2
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Potential therapeutic effects of interleukin-35 on the differentiation of naïve T cells into Helios +Foxp3 + Tregs in clinical and experimental acute respiratory distress syndrome

      research-article

      Read this article at

      ScienceOpenPublisherPMC
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Regulatory T lymphocytes are important targets for the treatment of acute respiratory distress syndrome (ARDS). IL-35 is a newly identified IL-12 cytokine family member that plays an important protective role in a variety of immune system diseases by regulating Treg cell differentiation; however, the role of IL-35 in the pathogenesis of ARDS is still unclear. Here, we found that IL-35 was significantly elevated in adult patients with ARDS compared to controls. Additionally, IL-35 was positively and significantly correlated with IL-6, IL-10 and the oxygenation index (PaO2/FiO2 ratio) but negatively correlated with TNF-α, IL-1β and APACHE II score during ARDS. Moreover, the proportion of Treg/CD4 + cells in the peripheral blood of ARDS patients and the expression of NF-κB in PMBCs were significantly higher than in healthy individuals. Recombinant IL-35 improved survival in a murine model of CLP-induced ARDS. Additionally, IL-35 administration decreased the inflammatory response, as reflected by lower levels of cytokines (including IL-2, TNF-α, IL-1β and IL-6) and less lung damage in CLP-induced ARDS. Furthermore, recombinant IL-35 reduced the apoptosis of lung tissue and the expression of NF-κB signalling in a CLP-induced ARDS model and increased the proportion of Treg cells in spleen and peripheral blood. In vitro experiments revealed that IL-35 can affect the phosphorylation of STAT5 during differentiation of naïve CD4 + T lymphocytes into Foxp3 +Helios + Tregs. Our findings suggest that IL-35 attenuates ARDS by promoting the differentiation of naïve CD4 + T cells into Foxp3 +Helios + Tregs, thereby providing a novel tool for anti-ARDS therapy.

          Related collections

          Most cited references82

          • Record: found
          • Abstract: found
          • Article: not found

          The non-canonical NF-κB pathway in immunity and inflammation

          Defects in the non-canonical pathway of NF-κB activation are associated with severe immune deficiencies, and aberrant activation of this pathway can cause autoimmune and inflammatory diseases. Here, the author investigates the activation, signalling mechanisms and the biological function of the non-canonical NF-κB pathway.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Regulatory T cells: mechanisms of differentiation and function.

            The immune system has evolved to mount an effective defense against pathogens and to minimize deleterious immune-mediated inflammation caused by commensal microorganisms, immune responses against self and environmental antigens, and metabolic inflammatory disorders. Regulatory T (Treg) cell-mediated suppression serves as a vital mechanism of negative regulation of immune-mediated inflammation and features prominently in autoimmune and autoinflammatory disorders, allergy, acute and chronic infections, cancer, and metabolic inflammation. The discovery that Foxp3 is the transcription factor that specifies the Treg cell lineage facilitated recent progress in understanding the biology of regulatory T cells. In this review, we discuss cellular and molecular mechanisms in the differentiation and function of these cells.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Reversing established sepsis with antagonists of endogenous high-mobility group box 1.

              Despite significant advances in intensive care therapy and antibiotics, severe sepsis accounts for 9% of all deaths in the United States annually. The pathological sequelae of sepsis are characterized by a systemic inflammatory response, but experimental therapeutics that target specific early inflammatory mediators [tumor necrosis factor (TNF) and IL-1beta] have not proven efficacious in the clinic. We recently identified high mobility group box 1 (HMGB1) as a late mediator of endotoxin-induced lethality that exhibits significantly delayed kinetics relative to TNF and IL-1beta. Here, we report that serum HMGB1 levels are increased significantly in a standardized model of murine sepsis, beginning 18 h after surgical induction of peritonitis. Specific inhibition of HMGB1 activity [with either anti-HMGB1 antibody (600 microg per mouse) or the DNA-binding A box (600 microg per mouse)] beginning as late as 24 h after surgical induction of peritonitis significantly increased survival (nonimmune IgG-treated controls = 28% vs. anti-HMGB1 antibody group = 72%, P < 0.03; GST control protein = 28% vs. A box = 68%, P < 0.03). Animals treated with either HMGB1 antagonist were protected against the development of organ injury, as evidenced by improved levels of serum creatinine and blood urea nitrogen. These observations demonstrate that specific inhibition of endogenous HMGB1 therapeutically reverses lethality of established sepsis indicating that HMGB1 inhibitors can be administered in a clinically relevant time frame.
                Bookmark

                Author and article information

                Journal
                Mol Immunol
                Mol Immunol
                Molecular Immunology
                Elsevier Ltd.
                0161-5890
                1872-9142
                16 January 2021
                April 2021
                16 January 2021
                : 132
                : 236-249
                Affiliations
                [a ]Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
                [b ]Department of Pathophysiology, Chongqing Medical University, Chongqing, China
                Author notes
                [* ]Corresponding author at: Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
                [** ]Corresponding author.
                [1]

                Both Chuan-jiang Wang and Ke Xie contributed equally to this work.

                Article
                S0161-5890(21)00009-2
                10.1016/j.molimm.2021.01.009
                8058740
                33494935
                8320c91c-c2fb-4dad-9c22-0405c164748e
                © 2021 Elsevier Ltd. All rights reserved.

                Since January 2020 Elsevier has created a COVID-19 resource centre with free information in English and Mandarin on the novel coronavirus COVID-19. The COVID-19 resource centre is hosted on Elsevier Connect, the company's public news and information website. Elsevier hereby grants permission to make all its COVID-19-related research that is available on the COVID-19 resource centre - including this research content - immediately available in PubMed Central and other publicly funded repositories, such as the WHO COVID database with rights for unrestricted research re-use and analyses in any form or by any means with acknowledgement of the original source. These permissions are granted for free by Elsevier for as long as the COVID-19 resource centre remains active.

                History
                : 8 June 2020
                : 2 January 2021
                : 5 January 2021
                Categories
                Article

                Immunology
                il-35,acute respiratory distress syndrome,cytokines,naïve cd4+ t lymphocytes,regulatory t cells,nf-κb

                Comments

                Comment on this article