time polymerase chain reaction). Intestinal synthesis was evaluated by immunohistochemistry and serum levels by ELISA. Statistical analysis was done using Prism GraphPad V6. Relative mRNA IL-39 expression was increased in patients with active UC and active CD compared to the remission UC, remission CD, and control group. High levels of relative mRNA expression of IL-39 (IL-23p19 subunit) were associated with histological activity. IHQ analysis showed increased IL-39 production in mucosa, submucosa, muscular, and serosa layer of patients with active disease. IL-39 serum production was increased in patients with UC. IL-39 gene's upregulation was found in patients with active IBD and was associated with severe histological activity in UC. This is the first report regarding the role of IL-39 in patients with IBD. The findings suggest that IL-39 might play a role as an inflammatory mediator in active IBD and could be considered a new alternative in treating this condition.
Collapse
Affiliation(s)
- Gabriela Fonseca-Camarillo
- Inflammatory Bowel Disease Clinic, Department of Gastroenterology, Instituto Nacional de Ciencias Médicas y Nutrición, Salvador Zubirán, #15, Col. Belisario Domínguez Sección XVI, 14080, Mexico City, CP, Mexico
- Department of Immunology, Instituto Nacional de Cardiología, Ignacio Chávez, Mexico, Mexico City
| | - Janette Furuzawa-Carballeda
- Department of Experimental Surgery, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Rafael Barreto-Zúñiga
- Department of Endoscopy, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Braulio Martínez-Benítez
- Department of Pathology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Jesús K Yamamoto-Furusho
- Inflammatory Bowel Disease Clinic, Department of Gastroenterology, Instituto Nacional de Ciencias Médicas y Nutrición, Salvador Zubirán, #15, Col. Belisario Domínguez Sección XVI, 14080, Mexico City, CP, Mexico.
| |
Collapse
|
6
|
Zhu Z, Peng Q, Duan X, Li J. Interleukin-12: Structure, Function, and Its Impact in Colorectal Cancer. J Interferon Cytokine Res 2024; 44:158-169. [PMID: 38498032 DOI: 10.1089/jir.2023.0190] [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: 03/19/2024] Open
Abstract
Interleukin 12 (IL-12) is a heterodimer consisting of 2 subunits, p35 and p40, with unique associations and interacting functions with its family members. IL-12 is one of the most important cytokines regulating the immune system response and is integral to adaptive immunity. IL-12 has shown marked therapeutic potential in a variety of tumor types. This review therefore summarizes the characteristics of IL-12 and its application in tumor treatment, focusing on its antitumor effects in colorectal cancer (CRC) and potential radiosensitization mechanisms. We aim to provide a current reference for IL-12 and other potential CRC treatment strategies.
Collapse
Affiliation(s)
- Ziwei Zhu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Qian Peng
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Xingmei Duan
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine University of Electronic Science and Technology of China, Chengdu, People's Republic of. China
| | - Jie Li
- School of Medicine, Southwest Medical University of China, Luzhou, People's Republic of China
- Department of Radiotherapy, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| |
Collapse
|
7
|
Imamichi T, Chen Q, Sowrirajan B, Yang J, Laverdure S, Marquez M, Mele AR, Watkins C, Adelsberger JW, Higgins J, Sui H. Interleukin-27-induced HIV-resistant dendritic cells suppress reveres transcription following virus entry in an SPTBN1, autophagy, and YB-1 independent manner. PLoS One 2023; 18:e0287829. [PMID: 37910521 PMCID: PMC10619827 DOI: 10.1371/journal.pone.0287829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 10/03/2023] [Indexed: 11/03/2023] Open
Abstract
Interleukin (IL)-27, a member of the IL-12 family of cytokines, induces human immunodeficiency virus (HIV)-resistant monocyte-derived macrophages and T cells. This resistance is mediated via the downregulation of spectrin beta, non-erythrocytic 1 (SPTBN1), induction of autophagy, or suppression of the acetylation of Y-box binding protein-1 (YB-1); however, the role of IL-27 administration during the induction of immature monocyte-derived dendritic cells (iDC) is poorly investigated. In the current study, we investigated the function of IL-27-induced iDC (27DC) on HIV infection. 27DC inhibited HIV infection by 95 ± 3% without significant changes in the expression of CD4, CCR5, and SPTBN1 expression, autophagy induction and acetylation of YB-1 compared to iDC. An HIV proviral DNA copy number assay displayed that 27DC suppressed reverse transcriptase (RT) reaction without influencing the virus entry. A DNA microarray analysis was performed to identify the differentially expressed genes between 27DC and iDC. Compared to iDC, 51 genes were differentially expressed in 27DC, with more than 3-fold changes in four independent donors. Cross-reference analysis with the reported 2,214 HIV regulatory host genes identified nine genes as potential interests: Ankyrin repeat domain 22, Guanylate binding protein (GBP)-1, -2, -4, -5, Stabilin 1, Serpin family G member 1 (SERPING1), Interferon alpha inducible protein 6, and Interferon-induced protein with tetratricopeptide repeats 3. A knock-down study using si-RNA failed to determine a key factor associated with the anti-HIV activity due to the induction of robust amounts of off-target effects. Overexpression of each protein in cells had no impact on HIV infection. Thus, we could not define the mechanism of the anti-HIV effect in 27DC. However, our findings indicated that IL-27 differentiates monocytes into HIV-resistant DC, and the inhibitory mechanism differs from IL-27-induced HIV-resistant macrophages and T cells.
Collapse
Affiliation(s)
- Tomozumi Imamichi
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Qian Chen
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Bharatwaj Sowrirajan
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Jun Yang
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Sylvain Laverdure
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Mayra Marquez
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Anthony R. Mele
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Catherine Watkins
- AIDS monitoring Laboratory, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Joseph W. Adelsberger
- AIDS monitoring Laboratory, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Jeanette Higgins
- AIDS monitoring Laboratory, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Hongyan Sui
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| |
Collapse
|
8
|
Yoon T, Ha JW, Ko E, Song JJ, Park YB, Ahn SS, Lee SW. Vasculitis Activity-Predicting Ability of IL-12 Family Cytokines in Patients with Microscopic Polyangiitis and Granulomatosis with Polyangiitis. Yonsei Med J 2023; 64:604-611. [PMID: 37727919 PMCID: PMC10522880 DOI: 10.3349/ymj.2023.0226] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/07/2023] [Accepted: 07/09/2023] [Indexed: 09/21/2023] Open
Abstract
PURPOSE The present study investigated and compared the antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) activity-predicting ability of the serum concentrations of the four interleukin (IL)-12 family cytokines including IL-23, IL-27, IL-35, and IL-39 in patients with microscopic polyangiitis (MPA) and granulomatosis with polyangiitis (GPA). MATERIALS AND METHODS The present study included 70 patients with MPA and GPA. Clinical and laboratory data, particularly Birmingham Vasculitis Activity Score (BVAS), at the time of blood collection were obtained. The serum concentrations of IL-23, IL-27, IL-35, and IL-37 were measured using sera stored at -80℃. Patients were divided into two groups: the upper half of BVAS (BVAS ≥12) and the lower half of BVAS (BVAS <12). RESULTS The serum concentrations of IL-23 and IL-27 reflected AAV activity. Patients with the upper half of BVAS exhibited significantly higher serum concentrations of IL-23 and IL-27 than those without. Patients with the serum concentrations of IL-23 ≥132.1 pg/mL or IL-27 ≥684.7 pg/mL exhibited higher frequency and risk for the upper half of BVAS than those without [relative risks (RR) 5.143 and RR 4.091, respectively]. The serum concentrations of IL-27 were associated with age ≥65 years and proteinase 3-ANCA (or C-ANCA) negativity, whereas, those of IL-23 were associated with MPA. However, the serum concentrations of IL-35 and IL-39 were not useful in predicting AAV activity in this study. CONCLUSION The present study is the first to demonstrate that among the various members of IL-12 family cytokines, the serum concentrations of IL-23 and IL-27 possess AAV activity-predicting ability.
Collapse
Affiliation(s)
- Taejun Yoon
- Department of Medical Science, BK2 Plus Project, Yonsei University College of Medicine, Seoul, Korea
| | - Jang Woo Ha
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Eunhee Ko
- Department of Medical Science, BK2 Plus Project, Yonsei University College of Medicine, Seoul, Korea
| | - Jason Jungsik Song
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - Yong-Beom Park
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - Sung Soo Ahn
- Division of Rheumatology, Department of Internal Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Korea.
| | - Sang-Won Lee
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
9
|
Mora-Boza A, Ghebrezadik SG, Leisen JE, García AJ. Rapid and Facile Light-Based Approach to Fabricate Protease-Degradable Poly(ethylene glycol)-norbornene Microgels for Cell Encapsulation. Adv Healthc Mater 2023; 12:e2300942. [PMID: 37235850 PMCID: PMC10592588 DOI: 10.1002/adhm.202300942] [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: 03/23/2023] [Revised: 05/15/2023] [Indexed: 05/28/2023]
Abstract
Thiol-norbornene photoclickable poly (ethylene glycol) (PEG)-based (PEG-NB) hydrogels are attractive biomaterials for cell encapsulation, drug delivery, and regenerative medicine applications. Although many crosslinking strategies and chemistries have been developed for PEG-NB bulk hydrogels, fabrication approaches of PEG-NB microgels have not been extensively explored. Here, a fabrication strategy for 4-arm amide-linked PEG-NB (PEG-4aNB) microgels using flow-focusing microfluidics for human mesenchymal stem/stromal cell (hMSCs) encapsulation is presented. PEG-4aNB photochemistry allows high-throughput, ultrafast generation, and cost-effective synthesis of monodispersed microgels (diameter 340 ± 18, 380 ± 24, and 420 ± 15 µm, for 6, 8, and 10 wt% of PEG-4aNB, respectively) using an in situ crosslinking methodology in a microfluidic device. PEG-4aNB microgels show in vitro degradability due to the incorporation of a protease-degradable peptide during photocrosslinking and encapsulated cells show excellent viability and metabolic activity for at least 13 days of culture. Furthermore, the secretory profile (i.e., MMP-13, ICAM-1, PD-L1, CXCL9, CCL3/MIP-1, IL-6, IL-12, IL-17E, TNF-α, CCL2/MCP-1) of encapsulated hMSCs shows increased expression in response to IFN-γ stimulation. Collectively, this work shows a versatile and facile approach for the fabrication of protease-degradable PEG-4aNB microgels for cell encapsulation.
Collapse
Affiliation(s)
- Ana Mora-Boza
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
- CÚRAM, University of Galway, Galway, H91 W2TY, Ireland
| | - Saron G Ghebrezadik
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
- Agnes Scott College, Decatur, GA, 30030, USA
| | - Johannes E Leisen
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
| | - Andrés J García
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
| |
Collapse
|
10
|
Imamichi T, Chen Q, Sowrirajan B, Yang J, Laverdure S, Mele AR, Watkins C, Adelsberger JW, Higgins J, Sui H. Interleukin-27-induced HIV-resistant dendritic cells suppress reveres transcription following virus entry in an SPTBN1, Autophagy, and YB-1 independent manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.12.544550. [PMID: 37546823 PMCID: PMC10402176 DOI: 10.1101/2023.06.12.544550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Interleukin (IL)-27, a member of the IL-12 family of cytokines, induces human immunodeficiency virus (HIV)-resistant monocyte-derived macrophages and T cells. This resistance is mediated via the downregulation of spectrin beta, non-erythrocytic 1 (SPTBN1), induction of autophagy, or suppression of the acetylation of Y-box binding protein-1 (YB-1); however, the role of IL-27 administration during the induction of immature monocyte-derived dendritic cells (iDC) is poorly investigated. In the current study, we investigated the function of IL-27-induced iDC (27DC) on HIV infection. 27DC inhibited HIV infection by 95 ± 3 % without significant changes in the expression of CD4, CCR5, and SPTBN1 expression, autophagy induction and acetylation of YB-1 compared to iDC. An HIV proviral DNA copy number assay displayed that 27DC suppressed reverse transcriptase (RT) reaction without influencing the virus entry. A DNA microarray analysis was performed to identify the differentially expressed genes between 27DC and iDC. Compared to iDC, 51 genes were differentially expressed in 27DC, with more than 3-fold changes in four independent donors. Cross-reference analysis with the reported 2,214 HIV regulatory host genes identified nine genes as potential interests: Ankyrin repeat domain 22, Guanylate binding protein (GBP)-1, -2, -4, -5, Stabilin 1, Serpin family G member 1 (SERPING1), Interferon alpha inducible protein 6, and Interferon-induced protein with tetratricopeptide repeats 3. A knock-down study using si-RNA failed to determine a key factor associated with the anti-HIV activity due to the induction of robust amounts of off-target effects. Overexpression of each protein in cells had no impact on HIV infection. Thus, we could not define the mechanism of the anti-HIV effect in 27DC. However, our findings indicated that IL-27 differentiates monocytes into HIV-resistant DC, and the inhibitory mechanism differs from IL-27-induced HIV-resistant macrophages and T cells.
Collapse
Affiliation(s)
- Tomozumi Imamichi
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702
| | - Qian Chen
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702
| | - Bharatwaj Sowrirajan
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702
| | - Jun Yang
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702
| | - Sylvain Laverdure
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702
| | - Anthony R. Mele
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702
| | - Catherine Watkins
- AIDS monitoring Laboratory, Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, USA
| | - Joseph W. Adelsberger
- AIDS monitoring Laboratory, Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, USA
| | - Jeanette Higgins
- AIDS monitoring Laboratory, Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, USA
| | - Hongyan Sui
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702
| |
Collapse
|
11
|
Diao HY, Zhu W, Liu J, Yin S, Wang JH, Li CL. Salvianolic Acid A Improves Rat Kidney Injury by Regulating MAPKs and TGF-β1/Smads Signaling Pathways. Molecules 2023; 28:3630. [PMID: 37110864 PMCID: PMC10144349 DOI: 10.3390/molecules28083630] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 03/30/2023] [Accepted: 04/03/2023] [Indexed: 04/29/2023] Open
Abstract
Salvianolic acid A (SAA) is one of the major components in Salvia miltiorrhiza Bge., with various pharmacological activities, and is likely to be a promising agent for the treatment of kidney diseases. The purpose of this study was to explore the protective effect and mechanisms of SAA on kidney disease. In this study, the improvement effects of SAA (10, 20, 40 mg/kg, i.g.) on kidney injury rats were investigated by detecting the levels of KIM-1, NGAL in serum and UP in the urine of AKI model rats established with gentamicin, as well as the levels of SCr and UREA in serum and IL-6, IL-12, MDA and T-SOD in the kidneys of CKD model rats established with 5/6 nephrectomy. HE and Masson staining were used to observe the histopathological changes in the kidney. Network pharmacology and Western blotting were used to explore the mechanism of SAA in improving kidney injury. The results showed that SAA improved kidney function in kidney injury rats by reducing the kidney index and pathological injury by HE and Masson staining, reducing the levels of KIM-1, NGAL and UP in AKI rats and UREA, SCr and UP in CKD rats, as well as exerting anti-inflammatory and anti-oxidative stress effects by inhibiting the release of IL-6 and IL-12, reducing MDA and increasing T-SOD. Western blotting results showed that SAA significantly reduced the phosphorylation levels of ERK1/2, p38, JNK and smad2/3, and the expression of TLR-4 and smad7. In conclusion, SAA plays a significant role in improving kidney injury in rats and the mechanism may be achieved by regulating the MAPKs and TGF-β1/smads signaling pathways.
Collapse
Affiliation(s)
- Hai-Yang Diao
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Wei Zhu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jie Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Sheng Yin
- Department of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jin-Hui Wang
- Department of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Chun-Li Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
12
|
Wang X, Zhang A, Qiu X, Yang K, Zhou H. The IL-12 family cytokines in fish: Molecular structure, expression profile and function. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 141:104643. [PMID: 36632929 DOI: 10.1016/j.dci.2023.104643] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 01/02/2023] [Accepted: 01/08/2023] [Indexed: 06/17/2023]
Abstract
Interleukin (IL)-12 family cytokines including IL-12, IL-23, IL-27, IL-35 and IL-39 are heterodimeric cytokines composed of two subunits, an α-chain (entitled p35, p19 and p28) and a β-chain (namely p40 and Epstein-Barr virus-induced gene 3, EBI3). Unlike in mammals, specific whole genome duplication events in fish may generate more paralogues of these subunits as the components of IL-12 family cytokines. Although all subunit genes of IL-12 family have been isolated and identified in various fish species, some important issues on fish IL-12 family are needed to be addressed compared to the extensive study in mammals: Whether the expansion of these subunit genes results in the generation of multiple isoforms of the family cytokines; Whether the related receptor genes have similar complex repertoire corresponding to their ligands; How about the expression kinetics of these subunit paralogues particularly under the circumstance of pathogen infection and immune stimulation; How about the functional properties of IL-12 family in fish. In the past ten years, these concerns have received increasing attentions to establish the biological significance of this family cytokines in fish immunity. In this review, we summarized the current understanding of IL-12 family with a special focus on the molecular structures, inducible expression profiles and functions of IL-12 family members in fish.
Collapse
Affiliation(s)
- Xinyan Wang
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Anying Zhang
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Xingyang Qiu
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Kun Yang
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Hong Zhou
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, People's Republic of China.
| |
Collapse
|
13
|
Harrison SR, Marzo-Ortega H. Have Therapeutics Enhanced Our Knowledge of Axial Spondyloarthritis? Curr Rheumatol Rep 2023; 25:56-67. [PMID: 36652160 PMCID: PMC9958165 DOI: 10.1007/s11926-023-01097-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PURPOSE OF REVIEW An overview of how the treatment landscape of axial spondyloarthritis (axSpA) has shaped our understanding of the disease. RECENT FINDINGS Prior to the millennium, non-steroidal anti-inflammatory drugs (NSAIDs) were the only treatment for axSpA, yet only 30% of patients responded and many developed side effects. In 2003, the first biological disease-modifying drug (bDMARD) was licensed for axSpA which substantially improved outcomes in comparison to NSAIDs. In 2022, there are now several bDMARDs for axSpA; however, they too are not universally efficacious in treating axial inflammation and may have deleterious effects on extramusculoskeletal manifestations. Nevertheless, successful or not, each bDMARD gives invaluable insight into axSpA immunobiology. This review discusses how much we have learned from the use of bDMARDs in axSpA, how this has redefined our understanding of the disease, and how we might use this knowledge to develop new and better treatments for axSpA in the future.
Collapse
Affiliation(s)
- S R Harrison
- The University of Leeds, Leeds Institute for Rheumatic and Musculoskeletal Medicine (LIRMM), NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals Trust, Leeds, UK
- The University of Leeds, Leeds Institute of Cardiovascular and Metabolic Medicine, the LIGHT building, Clarendon Way, Leeds, UK
| | - H Marzo-Ortega
- The University of Leeds, Leeds Institute for Rheumatic and Musculoskeletal Medicine (LIRMM), NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals Trust, Leeds, UK.
| |
Collapse
|
14
|
Kim D, Kim S, Kang MS, Yin Z, Min B. Cell type specific IL-27p28 (IL-30) deletion in mice uncovers an unexpected regulatory function of IL-30 in autoimmune inflammation. Sci Rep 2023; 13:1812. [PMID: 36725904 PMCID: PMC9892501 DOI: 10.1038/s41598-023-27413-w] [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: 09/02/2022] [Accepted: 01/02/2023] [Indexed: 02/03/2023] Open
Abstract
IL-27 is an IL-12 family cytokine with immune regulatory properties, capable of modulating inflammatory responses, including autoimmunity. While extensive studies investigated the major target cells of IL-27 mediating its functions, the source of IL-27 especially during tissue specific autoimmune inflammation has not formally been examined. IL-27p28 subunit, also known as IL-30, was initially discovered as an IL-27-specific subunit, and it has thus been deemed as a surrogate marker to denote IL-27 expression. However, IL-30 can be secreted independently of Ebi3, a subunit that forms bioactive IL-27 with IL-30. Moreover, IL-30 itself may act as a negative regulator antagonizing IL-27. In this study, we exploited various cell type specific IL-30-deficient mouse models and examined the source of IL-30 in a T cell mediated autoimmune neuroinflammation. We found that IL-30 expressed by infiltrating and CNS resident APC subsets, infiltrating myeloid cells and microglia, is central in limiting the inflammation. However, dendritic cell-derived IL-30 was dispensable for the disease development. Unexpectedly, in cell type specific IL-30 deficient mice that develop severe EAE, IL-30 expression in the remaining wild-type APC subsets is disproportionately increased, suggesting that increased endogenous IL-30 production may be involved in the severe pathogenesis. In support, systemic recombinant IL-30 administration exacerbates EAE severity. Our results demonstrate that dysregulated endogenous IL-30 expression may interfere with immune regulatory functions of IL-27, promoting encephalitogenic inflammation in vivo.
Collapse
Affiliation(s)
- Dongkyun Kim
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Sohee Kim
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Myung-Su Kang
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Zhinan Yin
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, China
| | - Booki Min
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
15
|
Shi Y, Chen K, Zhao X, Lu Y, Huang W, Guo J, Ji N, Jia Z, Xiao H, Dang H, Zou J, Wang J. IL-27 suppresses spring viremia of carp virus replication in zebrafish. FISH & SHELLFISH IMMUNOLOGY 2023; 133:108530. [PMID: 36632914 DOI: 10.1016/j.fsi.2023.108530] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/06/2023] [Accepted: 01/08/2023] [Indexed: 06/17/2023]
Abstract
Interleukin (IL) 27 is a member of the IL-12 family and is a heterodimeric cytokine composed of IL-27A and Epstein-Barr virus-induced 3 (EBI3). It plays an important role in regulating inflammation and cancer progression. IL-27A not only functions by dimerizing with EBI3 but also acts alone. Here, we report that IL-27A and EBI3 suppress spring viremia of carp virus (SVCV) replication in zebrafish. Expression analysis reveals that il-27a and ebi3 were significantly upregulated in the ZF4 cells by SVCV and poly(I:C), and in the zebrafish caudal fin (ZFIN) cells overexpressed with SVCV genes. Interestingly, il-27a and ebi3 were not modulated by IFNφ1, indicating that they are not IFN stimulated genes (ISGs). Furthermore, overexpression of IL-27A and EBI3 alone inhibited SVCV replication in the EPC cells, but less potent than co-expression of IL-27A and EBI3. Intriguingly, IL-27A could not induce the expression of irf3, ifn, isg15 and mx1. Taken together, our results demonstrate that IL-27A and EBI3 activate innate antiviral response in an IFN independent manner in zebrafish.
Collapse
Affiliation(s)
- Yanjie Shi
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Kangyong Chen
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Xin Zhao
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Yanan Lu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Wenji Huang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Jiahong Guo
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Ning Ji
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Zhao Jia
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Hehe Xiao
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Huifeng Dang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Jun Zou
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266200, China.
| | - Junya Wang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China.
| |
Collapse
|
16
|
Rybchenko VS, Aliev TK, Panina AA, Kirpichnikov MP, Dolgikh DA. Targeted Cytokine Delivery for Cancer Treatment: Engineering and Biological Effects. Pharmaceutics 2023; 15:pharmaceutics15020336. [PMID: 36839658 PMCID: PMC9960319 DOI: 10.3390/pharmaceutics15020336] [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: 12/09/2022] [Revised: 01/14/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Anti-tumor properties of several cytokines have already been investigated in multiple experiments and clinical trials. However, those studies evidenced substantial toxicities, even at low cytokine doses, and the lack of tumor specificity. These factors significantly limit clinical applications. Due to their high specificity and affinity, tumor-specific monoclonal antibodies or their antigen-binding fragments are capable of delivering fused cytokines to tumors and, therefore, of decreasing the number and severity of side effects, as well as of enhancing the therapeutic index. The present review surveys the actual antibody-cytokine fusion protein (immunocytokine) formats, their targets, mechanisms of action, and anti-tumor and other biological effects. Special attention is paid to the formats designed to prevent the off-target cytokine-receptor interactions, potentially inducing side effects. Here, we describe preclinical and clinical data and the efficacy of the antibody-mediated cytokine delivery approach, either as a single therapy or in combination with other agents.
Collapse
Affiliation(s)
- Vladislav S Rybchenko
- Bioengineering Department, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Teimur K Aliev
- Bioengineering Department, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Department of Chemistry, M.V. Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Anna A Panina
- Bioengineering Department, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Mikhail P Kirpichnikov
- Bioengineering Department, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Department of Biology, M.V. Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Dmitry A Dolgikh
- Bioengineering Department, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Department of Biology, M.V. Lomonosov Moscow State University, 119234 Moscow, Russia
| |
Collapse
|
17
|
Zhang P, Zhu H. Cytokines in Thyroid-Associated Ophthalmopathy. J Immunol Res 2022; 2022:2528046. [PMID: 36419958 PMCID: PMC9678454 DOI: 10.1155/2022/2528046] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 09/07/2023] Open
Abstract
Thyroid-associated ophthalmopathy (TAO), also known as thyroid eye disease (TED) or Graves' orbitopathy (GO), is a complex autoimmune condition causing visual impairment, disfigurement, and harm to patients' physical and mental health. The pathogenesis of TAO has not been fully elucidated, and the mainstream view is that coantigens shared by the thyroid and orbit trigger remodeling of extraocular muscles and orbital connective tissues through an inflammatory response. In recent years, cytokines and the immune responses they mediate have been crucial in disease progression, and currently, common evidence has shown that drugs targeting cytokines, such as tocilizumab, infliximab, and adalimumab, may be novel targets for therapy. In this review, we summarize the research development of different cytokines in TAO pathogenesis in the hope of discovering new therapeutic targets.
Collapse
Affiliation(s)
- Pengbo Zhang
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Huang Zhu
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| |
Collapse
|
18
|
Zhou L, Tang C, Li X, Feng F. IL-6/IL-10 mRNA expression ratio in tumor tissues predicts prognosis in gastric cancer patients without distant metastasis. Sci Rep 2022; 12:19427. [PMID: 36371539 PMCID: PMC9653417 DOI: 10.1038/s41598-022-24189-3] [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: 06/30/2022] [Accepted: 11/11/2022] [Indexed: 11/13/2022] Open
Abstract
There was growing evidence that inflammatory responses played significant roles in malignancies. However, the impact of pro-inflammatory-to-anti-inflammatory factor ratio in tumor tissues has not been investigated in gastric cancer (GC) yet. We collected patient data from The Cancer Genome Atlas (TCGA) database. A total of 270 stomach adenocarcinoma (STAD) patients without distant metastasis were included in the study. After screening 12 candidate pro-inflammatory-to-anti-inflammatory pairs, only the IL-6/IL-10 mRNA expression ratio in tumor tissues had a significant effect on overall survival (OS) of STAD patients (P = 0.014). X-tile analysis showed that the greatest survival differences were obtained when the cutoff value of IL-6/IL-10 mRNA expression ratio was set at 1.3 and 5.5. With the low-ratio group (IL-6/IL-10 mRNA expression ratio: < 1.3) as reference, OS time for both the medium-ratio group (IL-6/IL-10 mRNA expression ratio: 1.3-5.5) and the high-ratio group (IL-6/IL-10 mRNA expression ratio: > 5.5) was significantly shorter (P < 0.05). Multivariate Cox regression analyses indicated that IL-6/IL-10 mRNA expression ratio was an independent prognostic factor for OS and disease-specific survival (DSS). These findings provided a novel and powerful tool for a more rational management of GC patients.
Collapse
Affiliation(s)
- Li Zhou
- grid.263761.70000 0001 0198 0694Department of Oncology, Suzhou Ninth People′s Hospital, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, 215200 China
| | - Chuangang Tang
- grid.452207.60000 0004 1758 0558Department of General Surgery, Xuzhou Central Hospital, The Affiliated Xuzhou Hospital of Medical College of Southeast University, Xuzhou, 221009 China ,grid.452207.60000 0004 1758 0558Department of Breast Surgery, Xuzhou Central Hospital, The Affiliated Xuzhou Hospital of Medical College of Southeast University, Xuzhou, 221009 China
| | - Xiaoxin Li
- grid.452207.60000 0004 1758 0558Department of Pathology, Xuzhou Central Hospital, The Affiliated Xuzhou Hospital of Medical College of Southeast University, Xuzhou, 221009 China
| | - Fang Feng
- grid.263761.70000 0001 0198 0694Department of Oncology, Suzhou Ninth People′s Hospital, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, 215200 China
| |
Collapse
|
19
|
Składanowska K, Bloch Y, Strand J, White KF, Hua J, Aldridge D, Welin M, Logan DT, Soete A, Merceron R, Murphy C, Provost M, Bazan JF, Hunter CA, Hill JA, Savvides SN. Structural basis of activation and antagonism of receptor signaling mediated by interleukin-27. Cell Rep 2022; 41:111490. [PMID: 36261006 PMCID: PMC9597551 DOI: 10.1016/j.celrep.2022.111490] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/14/2022] [Accepted: 09/21/2022] [Indexed: 11/19/2022] Open
Abstract
Interleukin-27 (IL-27) uniquely assembles p28 and EBI3 subunits to a heterodimeric cytokine that signals via IL-27Rα and gp130. To provide the structural framework for receptor activation by IL-27 and its emerging therapeutic targeting, we report here crystal structures of mouse IL-27 in complex with IL-27Rα and of human IL-27 in complex with SRF388, a monoclonal antibody undergoing clinical trials with oncology indications. One face of the helical p28 subunit interacts with EBI3, while the opposite face nestles into the interdomain elbow of IL-27Rα to juxtapose IL-27Rα to EBI3. This orients IL-27Rα for paired signaling with gp130, which only uses its immunoglobulin domain to bind to IL-27. Such a signaling complex is distinct from those mediated by IL-12 and IL-23. The SRF388 binding epitope on IL-27 overlaps with the IL-27Rα interaction site explaining its potent antagonistic properties. Collectively, our findings will facilitate the mechanistic interrogation, engineering, and therapeutic targeting of IL-27.
Collapse
Affiliation(s)
- Katarzyna Składanowska
- Unit for Structural Biology, Department of Biochemistry and Microbiology Ghent University, Technologiepark 71, 9052 Ghent, Belgium; Unit for Structural Biology, VIB-UGent Center for Inflammation Research, Technologiepark 71, 9052 Ghent, Belgium
| | - Yehudi Bloch
- Unit for Structural Biology, Department of Biochemistry and Microbiology Ghent University, Technologiepark 71, 9052 Ghent, Belgium; Unit for Structural Biology, VIB-UGent Center for Inflammation Research, Technologiepark 71, 9052 Ghent, Belgium
| | - Jamie Strand
- Surface Oncology, 50 Hampshire Street, Cambridge, MA 02139, USA
| | - Kerry F White
- Surface Oncology, 50 Hampshire Street, Cambridge, MA 02139, USA
| | - Jing Hua
- Surface Oncology, 50 Hampshire Street, Cambridge, MA 02139, USA
| | - Daniel Aldridge
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Martin Welin
- SARomics Biostructures AB, Medicon Village, Scheelevägen 2, 223 63 Lund, Sweden
| | - Derek T Logan
- SARomics Biostructures AB, Medicon Village, Scheelevägen 2, 223 63 Lund, Sweden
| | - Arne Soete
- Department of Biomedical Molecular Biology, Faculty of Science, Ghent University, Ghent, Belgium; Data Mining and Modeling for Biomedicine, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Romain Merceron
- Unit for Structural Biology, Department of Biochemistry and Microbiology Ghent University, Technologiepark 71, 9052 Ghent, Belgium; Unit for Structural Biology, VIB-UGent Center for Inflammation Research, Technologiepark 71, 9052 Ghent, Belgium
| | - Casey Murphy
- Unit for Structural Biology, Department of Biochemistry and Microbiology Ghent University, Technologiepark 71, 9052 Ghent, Belgium; Unit for Structural Biology, VIB-UGent Center for Inflammation Research, Technologiepark 71, 9052 Ghent, Belgium
| | - Mathias Provost
- Unit for Structural Biology, Department of Biochemistry and Microbiology Ghent University, Technologiepark 71, 9052 Ghent, Belgium; Unit for Structural Biology, VIB-UGent Center for Inflammation Research, Technologiepark 71, 9052 Ghent, Belgium
| | - J Fernando Bazan
- Unit for Structural Biology, VIB-UGent Center for Inflammation Research, Technologiepark 71, 9052 Ghent, Belgium; ħ Bioconsulting, Stillwater, MN, USA
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jonathan A Hill
- Surface Oncology, 50 Hampshire Street, Cambridge, MA 02139, USA.
| | - Savvas N Savvides
- Unit for Structural Biology, Department of Biochemistry and Microbiology Ghent University, Technologiepark 71, 9052 Ghent, Belgium; Unit for Structural Biology, VIB-UGent Center for Inflammation Research, Technologiepark 71, 9052 Ghent, Belgium.
| |
Collapse
|
20
|
Role of Innate and Adaptive Cytokines in the Survival of COVID-19 Patients. Int J Mol Sci 2022; 23:ijms231810344. [PMID: 36142255 PMCID: PMC9499609 DOI: 10.3390/ijms231810344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/02/2022] [Accepted: 09/02/2022] [Indexed: 11/16/2022] Open
Abstract
SARS-CoV-2 is a new coronavirus characterized by a high infection and transmission capacity. A significant number of patients develop inadequate immune responses that produce massive releases of cytokines that compromise their survival. Soluble factors are clinically and pathologically relevant in COVID-19 survival but remain only partially characterized. The objective of this work was to simultaneously study 62 circulating soluble factors, including innate and adaptive cytokines and their soluble receptors, chemokines and growth and wound-healing/repair factors, in severe COVID-19 patients who survived compared to those with fatal outcomes. Serum samples were obtained from 286 COVID-19 patients and 40 healthy controls. The 62 circulating soluble factors were quantified using a Luminex Milliplex assay. Results. The patients who survived had decreased levels of the following 30 soluble factors of the 62 studied compared to those with fatal outcomes, therefore, these decreases were observed for cytokines and receptors predominantly produced by the innate immune system—IL-1α, IL-1α, IL-18, IL-15, IL-12p40, IL-6, IL-27, IL-1Ra, IL-1RI, IL-1RII, TNFα, TGFα, IL-10, sRAGE, sTNF-RI and sTNF-RII—for the chemokines IL-8, IP-10, MCP-1, MCP-3, MIG and fractalkine; for the growth factors M-CSF and the soluble receptor sIL2Ra; for the cytokines involved in the adaptive immune system IFNγ, IL-17 and sIL-4R; and for the wound-repair factor FGF2. On the other hand, the patients who survived had elevated levels of the soluble factors TNFβ, sCD40L, MDC, RANTES, G-CSF, GM-CSF, EGF, PDGFAA and PDGFABBB compared to those who died. Conclusions. Increases in the circulating levels of the sCD40L cytokine; MDC and RANTES chemokines; the G-CSF and GM-CSF growth factors, EGF, PDGFAA and PDGFABBB; and tissue-repair factors are strongly associated with survival. By contrast, large increases in IL-15, IL-6, IL-18, IL-27 and IL-10; the sIL-1RI, sIL1RII and sTNF-RII receptors; the MCP3, IL-8, MIG and IP-10 chemokines; the M-CSF and sIL-2Ra growth factors; and the wound-healing factor FGF2 favor fatal outcomes of the disease.
Collapse
|
21
|
The role of temporal changes of pro-inflammatory cytokines in the development of adverse cardiac remodeling after ST-elevation myocardial infarction. Adv Cardiol 2022; 18:217-227. [PMID: 36751290 PMCID: PMC9885240 DOI: 10.5114/aic.2022.120938] [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: 08/04/2022] [Accepted: 10/10/2022] [Indexed: 11/18/2022]
Abstract
Introduction Increasing evidence supports the view that pro-inflammatory cytokines play a role in fibrosis after myocardial infarction (MI). It has been suggested that interleukin (IL)-12p40, a pro-inflammatory cytokine, can induce interferon γ (IFN-γ) and matrix metalloproteinase (MMP). However, the role of IL-12p40 in adverse cardiac remodeling (AR) after ST-elevation MI (STEMI) is unclear. Aim To examine the role of temporal changes of pro-inflammatory cytokines in the development of post-STEMI AR. Material and methods A total of 43 patients with STEMI for the first time ever were prospectively analyzed. In cardiac magnetic resonance imaging at 6 months after STEMI, a decrease of left ventricular end-diastolic volume by ≥ 12% was defined as reverse cardiac remodeling (RR), and a 12% increase was defined as AR. Cytokine concentrations were measured on the first day (baseline) and 2 weeks after STEMI. Results Mean IL-12p40 (59.1 ±14.5 vs. 46.7 ±9.1 pq/ml, p = 0.001), median IFN-γ (20.4 vs. 16.2 pq/ml, p = 0.048) and median MMP-2 (33866 vs. 20691 pq/ml, p = 0.011) baseline concentrations were higher in AR than RR. In patients with AR, IL-12p40 level was lower at 2 weeks than baseline (p < 0.001). There was a positive correlation between the baseline concentrations of IL-12p40, IFN-γ, MMP-2, C-reactive protein and infarct size (p < 0.05). Increased IL-12p40 and MMP-2 baseline levels were independently associated with AR (OR = 1.14, p = 0.010; OR = 1.08, p = 0.035). Conclusions In the initial phase of MI, greater release of pro-inflammatory cytokines was associated with increased MMP-2 levels. Elevated expression of IL-12 and MMP-2 had an independent association with AR. This may be related to the excessive inflammatory response in the initial phase of MI.
Collapse
|
22
|
Zhang H, Jiang HL, Dai SM. No Significant Effects of IL-23 on Initiating and Perpetuating the Axial Spondyloarthritis: The Reasons for the Failure of IL-23 Inhibitors. Front Immunol 2022; 13:818413. [PMID: 35222393 PMCID: PMC8868936 DOI: 10.3389/fimmu.2022.818413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/19/2022] [Indexed: 11/13/2022] Open
Abstract
Axial spondyloarthritis (axSpA) is comprised of ankylosing spondylitis (AS) and non-radiographic axSpA. In recent years, the involvement of the interleukin (IL)-23/IL-17 axis in the pathophysiology of axSpA has been widely proposed. Since IL-23 is an upstream activating cytokine of IL-17, theoretically targeting IL-23 should be effective in axSpA, especially after the success of the treatment with IL-17 blockers in the disorder. Unfortunately, IL-23 blockade did not show meaningful efficacy in clinical trials of AS. In this review, we analyzed the possible causes of the failure of IL-23 blockers in AS: 1) the available data from an animal model is not able to support that IL-23 is involved in a preclinical rather than clinical phase of axSpA; 2) Th17 cells are not principal inflammatory cells in the pathogenesis of axSpA; 3) IL-17 may be produced independently of IL-23 in several immune cell types other than Th17 cells in axSpA; 4) no solid evidence supports IL-23 as a pathogenic factor to induce enthesitis and bone formation. Taken together, IL-23 is not a principal proinflammatory cytokine in the pathogenesis of axSpA.
Collapse
Affiliation(s)
| | | | - Sheng-Ming Dai
- Department of Rheumatology & Immunology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| |
Collapse
|
23
|
Huntula S, Lalert L, Punsawad C. The Effects of Exercise on Aging-Induced Exaggerated Cytokine Responses: An Interdisciplinary Discussion. SCIENTIFICA 2022; 2022:3619362. [PMID: 35106183 PMCID: PMC8801319 DOI: 10.1155/2022/3619362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 01/10/2022] [Indexed: 06/14/2023]
Abstract
Aging is generally known to be associated with dynamic biological changes, physiological dysfunction, and environmental and psychological decline. Several studies have suggested that aging is associated with increased inflammatory cytokines, causing several diseases. However, the effect of exercise on aging has been less delineated, and the relationships between cytokine activation, aging, and exercise also need further study. Here, we discuss some ideas about the effect of exercise on aging-induced exaggerated cytokine responses and discuss the possible roles of the aging-induced exaggerated cytokine response following exercise. Evidence from these findings suggests that exercise is a beneficially applicable model to use in studies on the mechanisms underlying the age-associated gradated cytokine response, and these results may provide guidelines for health professionals with diverse backgrounds.
Collapse
Affiliation(s)
- Soontaraporn Huntula
- Department of Sport and Exercise Science, School of Medicine, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Laddawan Lalert
- Department of Medical Science, School of Medicine, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Chuchard Punsawad
- Department of Medical Science, School of Medicine, Walailak University, Nakhon Si Thammarat 80160, Thailand
- Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat 80160, Thailand
| |
Collapse
|
24
|
Lay CS, Bridges A, Goulding J, Briddon SJ, Soloviev Z, Craggs PD, Hill SJ. Probing the binding of interleukin-23 to individual receptor components and the IL-23 heteromeric receptor complex in living cells using NanoBRET. Cell Chem Biol 2022; 29:19-29.e6. [PMID: 34038748 PMCID: PMC8790524 DOI: 10.1016/j.chembiol.2021.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/06/2021] [Accepted: 05/04/2021] [Indexed: 02/07/2023]
Abstract
Interleukin-23 (IL-23) is a pro-inflammatory cytokine involved in the host defense against pathogens but is also implicated in the development of several autoimmune disorders. The IL-23 receptor has become a key target for drug discovery, but the exact mechanism of the receptor ligand interaction remains poorly understood. In this study the affinities of IL-23 for its individual receptor components (IL23R and IL12Rβ1) and the heteromeric complex formed between them have been measured in living cells using NanoLuciferase-tagged full-length proteins. Here, we demonstrate that TAMRA-tagged IL-23 has a greater than 7-fold higher affinity for IL12Rβ1 than IL23R. However, in the presence of both receptor subunits, IL-23 affinity is increased more than three orders of magnitude to 27 pM. Furthermore, we show that IL-23 induces a potent change in the position of the N-terminal domains of the two receptor subunits, consistent with a conformational change in the heteromeric receptor structure.
Collapse
Affiliation(s)
- Charles S Lay
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK; Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, The Midlands, UK; Medicine Design, Medicinal Science and Technology, GlaxoSmithKline, Stevenage SG1 2NY, UK
| | - Angela Bridges
- Protein and Cellular Sciences, Medicinal Science and Technology, GlaxoSmithKline, Stevenage SG1 2NY, UK
| | - Joelle Goulding
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK; Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, The Midlands, UK
| | - Stephen J Briddon
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK; Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, The Midlands, UK
| | - Zoja Soloviev
- Protein and Cellular Sciences, Medicinal Science and Technology, GlaxoSmithKline, Stevenage SG1 2NY, UK
| | - Peter D Craggs
- Medicine Design, Medicinal Science and Technology, GlaxoSmithKline, Stevenage SG1 2NY, UK; GSK-Francis Crick Institute Linklabs, Medicinal Science and Technology, GlaxoSmithKline, Stevenage SG1 2NY, UK.
| | - Stephen J Hill
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK; Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, The Midlands, UK.
| |
Collapse
|
25
|
Ritter K, Rousseau J, Hölscher C. Interleukin-27 in Tuberculosis: A Sheep in Wolf’s Clothing? Front Immunol 2022; 12:810602. [PMID: 35116036 PMCID: PMC8803639 DOI: 10.3389/fimmu.2021.810602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 12/22/2021] [Indexed: 12/12/2022] Open
Abstract
In tuberculosis (TB), protective inflammatory immune responses and the pathological sequelae of chronic inflammation significantly depend on a timely balance of cytokine expression. In contrast to other anti-inflammatory cytokines, interleukin (IL)-27 has fundamental effects in experimental Mycobacterium tuberculosis (Mtb) infection: the absence of IL-27-mediated signalling promotes a better control of mycobacterial growth on the one hand side but also leads to a chronic hyperinflammation and immunopathology later during infection. Hence, in the context of novel host-directed therapeutic approaches and vaccination strategies for the management of TB, the timely restricted blockade of IL-27 signalling may represent an advanced treatment option. In contrast, administration of IL-27 itself may allow to treat the immunopathological consequences of chronic TB. In both cases, a better knowledge of the cell type-specific and kinetic effects of IL-27 after Mtb infection is essential. This review summarizes IL-27-mediated mechanisms affecting protection and immunopathology in TB and discusses possible therapeutic applications.
Collapse
Affiliation(s)
- Kristina Ritter
- Infection Immunology, Research Centre Borstel, Borstel, Germany
| | - Jasmin Rousseau
- Infection Immunology, Research Centre Borstel, Borstel, Germany
| | - Christoph Hölscher
- Infection Immunology, Research Centre Borstel, Borstel, Germany
- German Centre for Infection Research (DZIF), Partner Site Hamburg-Borstel-Lübeck-Riems, Borstel, Germany
- *Correspondence: Christoph Hölscher,
| |
Collapse
|
26
|
Branisteanu D, Cojocaru C, Diaconu R, Porumb E, Alexa A, Nicolescu A, Brihan I, Bogdanici C, Branisteanu G, Dimitriu A, Zemba M, Anton N, Toader M, Grechin A, Branisteanu D. Update on the etiopathogenesis of psoriasis (Review). Exp Ther Med 2022; 23:201. [PMID: 35126704 PMCID: PMC8794554 DOI: 10.3892/etm.2022.11124] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 11/22/2021] [Indexed: 11/05/2022] Open
Affiliation(s)
- Daciana Branisteanu
- Department of Dermatology, ‘Grigore T. Popa’ University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Catalina Cojocaru
- Department of Dermatology, Railway Clinical Hospital, 700506 Iasi, Romania
| | - Roxana Diaconu
- Department of Dermatology, Railway Clinical Hospital, 700506 Iasi, Romania
| | - Elena Porumb
- Department of Dermatology, ‘Sf. Spiridon’ Clinical Emergency County Hospital, 700111 Iasi, Romania
| | - Anisia Alexa
- Department of Ophthalmology, ‘Grigore T. Popa’ University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Alin Nicolescu
- Department of Dermatology, ‘Roma’ Medical Center for Diagnosis and Treatment, 011773 Bucharest, Romania
| | - Ilarie Brihan
- Department of Dermatology, Dermatology Clinic, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | - Camelia Bogdanici
- Department of Ophthalmology, ‘Grigore T. Popa’ University of Medicine and Pharmacy, 700115 Iași, Romania
| | - George Branisteanu
- Faculty of Medicine, ‘Grigore T. Popa’ University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Andreea Dimitriu
- Department of Dermatology, ‘Arcadia’ Hospitals and Medical Centers, 700620 Iasi, Romania
| | - Mihail Zemba
- Department of Ophthalmology, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Nicoleta Anton
- Department of Ophthalmology, ‘Grigore T. Popa’ University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Mihaela Toader
- Department of Oral Dermatology, ‘Grigore T. Popa’ University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Adrian Grechin
- Department of Ophthalmology, ‘Sf. Spiridon’ Clinical Emergency County Hospital, 700111 Iasi, Romania
| | - Daniel Branisteanu
- Department of Ophthalmology, ‘Grigore T. Popa’ University of Medicine and Pharmacy, 700115 Iași, Romania
| |
Collapse
|
27
|
Ye C, Yano H, Workman CJ, Vignali DAA. Interleukin-35: Structure, Function and Its Impact on Immune-Related Diseases. J Interferon Cytokine Res 2021; 41:391-406. [PMID: 34788131 DOI: 10.1089/jir.2021.0147] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The balance between inflammatory and anti-inflammatory immune responses is maintained through immunoregulatory cell populations and immunosuppressive cytokines. Interleukin-35 (IL-35), an inhibitory cytokine that belongs to the IL-12 family, is capable of potently suppressing T cell proliferation and inducing IL-35-producing induced regulatory T cells (iTr35) to limit inflammatory responses. Over the past decade, a growing number of studies have indicated that IL-35 plays an important role in controlling immune-related disorders, including autoimmune diseases, infectious diseases, and cancer. In this review, we summarize the current knowledge about the biology of IL-35 and its contribution in different diseases, and we discuss the potential of and barriers to harnessing IL-35 as a clinical biomarker or immunotherapy.
Collapse
Affiliation(s)
- Cheng Ye
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Hiroshi Yano
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY, USA
| | - Creg J Workman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA.,Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
28
|
Corylin Ameliorates LPS-Induced Acute Lung Injury via Suppressing the MAPKs and IL-6/STAT3 Signaling Pathways. Pharmaceuticals (Basel) 2021; 14:ph14101046. [PMID: 34681270 PMCID: PMC8537250 DOI: 10.3390/ph14101046] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/08/2021] [Accepted: 10/08/2021] [Indexed: 12/18/2022] Open
Abstract
Acute lung injury (ALI) is a high mortality disease with acute inflammation. Corylin is a compound isolated from the whole plant of Psoralea corylifolia L. and has been reported to have anti-inflammatory activities. Herein, we investigated the therapeutic potential of corylin on lipopolysaccharides (LPS)-induced ALI, both in vitro and in vivo. The levels of proinflammatory cytokine secretions were analyzed by ELISA; the expressions of inflammation-associated proteins were detected using Western blot; and the number of immune cell infiltrations in the bronchial alveolar lavage fluid (BALF) were detected by multicolor flow cytometry and lung tissues by hematoxylin and eosin (HE) staining, respectively. Experimental results indicated that corylin attenuated LPS-induced IL-6 production in human bronchial epithelial cells (HBEC3-KT cells). In intratracheal LPS-induced ALI mice, corylin attenuated tissue damage, suppressed inflammatory cell infiltration, and decreased IL-6 and TNF-α secretions in the BALF and serum. Moreover, it further inhibited the phosphorylation of mitogen-activated protein kinases (MAPKs), including p-JNK, p-ERK, p-p38, and repressed the activation of signal transducer and activator of transcription 3 (STAT3) in lungs. Collectively, our results are the first to demonstrate the anti-inflammatory effects of corylin on LPS-induced ALI and suggest corylin has significant potential as a novel therapeutic agent for ALI.
Collapse
|
29
|
Watanabe A, Mizoguchi I, Hasegawa H, Katahira Y, Inoue S, Sakamoto E, Furusaka Y, Sekine A, Miyakawa S, Murakami F, Xu M, Yoneto T, Yoshimoto T. A Chaperone-Like Role for EBI3 in Collaboration With Calnexin Under Inflammatory Conditions. Front Immunol 2021; 12:757669. [PMID: 34603342 PMCID: PMC8484754 DOI: 10.3389/fimmu.2021.757669] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 09/02/2021] [Indexed: 01/31/2023] Open
Abstract
The interleukin-6 (IL-6)/IL-12 family of cytokines plays critical roles in the induction and regulation of innate and adaptive immune responses. Among the various cytokines, only this family has the unique characteristic of being composed of two distinct subunits, α- and β-subunits, which form a heterodimer with subunits that occur in other cytokines as well. Recently, we found a novel intracellular role for one of the α-subunits, Epstein-Barr virus-induced gene 3 (EBI3), in promoting the proper folding of target proteins and augmenting its expression at the protein level by binding to its target protein and a well-characterized lectin chaperone, calnexin, presumably through enhancing chaperone activity. Because calnexin is ubiquitously and constitutively expressed but EBI3 expression is inducible, these results could open an avenue to establish a new paradigm in which EBI3 plays an important role in further increasing the expression of target molecules at the protein level in collaboration with calnexin under inflammatory conditions. This theory well accounts for the heterodimer formation of EBI3 with p28, and probably with p35 and p19 to produce IL-27, IL-35, and IL-39, respectively. In line with this concept, another β-subunit, p40, plays a critical role in the assembly-induced proper folding of p35 and p19 to produce IL-12 and IL-23, respectively. Thus, chaperone-like activities in proper folding and maturation, which allow the secretion of biologically active heterodimeric cytokines, have recently been highlighted. This review summarizes the current understanding of chaperone-like activities of EBI3 to form heterodimers and other associations together with their possible biological implications.
Collapse
Affiliation(s)
- Aruma Watanabe
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Izuru Mizoguchi
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Hideaki Hasegawa
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Yasuhiro Katahira
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Shinya Inoue
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Eri Sakamoto
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Yuma Furusaka
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Ami Sekine
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Satomi Miyakawa
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Fumihiro Murakami
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Mingli Xu
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Toshihiko Yoneto
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Takayuki Yoshimoto
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
30
|
Chen Q, Zhang Z, Chen S, Chen J, Cheng Y, Liu A, Li B, Chen Z, Zheng Y, Ga M, Du L, Wang F. Genome-Wide Differential Expression Profiling of Pulmonary circRNAs Associated With Immune Reaction to Pasteurella multocida in Goats. Front Vet Sci 2021; 8:615405. [PMID: 34235193 PMCID: PMC8256745 DOI: 10.3389/fvets.2021.615405] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 04/08/2021] [Indexed: 01/26/2023] Open
Abstract
Pasteurella multocida is a highly versatile pathogen that infects a wide range of animals, including goats, causing pneumonia and hemorrhagic septicemia. Circular RNA (circRNA) is a type of non-coding RNA that plays an important role in regulating cellular metabolism. However, whether and how circRNA is involved in regulating immune responses in the goat lung has not been reported. Thus, this study was designed to examine the function of circRNA in goats infected with Pasteurella multocida. Goats were assigned into one of two groups: an uninfected control group (CK) and an infected group challenged with P. multocida. Compared with the CK group, which remained healthy, the infected goats showed clinical signs of infection, including depression, cough, nasal discharge, and dyspnea, along with elevated body temperature and lesions in the lung. Whole-transcriptome sequencing and small RNA sequencing were then performed using lung samples from goats from each group. A total of 138 circRNA, 56 microRNAs (miRNA), and 2,673 messenger RNA (mRNA) molecules were significantly differentially expressed in the P. multocida-infected group compared with the CK group. Randomly selected differentially expressed circRNA, miRNA, and mRNA molecules (n = 5 per group) were then validated by quantitative reverse-transcriptase polymerase chain reaction analysis. Gene ontology (GO) analysis of the source genes indicated that six immune-related terms were enriched among the differentially expressed cirRNA molecules, including inflammatory response, immune effector process, cell activation involved in immune response, cytokine-mediated signaling pathway, response to endogenous stimulus, and immune response. The corresponding circRNA molecules were then selected for construction of a competitive endogenous RNA network to identify networks that may be involved in the immune response to P. multocida infection. The results indicated that P. multocida HN01 may cause pneumonia and stimulate an immune response in goats via regulation of circRNA expression. This study presents the first comprehensive circRNA profile in response to P. multocida infection in goats, thus, providing a basis for understanding the function of circRNA in the host immune response to P. multocida infection.
Collapse
Affiliation(s)
- Qiaoling Chen
- Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, College of Animal Science and Technology, Hainan University, Haikou, China
| | - Zhenxing Zhang
- Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, College of Animal Science and Technology, Hainan University, Haikou, China
| | - Si Chen
- Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, College of Animal Science and Technology, Hainan University, Haikou, China
| | - Jie Chen
- Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, College of Animal Science and Technology, Hainan University, Haikou, China
| | - Yiwen Cheng
- Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, College of Animal Science and Technology, Hainan University, Haikou, China
| | - Ang Liu
- Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, College of Animal Science and Technology, Hainan University, Haikou, China
| | - Bin Li
- Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, College of Animal Science and Technology, Hainan University, Haikou, China
| | - Zhen Chen
- Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, College of Animal Science and Technology, Hainan University, Haikou, China
| | - Yiying Zheng
- Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, College of Animal Science and Technology, Hainan University, Haikou, China
| | - Manchuriga Ga
- Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, College of Animal Science and Technology, Hainan University, Haikou, China
| | - Li Du
- Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, College of Animal Science and Technology, Hainan University, Haikou, China
| | - Fengyang Wang
- Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, College of Animal Science and Technology, Hainan University, Haikou, China
| |
Collapse
|
31
|
Mohd Shukri ND, Farah Izati A, Wan Ghazali WS, Che Hussin CM, Wong KK. CD3 +CD4 +gp130 + T Cells Are Associated With Worse Disease Activity in Systemic Lupus Erythematosus Patients. Front Immunol 2021; 12:675250. [PMID: 34149710 PMCID: PMC8213373 DOI: 10.3389/fimmu.2021.675250] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 05/10/2021] [Indexed: 01/02/2023] Open
Abstract
The receptors for IL-35, IL-12Rβ2 and gp130, have been implicated in the inflammatory pathophysiology of autoimmune diseases. In this study, we set out to investigate the serum IL-35 levels and the surface levels of IL-12Rβ2 and gp130 in CD3+CD4+, CD3+CD4─ and CD3─CD4─ lymphocyte subpopulations in systemic lupus erythematosus (SLE) patients (n=50) versus healthy controls (n=50). The potential T cell subsets associated with gp130 transcript (i.e. IL6ST) expression in CD4+ T cells of SLE patients was also examined in publicly-available gene expression profiling (GEP) datasets. Here, we report that serum IL-35 levels were significantly higher in SLE patients than healthy controls (p=0.038) but it was not associated with SLEDAI-2K scores. The proportions of IL-12Rβ2+ and gp130+ cells in SLE patients did not differ significantly with those of healthy controls in all lymphocyte subpopulations investigated. Essentially, higher SLEDAI-2K scores were positively correlated with increased proportion of gp130+ cells, but not IL-12Rβ2+ cells, on CD3+CD4+ T cells (r=0.425, p=0.002, q=0.016). Gene Set Enrichment Analysis (GSEA) of a GEP dataset of CD4+ T cells isolated from SLE patients (n=8; GSE4588) showed that IL6ST expression was positively associated with genes upregulated in CD4+ T cells vs myeloid or B cells (q<0.001). In an independent GEP dataset of CD4+ T cells isolated from SLE patients (n=9; GSE1057), IL6ST expression was induced upon anti-CD3 stimulation, and that Treg, TCM and CCR7+ T cells gene sets were significantly enriched (q<0.05) by genes highly correlated with IL6ST expression (n=92 genes; r>0.75 with IL6ST expression) upon anti-CD3 stimulation in these SLE patients. In conclusion, gp130 signaling in CD3+CD4+ T cell subsets may contribute to increased disease activity in SLE patients, and it represents a promising therapeutic target for inhibition in the disease.
Collapse
Affiliation(s)
- Nur Diyana Mohd Shukri
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia.,Hospital Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Aziz Farah Izati
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia.,Hospital Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Wan Syamimee Wan Ghazali
- Hospital Universiti Sains Malaysia, Kubang Kerian, Malaysia.,Department of Internal Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Che Maraina Che Hussin
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia.,Hospital Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Kah Keng Wong
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia.,Hospital Universiti Sains Malaysia, Kubang Kerian, Malaysia
| |
Collapse
|
32
|
Guo K, Zhang X. Cytokines that Modulate the Differentiation of Th17 Cells in Autoimmune Uveitis. J Immunol Res 2021; 2021:6693542. [PMID: 33816637 PMCID: PMC7990547 DOI: 10.1155/2021/6693542] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/01/2021] [Accepted: 03/04/2021] [Indexed: 02/06/2023] Open
Abstract
Increasing evidence has suggested that T helper 17 (Th17) cells play a central role in the pathogenesis of ocular immune disease. The association between pathogenic Th17 cells and the development of uveitis has been confirmed in experimental and clinical studies. Several cytokines affect the initiation and stabilization of the differentiation of Th17 cells. Therefore, understanding the mechanism of related cytokines in the differentiation of Th17 cells is important for exploring the pathogenesis and the potential therapeutic targets of uveitis. This article briefly describes the structures, mechanisms, and targeted drugs of cytokines-including interleukin (IL)-6, transforming growth factor-β1 (TGF-β1), IL-1β, IL-23, IL-27, IL-35, IL-2, IL-4, IL-21, and interferon (IFN)-γ-which have an important influence on the differentiation of Th17 cells and discusses their potential as therapeutic targets for treating autoimmune uveitis.
Collapse
Affiliation(s)
- Kailei Guo
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300384, China
| | - Xiaomin Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300384, China
| |
Collapse
|
33
|
The immunological impact of adenovirus early genes on vaccine-induced responses in mice and nonhuman primates. J Virol 2021; 95:JVI.02253-20. [PMID: 33441339 PMCID: PMC8092689 DOI: 10.1128/jvi.02253-20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Adenovirus (Ad) is being explored for use in the prevention and treatment of a variety of infectious diseases and cancers. Ad with a deletion in early region 3 (ΔE3) provokes a stronger immune response than Ad with deletions in early regions 1 and E3 (ΔE1/ΔE3). The ΔE1/ΔE3 Ads are more popular because they can carry a larger transgene and because of the deleted E1 (E1A and E1B), are perceived safer for clinical use. Ad with a deletion in E1B55K (ΔE1B55K) has been in phase III clinical trials for use in cancer therapy in the US and has been approved for use in head and neck tumor therapy in China, demonstrating that Ad containing E1A are safe for clinical use. We have shown previously that ΔE1B55K Ad, even while promoting lower levels of an inserted transgene, promoted similar levels of transgene-specific immune responses as a ΔE3 Ad. Products of the Ad early region 4 (E4) limit the ability of cells to mount an innate immune response. Using this knowledge, we deleted the Ad E4 open reading frames 1-4 (E4orf1-4) from the ΔE1B55K Ad. Here, we show that innate cytokine network genes are elevated in the ΔE4 Ad-infected cells beyond that of ΔE3 Ad-infected cells. Further, in immunized mice the IgG2a subclass was favored as was the IgG1 subclass in immunized nonhuman primates. Thus, Ad E4 impacts immune responses in cells, in immunized mice, and immunized nonhuman primates. These Ad may offer advantages that are beneficial for clinical use.Importance: Adenovirus (Ad) is being explored for use in the prevention and treatment of a variety of infectious diseases and cancers. Here we provide evidence in cells, mice, and nonhuman primates supporting the notion that Ad early gene-products limit specific immune responses. Ad constructed with deletions in early genes and expressing HIV envelope protein was shown to induce greater HIV-specific cellular immune responses and higher titer antibodies compared to the parental Ad with the early genes. In addition to eliciting enhanced immunity, the deleted Ad possesses more space for insertion of additional or larger transgenes needed for targeting other infectious agents or cancers.
Collapse
|
34
|
Hasegawa H, Mizoguchi I, Orii N, Inoue S, Katahira Y, Yoneto T, Xu M, Miyazaki T, Yoshimoto T. IL-23p19 and CD5 antigen-like form a possible novel heterodimeric cytokine and contribute to experimental autoimmune encephalomyelitis development. Sci Rep 2021; 11:5266. [PMID: 33664371 PMCID: PMC7933155 DOI: 10.1038/s41598-021-84624-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 02/18/2021] [Indexed: 02/07/2023] Open
Abstract
Among various cytokines, interleukin (IL)-12 family cytokines have very unique characteristics in that they are composed of two distinct subunits and these subunits are shared with each other. IL-23, one of the IL-12 family cytokines, consists of p19 and p40 subunits, is mainly produced by antigen-presenting cells, and plays a critical role in the expansion and maintenance of pathogenic helper CD4+ T (Th)17 cells. Since we initially found that p19 is secreted in the culture supernatant of activated CD4+ T cells, we have further investigated the role of p19. p19 was revealed to associate with CD5 antigen-like (CD5L), which is a repressor of Th17 pathogenicity and is highly expressed in non-pathogenic Th17 cells, to form a composite p19/CD5L. This p19/CD5L was shown to activate STAT5 and enhance the differentiation into granulocyte macrophage colony-stimulating factor (GM-CSF)-producing CD4+ T cells. Both CD4+ T cell-specific conditional p19-deficient mice and complete CD5L-deficient mice showed significantly alleviated experimental autoimmune encephalomyelitis (EAE) with reduced frequency of GM-CSF+CD4+ T cells. During the course of EAE, the serum level of p19/CD5L, but not CD5L, correlated highly with the clinical symptoms. Thus, the composite p19/CD5L is a possible novel heterodimeric cytokine that contributes to EAE development with GM-CSF up-regulation.
Collapse
Affiliation(s)
- Hideaki Hasegawa
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Izuru Mizoguchi
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Naoko Orii
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Shinya Inoue
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Yasuhiro Katahira
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Toshihiko Yoneto
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Mingli Xu
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Toru Miyazaki
- Laboratory of Molecular Biomedicine for Pathogenesis, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Takayuki Yoshimoto
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan.
| |
Collapse
|
35
|
Abstract
IL-6 is involved both in immune responses and in inflammation, hematopoiesis, bone metabolism and embryonic development. IL-6 plays roles in chronic inflammation (closely related to chronic inflammatory diseases, autoimmune diseases and cancer) and even in the cytokine storm of corona virus disease 2019 (COVID-19). Acute inflammation during the immune response and wound healing is a well-controlled response, whereas chronic inflammation and the cytokine storm are uncontrolled inflammatory responses. Non-immune and immune cells, cytokines such as IL-1β, IL-6 and tumor necrosis factor alpha (TNFα) and transcription factors nuclear factor-kappa B (NF-κB) and signal transducer and activator of transcription 3 (STAT3) play central roles in inflammation. Synergistic interactions between NF-κB and STAT3 induce the hyper-activation of NF-κB followed by the production of various inflammatory cytokines. Because IL-6 is an NF-κB target, simultaneous activation of NF-κB and STAT3 in non-immune cells triggers a positive feedback loop of NF-κB activation by the IL-6-STAT3 axis. This positive feedback loop is called the IL-6 amplifier (IL-6 Amp) and is a key player in the local initiation model, which states that local initiators, such as senescence, obesity, stressors, infection, injury and smoking, trigger diseases by promoting interactions between non-immune cells and immune cells. This model counters dogma that holds that autoimmunity and oncogenesis are triggered by the breakdown of tissue-specific immune tolerance and oncogenic mutations, respectively. The IL-6 Amp is activated by a variety of local initiators, demonstrating that the IL-6-STAT3 axis is a critical target for treating diseases.
Collapse
Affiliation(s)
- Toshio Hirano
- National Institutes for Quantum and Radiological Science and Technology, Anagawa, Inage-ku, Chiba, Japan
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
36
|
Mizoguchi I, Ohashi M, Hasegawa H, Chiba Y, Orii N, Inoue S, Kawana C, Xu M, Sudo K, Fujita K, Kuroda M, Hashimoto SI, Matsushima K, Yoshimoto T. EBV-induced gene 3 augments IL-23Rα protein expression through a chaperone calnexin. J Clin Invest 2021; 130:6124-6140. [PMID: 32809973 DOI: 10.1172/jci122732] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/05/2020] [Indexed: 12/27/2022] Open
Abstract
Epstein-Barr virus-induced gene 3 (EBI3) is a subunit common to IL-27, IL-35, and IL-39. Here, we explore an intracellular role of EBI3 that is independent of its function in cytokines. EBI3-deficient naive CD4+ T cells had reduced IFN-γ production and failed to induce T cell-dependent colitis in mice. Similarly reduced IFN-γ production was observed in vitro in EBI3-deficient CD4+ T cells differentiated under pathogenic Th17 polarizing conditions with IL-23. This is because the induction of expression of one of the IL-23 receptor (IL-23R) subunits, IL-23Rα, but not another IL-23R subunit, IL-12Rβ1, was selectively decreased at the protein level, but not the mRNA level. EBI3 augmented IL-23Rα expression via binding to the chaperone molecule calnexin and to IL-23Rα in a peptide-dependent manner, but not a glycan-dependent manner. Indeed, EBI3 failed to augment IL-23Rα expression in the absence of endogenous calnexin. Moreover, EBI3 poorly augmented the expression of G149R, an IL-23Rα variant that protects against the development of human colitis, because binding of EBI3 to the variant was reduced. Taken together with the result that EBI3 expression is inducible in T cells, the present results suggest that EBI3 plays a critical role in augmenting IL-23Rα protein expression via calnexin under inflammatory conditions.
Collapse
Affiliation(s)
- Izuru Mizoguchi
- Department of Immunoregulation, Institute of Medical Science
| | - Mio Ohashi
- Department of Immunoregulation, Institute of Medical Science
| | | | - Yukino Chiba
- Department of Immunoregulation, Institute of Medical Science
| | - Naoko Orii
- Department of Immunoregulation, Institute of Medical Science
| | - Shinya Inoue
- Department of Immunoregulation, Institute of Medical Science
| | - Chiaki Kawana
- Department of Immunoregulation, Institute of Medical Science
| | - Mingli Xu
- Department of Immunoregulation, Institute of Medical Science
| | | | - Koji Fujita
- Department of Molecular Pathology, Tokyo Medical University, Tokyo, Japan
| | - Masahiko Kuroda
- Department of Molecular Pathology, Tokyo Medical University, Tokyo, Japan
| | - Shin-Ichi Hashimoto
- Department of Laboratory Medicine, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Ishikawa, Japan
| | - Kouji Matsushima
- Department of Molecular Preventive Medicine, School of Medicine, University of Tokyo, Tokyo, Japan
| | | |
Collapse
|
37
|
Breban M, Glatigny S, Cherqaoui B, Beaufrère M, Lauraine M, Rincheval-Arnold A, Gaumer S, Guénal I, Araujo LM. Lessons on SpA pathogenesis from animal models. Semin Immunopathol 2021; 43:207-219. [PMID: 33449154 DOI: 10.1007/s00281-020-00832-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 12/15/2020] [Indexed: 12/31/2022]
Abstract
Understanding the complex mechanisms underlying a disorder such as spondyloarthritis (SpA) may benefit from studying animal models. Several suitable models have been developed, in particular to investigate the role of genetic factors predisposing to SpA, including HLA-B27, ERAP1, and genes related to the interleukin (IL)-23/IL-17 axis. One of the best examples of such research is the HLA-B27 transgenic rat model that fostered the emergence of original theories regarding HLA-B27 pathogenicity, including dysregulation of innate immunity, contribution of the adaptive immune system to chronic inflammation, and influence of the microbiota on disease development. Very recently, a new model of HLA-B27 transgenic Drosophila helped to expand further some of those theories in an unexpected direction involving the TGFβ/BMP family of mediators. On the other hand, several spontaneous, inducible, and/or genetically modified mouse models-including SKG mouse, TNFΔARE mouse and IL-23-inducible mouse model of SpA-have highlighted the importance of TNFα and IL-23/IL-17 axis in the development of SpA manifestations. Altogether, those animal models afford not only to study disease mechanism but also to investigate putative therapeutic targets.
Collapse
Affiliation(s)
- Maxime Breban
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, 2 ave de la Source de la Bièvre, 78180, Montigny-le-Bretonneux, France. .,Laboratoire d'Excellence Inflamex, Université Paris Descartes, Sorbonne-Paris-Cité, Paris, France. .,Service de Rhumatologie, Hôpital Ambroise Paré, AP-HP, 9 ave Charles de Gaulle, 92100, Boulogne, France.
| | - Simon Glatigny
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, 2 ave de la Source de la Bièvre, 78180, Montigny-le-Bretonneux, France.,Laboratoire d'Excellence Inflamex, Université Paris Descartes, Sorbonne-Paris-Cité, Paris, France
| | - Bilade Cherqaoui
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, 2 ave de la Source de la Bièvre, 78180, Montigny-le-Bretonneux, France.,Laboratoire d'Excellence Inflamex, Université Paris Descartes, Sorbonne-Paris-Cité, Paris, France
| | - Marie Beaufrère
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, 2 ave de la Source de la Bièvre, 78180, Montigny-le-Bretonneux, France.,Laboratoire d'Excellence Inflamex, Université Paris Descartes, Sorbonne-Paris-Cité, Paris, France
| | - Marc Lauraine
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, 2 ave de la Source de la Bièvre, 78180, Montigny-le-Bretonneux, France.,Laboratoire d'Excellence Inflamex, Université Paris Descartes, Sorbonne-Paris-Cité, Paris, France
| | - Aurore Rincheval-Arnold
- LGBC, EA4589, UVSQ/Université Paris-Saclay, EPHE/PSL Research University, 2 ave de la Source de la Bièvre, 78180, Montigny-le-Bretonneux, France
| | - Sébastien Gaumer
- LGBC, EA4589, UVSQ/Université Paris-Saclay, EPHE/PSL Research University, 2 ave de la Source de la Bièvre, 78180, Montigny-le-Bretonneux, France
| | - Isabelle Guénal
- LGBC, EA4589, UVSQ/Université Paris-Saclay, EPHE/PSL Research University, 2 ave de la Source de la Bièvre, 78180, Montigny-le-Bretonneux, France
| | - Luiza M Araujo
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, 2 ave de la Source de la Bièvre, 78180, Montigny-le-Bretonneux, France.,Laboratoire d'Excellence Inflamex, Université Paris Descartes, Sorbonne-Paris-Cité, Paris, France
| |
Collapse
|
38
|
Suzuki K, Hayashida H. Effect of Exercise Intensity on Cell-Mediated Immunity. Sports (Basel) 2021; 9:sports9010008. [PMID: 33440732 PMCID: PMC7826544 DOI: 10.3390/sports9010008] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/13/2020] [Accepted: 01/06/2021] [Indexed: 12/16/2022] Open
Abstract
Moderate-intensity exercise is considered to enhance immune function and to be useful for preventing acute upper respiratory infections and similar conditions. Many people practice low-intensity short-duration exercise with the expectation of a beneficial effect on immunocompetency. However, it is difficult to affirm the existence of definite evidence of such a benefit. In this article, we discuss the effects of low-intensity short-duration exercise on cell-mediated immunity, and contrast them to the effects of high-intensity and long-duration exercise. Whereas high-intensity exercise induces inflammation and reduces cell-mediated immune system function, low-intensity exercise does not appear to have a large effect on either inflammation or cell-mediated immune function. Low-intensity exercises such as walking and yoga, which are helpful to relieve stress, cannot be considered as harmful to the immune system. Although yoga was shown to impose fewer restrictions on breathing and physical strain, the evidence that yoga enhances cell-mediated immunity remains insufficient. Therefore, further studies are needed to examine the exercise mode that may be most effective for improvement of immune functions.
Collapse
Affiliation(s)
- Katsuhiko Suzuki
- Faculty of Sport Sciences, Waseda University, 2-579-15 Mikajima, Tokorozawa 359-1192, Japan
- Correspondence: ; Tel.: +81-4-2947-6898
| | - Harumi Hayashida
- Faculty of Culture and Sport Policy, Toin University of Yokohama, 1614 Kurogane-cho, Aoba-ku, Yokohama 225-8503, Japan;
| |
Collapse
|
39
|
Li Y, Gong L, Weng L, Pan X, Liu C, Li M. Interleukin-39 exacerbates concanavalin A-induced liver injury. Immunopharmacol Immunotoxicol 2021; 43:94-99. [PMID: 33412981 DOI: 10.1080/08923973.2020.1869778] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
BACKGROUND Interleukin (IL)-39 is a novel member of IL-12 family and has been reported to play a pro-inflammatory role in lupus-like mice, but its function in concanavalin A (ConA)-induced liver injury is currently unclear. MATERIALS AND METHODS In this study, we investigated the effects of IL-39 expression in a mouse model of ConA induced-hepatitis. We first showed that delivery of plasmid DNA encoding mouse IL-39 using the hydrodynamic tail vein injection method increased IL-39 mRNA and protein levels in the liver. We then administrated mice with IL-39 plasmid before ConA injection and measured serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels, inflammatory infiltration, and hepatocyte necrosis in the liver. Additionally, we further explored the potential mechanism of IL-39 in ConA-induced liver injury by measuring several inflammatory mediators. RESULTS We found that ectopic IL-39 expression promoted the ConA-induced increase in serum ALT and AST levels, inflammatory infiltration, and hepatocyte necrosis in the liver. We also observed that IL-39 plasmid administration significantly increased serum and liver interferon-γ, tumor necrosis factor-α, and IL-17A levels, but did not affect serum and liver IL-10 levels in ConA-induced hepatitis. CONCLUSION Our results suggest that IL-39 can exacerbate ConA-induced hepatitis and may be a therapeutic target in inflammatory liver disease.
Collapse
Affiliation(s)
- Yan Li
- The Affiliated Hospital of Medical School of Ningbo University, and Department of Immunology, Ningbo University School of Medicine, Ningbo, China
| | - Luping Gong
- The Affiliated Hospital of Medical School of Ningbo University, and Department of Immunology, Ningbo University School of Medicine, Ningbo, China.,School of Marine Sciences, Ningbo University, Ningbo, China
| | - Linjie Weng
- The Affiliated Hospital of Medical School of Ningbo University, and Department of Immunology, Ningbo University School of Medicine, Ningbo, China.,School of Marine Sciences, Ningbo University, Ningbo, China
| | - Xiuhe Pan
- The Affiliated Hospital of Medical School of Ningbo University, and Department of Immunology, Ningbo University School of Medicine, Ningbo, China
| | - Chaobo Liu
- The Affiliated Hospital of Medical School of Ningbo University, and Department of Immunology, Ningbo University School of Medicine, Ningbo, China
| | - Mingcai Li
- The Affiliated Hospital of Medical School of Ningbo University, and Department of Immunology, Ningbo University School of Medicine, Ningbo, China
| |
Collapse
|
40
|
Mirlekar B, Pylayeva-Gupta Y. IL-12 Family Cytokines in Cancer and Immunotherapy. Cancers (Basel) 2021; 13:E167. [PMID: 33418929 PMCID: PMC7825035 DOI: 10.3390/cancers13020167] [Citation(s) in RCA: 131] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/22/2020] [Accepted: 12/29/2020] [Indexed: 12/16/2022] Open
Abstract
The IL-12 family cytokines are a group of unique heterodimeric cytokines that include IL-12, IL-23, IL-27, IL-35 and, most recently, IL-39. Recent studies have solidified the importance of IL-12 cytokines in shaping innate and adaptive immune responses in cancer and identified multipronged roles for distinct IL-12 family members, ranging from effector to regulatory immune functions. These cytokines could serve as promising candidates for the development of immunomodulatory therapeutic approaches. Overall, IL-12 can be considered an effector cytokine and has been found to engage anti-tumor immunity by activating the effector Th1 response, which is required for the activation of cytotoxic T and NK cells and tumor clearance. IL-23 and IL-27 play dual roles in tumor immunity, as they can both activate effector immune responses and promote tumor growth by favoring immune suppression. IL-35 is a potent regulatory cytokine and plays a largely pro-tumorigenic role by inhibiting effector T cells. In this review, we summarize the recent findings on IL-12 family cytokines in the control of tumor growth with an emphasis primarily on immune regulation. We underscore the clinical implications for the use of these cytokines either in the setting of monotherapy or in combination with other conventional therapies for the more effective treatment of malignancies.
Collapse
Affiliation(s)
- Bhalchandra Mirlekar
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA;
| | - Yuliya Pylayeva-Gupta
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA;
- Department of Genetics, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA
| |
Collapse
|
41
|
Kang S, Narazaki M, Metwally H, Kishimoto T. Historical overview of the interleukin-6 family cytokine. J Exp Med 2020; 217:151633. [PMID: 32267936 PMCID: PMC7201933 DOI: 10.1084/jem.20190347] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 11/20/2019] [Accepted: 03/09/2020] [Indexed: 02/06/2023] Open
Abstract
Interleukin-6 (IL-6) has been identified as a 26-kD secreted protein that stimulates B cells to produce antibodies. Later, IL-6 was revealed to have various functions that overlap with other IL-6 family cytokines and use the common IL-6 signal transducer gp130. IL-6 stimulates cells through multiple pathways, using both membrane and soluble IL-6 receptors. As indicated by the expanding market for IL-6 inhibitors, it has become a primary therapeutic target among IL-6 family cytokines. Here, we revisit the discovery of IL-6; discuss insights regarding the roles of this family of cytokines; and highlight recent advances in our understanding of regulation of IL-6 expression.
Collapse
Affiliation(s)
- Sujin Kang
- Department of Immune Regulation, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Masashi Narazaki
- Department of Advanced Clinical and Translational Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hozaifa Metwally
- Department of Immune Regulation, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Tadamitsu Kishimoto
- Department of Immune Regulation, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| |
Collapse
|
42
|
Ecoeur F, Weiss J, Schleeger S, Guntermann C. Lack of evidence for expression and function of IL-39 in human immune cells. PLoS One 2020; 15:e0242329. [PMID: 33259477 PMCID: PMC7707563 DOI: 10.1371/journal.pone.0242329] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 11/02/2020] [Indexed: 01/22/2023] Open
Abstract
Members of the IL-6/IL-12 cytokine family are critical regulators of innate and adaptive immunity and have emerged as key players controlling inflammatory and autoimmune disorders. This cytokine family comprises of IL-12, IL-23, IL-27, and IL-35, each consisting of distinct α- and β-cytokine subunits that form heterodimers. A new member of this family, IL-39, was identified in the murine species and was shown to consist of the IL-23p19 and Epstein-Barr Virus-induced 3 (EBI3) subunits. Subsequently, it was shown that IL-39 was implicated in the immunopathogenesis of murine experimental lupus erythematosus. The existence of IL-39 in the human system has yet to be confirmed. Based on the clinical success of IL-23p19 neutralizing approaches in moderate-to-severe psoriasis, anti-IL-23p19 antibodies in the clinic may not only neutralize IL-23, but additionally IL-39, implying that IL-39 might also contribute to the pathogenesis of psoriasis. It is therefore pivotal to demonstrate IL-39 expression and to characterize its function in the human system. In this study, we provided evidence for the existence of secreted heterodimeric p19 and EBI3 complexes in supernatants originating from p19 and EBI3 transfected HEK293FT cells. We attempted to detect IL-39 expression from stimulated human primary B cells, human keratinocytes and in vitro polarized human macrophages. Whereas, the expression of p19 and EBI3 mRNA was elevated, we failed to detect p19 and EBI3 heterodimers. Functional assays were conducted with conditioned media containing human IL-39 or with a human recombinant IL-39 Fc protein. Immune cells targeted by IL-39 in mouse, such as neutrophils and PBMCs, did not respond to human IL-39 stimulation and IL-39 failed to activate STAT3 in a reporter cell line. These results suggest that, while the secretion of p19/EBI3 complexes can be forced in human cells, it is secreted below the lower quantity of detection or it has no functional role.
Collapse
Affiliation(s)
- Florence Ecoeur
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Jessica Weiss
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Simone Schleeger
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Christine Guntermann
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Basel, Switzerland
- * E-mail:
| |
Collapse
|
43
|
Huang F, Ju YH, Wang HB, Li YN. Maternal vitamin D deficiency impairs Treg and Breg responses in offspring mice and deteriorates allergic airway inflammation. Allergy Asthma Clin Immunol 2020; 16:89. [PMID: 33072159 PMCID: PMC7557044 DOI: 10.1186/s13223-020-00487-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 10/06/2020] [Indexed: 02/06/2023] Open
Abstract
Background Vitamin D (VitD) can regulate immune responses and maternal VitD-deficiency can affect immune responses in the offspring. This study aimed at investigating the effects of maternal VitD-deficiency during pregnancy on Treg and Breg responses in offspring mice with house dust mite (HDM)-induced allergic airway inflammation. Methods Female BALB/c mice were randomized and fed with normal chow or VitD-deficient diet until their offspring weaned. The offspring mice were fed with normal chow and injected with vehicle or HDM to induce allergic airway inflammation. The levels of serum 25(OH)D, cytokines and infiltrate numbers as well as percentages of Tregs and Bregs in the bronchoalveolar lavage fluid (BALF) were analyzed. The relative levels of VitD receptor (VDR), VitD-binding protein (VDBP), Cytochromes P450 (CYP) 27b1, and CYP24A1 mRNA transcripts in the lungs of different groups of mice were measured. Results Maternal VitD-deficiency significantly reduced serum 25(OH)D levels in offspring mice. VitD-deficiency significantly increased the relative levels of VDR, VDBP and CYP27B1 mRNA transcripts, but decreased CYP24A1 expression in the lungs of mice. In comparison with the control mice, significantly elevated levels of pro-inflammatory cytokines, increased numbers of lymphocytes and eosinophils, but decreased levels of anti-inflammatory cytokines were detected in the BALF of VitD-deficient mice. VitD-deficiency significantly increased the frequency of Th1, Th2, Th9, Th17 cells, but decreased regulatory T (Tregs) and B cells (Bregs) in the BALF of mice with allergic airway inflammation. Conclusion Maternal VitD-deficiency lowed serum 25(OH)D levels and enhanced HDM-induced allergic airway inflammation in the offspring by impairing Breg and Treg responses.
Collapse
Affiliation(s)
- Fei Huang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, 130033 Jilin China
| | - Yang-Hua Ju
- Department of Pediatrics, the First Hospital of Jilin University, Changchun, 130021 Jilin China
| | - Hong-Bo Wang
- Department of Pediatrics, the First Hospital of Jilin University, Changchun, 130021 Jilin China
| | - Ya-Nan Li
- Department of Pediatrics, the First Hospital of Jilin University, Changchun, 130021 Jilin China.,Institute of Pediatrics, the First Hospital of Jilin University, Changchun, 130021 Jilin China
| |
Collapse
|
44
|
Saha B, Bhattacharjee S, Sarkar A, Bhor R, Pai K, Bodhale N. Conundrums in leishmaniasis. Cytokine 2020; 145:155304. [PMID: 33004260 DOI: 10.1016/j.cyto.2020.155304] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 09/15/2020] [Accepted: 09/17/2020] [Indexed: 11/19/2022]
Abstract
Parasites of the genus Leishmania cause the disease leishmaniasis. As the sandfly vector transfers the promastigotes into the skin of the human host, the infection is either cured or exacerbated. In the process, there emerge several unsolved paradoxes of leishmaniasis. Chronologically, as the infections starts in skin, the role of the salivary proteins in supporting the infection or the host response to these proteins influencing the induction of immunological memory becomes a conundrum. As the parasite invokes inflammation, the infiltrating neutrophils may act as "Trojan Horse" to transfer parasites to macrophages that, along with dendritic cells, carry the parasite to lymphoid organs to start visceralization. As the visceralized infection becomes chronic, the acutely enhanced monocytopoiesis takes a downturn while neutropenia and thrombocytopenia ensue with concomitant rise in splenic colony-forming-units. These responses are accompanied by splenic and hepatic granulomas, polyclonal activation of B cells and deviation of T cell responses. The granuloma formation is both a containment process and a form of immunopathogenesis. The heterogeneity in neutrophils and macrophages contribute to both cure and progression of the disease. The differentiation of T-helper subsets presents another paradox of visceral leishmaniasis, as the counteractive T cell subsets influence the curing or non-curing outcome. Once the parasites are killed by chemotherapy, in some patients the cured visceral disease recurs as a cutaneous manifestation post-kala azar dermal leishmaniasis (PKDL). As no experimental model exists, the natural history of PKDL remains almost a black box at the end of the visceral disease.
Collapse
Affiliation(s)
- Baibaswata Saha
- Centre of Advanced Study, Department of Zoology, Savitribai Phule Pune University, Pune 411007, India
| | - Surajit Bhattacharjee
- Department of Molecular Biology and Bioinformatics, Tripura Central University, Agartala, India
| | - Arup Sarkar
- Trident Academy of Creative Technology, Bhubaneshwar, Odisha 751024, India
| | - Renuka Bhor
- Centre of Advanced Study, Department of Zoology, Savitribai Phule Pune University, Pune 411007, India
| | - Kalpana Pai
- Centre of Advanced Study, Department of Zoology, Savitribai Phule Pune University, Pune 411007, India
| | - Neelam Bodhale
- Jagadis Bose National Science Talent Search, 1300 Rajdanga Road, Kolkata 700107, India; National Centre for Cell Science, Ganeshkhind, Pune 411007, India.
| |
Collapse
|
45
|
IL-12 and IL-23-Close Relatives with Structural Homologies but Distinct Immunological Functions. Cells 2020; 9:cells9102184. [PMID: 32998371 PMCID: PMC7600943 DOI: 10.3390/cells9102184] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 09/24/2020] [Accepted: 09/27/2020] [Indexed: 12/21/2022] Open
Abstract
Cytokines of the IL-12 family show structural similarities but have distinct functions in the immune system. Prominent members of this cytokine family are the pro-inflammatory cytokines IL-12 and IL-23. These two cytokines share cytokine subunits and receptor chains but have different functions in autoimmune diseases, cancer and infections. Accordingly, structural knowledge about receptor complex formation is essential for the development of new therapeutic strategies preventing and/or inhibiting cytokine:receptor interaction. In addition, intracellular signaling cascades can be targeted to inhibit cytokine-mediated effects. Single nucleotide polymorphisms can lead to alteration in the amino acid sequence and thereby influencing protein functions or protein–protein interactions. To understand the biology of IL-12 and IL-23 and to establish efficient targeting strategies structural knowledge about cytokines and respective receptors is crucial. A highly efficient therapy might be a combination of different drugs targeting extracellular cytokine:receptor assembly and intracellular signaling pathways.
Collapse
|
46
|
Koohyanizadeh F, Karaji AG, Falahi S, Rezaiemanesh A, Salari F. In silico prediction of deleterious single nucleotide polymorphisms in human interleukin 27 (IL-27) gene. Meta Gene 2020. [DOI: 10.1016/j.mgene.2020.100710] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
47
|
Yang MG, Tian S, Zhang Q, Han J, Liu C, Zhou Y, Zhu J, Jin T. Elevated serum interleukin-39 levels in patients with neuromyelitis optica spectrum disorders correlated with disease severity. Mult Scler Relat Disord 2020; 46:102430. [PMID: 32853892 DOI: 10.1016/j.msard.2020.102430] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 07/17/2020] [Accepted: 07/29/2020] [Indexed: 01/10/2023]
Abstract
AIMS Neuromyelitis optica spectrum disorders (NMOSD) is an inflammatory demyelinating autoimmune disorder in the central nervous system (CNS), which is mainly mediated by aquaporin 4 antibodies (AQP4-Ab). Interleukin (IL)-39 is a new pro-inflammatory cytokine which belongs to the IL-12 cytokine family. However, the role of IL-39 in patients with NMOSD is not completely understood. Therefore, the aim of this study is to explore the possible implication of IL-39 in the pathogenesis of NMOSD. METHODS In this study, 50 patients with NMOSD, 20 patients with relapsing-remitting multiple sclerosis (RRMS), 30 patients with non-inflammatory neurological disorders (NND) and 78 healthy controls (HCs) were recruited. The levels of serum IL-39 were measured using the enzyme-linked immunosorbent assay (ELISA). RESULTS Our study showed serum IL-39 levels in patients with NMOSD were significantly higher than that in RRMS patients, NND patients and HCs, and positively correlated with Expanded Disability Status Scale (EDSS) score. CONCLUSION These findings suggested that IL-39 may play a pro-inflammatory role in the pathogenesis of NMOSD and correlate with disease severity.
Collapse
Affiliation(s)
- Meng-Ge Yang
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Changchun, China
| | - Suyan Tian
- Division of Clinical Research, the First Hospital of Jilin University, Changchun, China
| | - Qingxiang Zhang
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Changchun, China
| | - Jinming Han
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Changchun, China
| | - Caiyun Liu
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Changchun, China
| | - Yang Zhou
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Changchun, China
| | - Jie Zhu
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Changchun, China; Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Tao Jin
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
48
|
Calender A, Weichhart T, Valeyre D, Pacheco Y. Current Insights in Genetics of Sarcoidosis: Functional and Clinical Impacts. J Clin Med 2020; 9:E2633. [PMID: 32823753 PMCID: PMC7465171 DOI: 10.3390/jcm9082633] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/05/2020] [Accepted: 08/11/2020] [Indexed: 12/17/2022] Open
Abstract
Sarcoidosis is a complex disease that belongs to the vast group of autoinflammatory disorders, but the etiological mechanisms of which are not known. At the crosstalk of environmental, infectious, and genetic factors, sarcoidosis is a multifactorial disease that requires a multidisciplinary approach for which genetic research, in particular, next generation sequencing (NGS) tools, has made it possible to identify new pathways and propose mechanistic hypotheses. Codified treatments for the disease cannot always respond to the most progressive forms and the identification of new genetic and metabolic tracks is a challenge for the future management of the most severe patients. Here, we review the current knowledge regarding the genes identified by both genome wide association studies (GWAS) and whole exome sequencing (WES), as well the connection of these pathways with the current research on sarcoidosis immune-related disorders.
Collapse
Affiliation(s)
- Alain Calender
- Department of Molecular and Medical genetics, Hospices Civils de Lyon, University Hospital, 69500 Bron, France;
- CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory, University Claude Bernard Lyon 1, 69007 Lyon, France
| | - Thomas Weichhart
- Center for Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, 1090 Vienna, Austria;
| | - Dominique Valeyre
- INSERM UMR 1272, Department of Pulmonology, Avicenne Hospital, University Sorbonne Paris Nord, Saint Joseph Hospital, AP-HP, 75014 Paris, France;
| | - Yves Pacheco
- Department of Molecular and Medical genetics, Hospices Civils de Lyon, University Hospital, 69500 Bron, France;
- CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory, University Claude Bernard Lyon 1, 69007 Lyon, France
| |
Collapse
|
49
|
Interleukin 39: a new member of interleukin 12 family. Cent Eur J Immunol 2020; 45:214-217. [PMID: 33456334 PMCID: PMC7792434 DOI: 10.5114/ceji.2020.97911] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 12/07/2017] [Indexed: 12/13/2022] Open
Abstract
Interleukin (IL)-12 family member is a heterodimer glycoprotein, composed of two covalently linked subunits, α and β chains. The α subunit consists of IL-23p19, IL-27p28, and IL-12p35, and the β subunit includes IL-12p40 and Epstein-Barr virus-induced gene (Ebi3). IL-39 is a new heterodimeric IL-12 family member composed of IL-23p19 and Ebi3 subunits. IL-39 is secreted by lipopolysaccharide-stimulated B cells. Other immune cells, such as dendritic cells and macrophages, express IL-39 mRNA. In lupus-like mice, GL7+B cells and CD138+plasma cells are highly activated and widely expressed, promoting high expression of IL-39. IL-39 mediates inflammatory responses through binding to a heterodimer of IL-23R/gp130 receptor and activation of signal transducer and activator of transcription (STAT)1/STAT3 signal molecules. The serum levels of IL-39 were significantly increased in patients with acute coronary syndrome compared with patients with normal coronary arteries. This review discusses the biological characteristics, receptor, and signal pathway as well as biological activity of IL-39 and its potential role in inflammation and other diseases.
Collapse
|
50
|
Reich K, Papp KA, Armstrong AW, Wasfi Y, Li S, Shen YK, Randazzo B, Song M, Kimball AB. Safety of guselkumab in patients with moderate-to-severe psoriasis treated through 100 weeks: a pooled analysis from the randomized VOYAGE 1 and VOYAGE 2 studies. Br J Dermatol 2020; 180:1039-1049. [PMID: 30485400 DOI: 10.1111/bjd.17454] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Long-term evaluation is required to confirm the safety profile of newer biologic agents. OBJECTIVES To report on pooled safety data from the ongoing VOYAGE 1 (NCT02207231) and VOYAGE 2 (NCT02207244) trials through 100 weeks of follow-up. METHODS Patients were randomized to either guselkumab 100 mg at weeks 0 and 4 and every 8 weeks thereafter; placebo at weeks 0, 4, 12 followed by guselkumab 100 mg at weeks 16 and 20 and every 8 weeks thereafter; or adalimumab 80 mg at week 0, 40 mg at week 1, and 40 mg every 2 weeks thereafter. Patients who received adalimumab crossed over to guselkumab at week 52 (VOYAGE 1) and at/after week 28 based on clinical response (VOYAGE 2). Open-label extensions, in which all patients received guselkumab, started at week 52 (VOYAGE 1) and week 76 (VOYAGE 2). Rates of adverse events (AEs) per 100 patient-years (PYs) are presented through 100 weeks of follow-up. RESULTS Through week 52, observed rates for guselkumab- and adalimumab-treated patients, respectively, were 262·45 per 100 PYs and 328·28 per 100 PYs for AEs, 6·20 per 100 PYs and 7·77 per 100 PYs for serious AEs (SAEs), 1·22 per 100 PYs and 1·79 per 100 PYs for serious infections (SIs), 0·28 per 100 PYs and 0·40 per 100 PYs for malignancies other than nonmelanoma skin cancers (NMSCs), 0·56 per 100 PYs and 0·40 per 100 PYs for NMSCs, and 0·47 per 100 PYs and 0·40 per 100 PYs for major adverse cardiovascular events (MACEs). Rates among patients treated with guselkumab through week 52 and week 100, respectively, were 262·45 per 100 PYs and 210·41 per 100 PYs for AEs, 6·20 and 6·29 per 100 PYs, for SAEs, 1·22 per 100 PYs and 1·06 per 100 PYs for SIs, 0·28 per 100 PYs and 0·38 per 100 PYs for malignancies, 0·56 per 100 PYs and 0·39 per 100 PYs for NMSCs, and 0·47 per 100 PYs and 0·38 per 100 PYs for MACEs. Among patients treated with adalimumab, rates of AEs, SAEs, SIs, malignancies, NMSCs, and MACEs showed some variability before and after crossover to guselkumab, although no new safety signals were noted after crossover. CONCLUSIONS The safety profile for guselkumab remains favourable through 100 weeks of treatment in patients with moderate-to-severe psoriasis.
Collapse
Affiliation(s)
- K Reich
- Dermatologikum Berlin and SCIderm Research Institute, Hamburg, Germany
| | - K A Papp
- K Papp Clinical Research and Probity Research, Inc., Waterloo, Canada
| | - A W Armstrong
- University of Southern California, Los Angeles, CA, U.S.A
| | - Y Wasfi
- Janssen Research & Development, LLC, Spring House, PA, U.S.A
| | - S Li
- Janssen Research & Development, LLC, Spring House, PA, U.S.A
| | - Y K Shen
- Janssen Research & Development, LLC, Spring House, PA, U.S.A
| | - B Randazzo
- Janssen Research & Development, LLC, Spring House, PA, U.S.A
| | - M Song
- Janssen Research & Development, LLC, Spring House, PA, U.S.A
| | - A B Kimball
- Harvard Medical School and Beth Israel Deaconess Medical Center, Inc., Boston, MA, U.S.A
| |
Collapse
|