27
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Hydrogen Sulfide Alleviates Lipopolysaccharide-Induced Diaphragm Dysfunction in Rats by Reducing Apoptosis and Inflammation through ROS/MAPK and TLR4/NF-κB Signaling Pathways

      Read this article at

      Bookmark
          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

          Diaphragm dysfunction is an important clinical problem worldwide. Hydrogen sulfide (H 2S) is involved in many physiological and pathological processes in mammals. However, the effect and mechanism of H 2S in diaphragm dysfunction have not been fully elucidated. In this study, we detected that the level of H 2S was decreased in lipopolysaccharide- (LPS-) treated L6 cells. Treatment with H 2S increased the proliferation and viability of LPS-treated L6 cells. We found that H 2S decreased reactive oxygen species- (ROS-) induced apoptosis through the mitogen-activated protein kinase (MAPK) signaling pathway in LPS-treated L6 cells. Administration of H 2S alleviated LPS-induced inflammation by mediating the toll-like receptor-4 (TLR-4)/nuclear factor-kappa B (NF- κB) signaling pathway in L6 cells. Furthermore, H 2S improved diaphragmatic function and structure through the reduction of inflammation and apoptosis in the diaphragm of septic rats. In conclusion, these findings indicate that H 2S ameliorates LPS-induced diaphragm dysfunction in rats by reducing apoptosis and inflammation through ROS/MAPK and TLR4/NF- κB signaling pathways. Novel slow-releasing H 2S donors can be designed and applied for the treatment of diaphragm dysfunction.

          Related collections

          Most cited references54

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

          Tumor-derived hydrogen sulfide, produced by cystathionine-β-synthase, stimulates bioenergetics, cell proliferation, and angiogenesis in colon cancer.

          The physiological functions of hydrogen sulfide (H2S) include vasorelaxation, stimulation of cellular bioenergetics, and promotion of angiogenesis. Analysis of human colon cancer biopsies and patient-matched normal margin mucosa revealed the selective up-regulation of the H2S-producing enzyme cystathionine-β-synthase (CBS) in colon cancer, resulting in an increased rate of H2S production. Similarly, colon cancer-derived epithelial cell lines (HCT116, HT-29, LoVo) exhibited selective CBS up-regulation and increased H2S production, compared with the nonmalignant colonic mucosa cells, NCM356. CBS localized to the cytosol, as well as the mitochondrial outer membrane. ShRNA-mediated silencing of CBS or its pharmacological inhibition with aminooxyacetic acid reduced HCT116 cell proliferation, migration, and invasion; reduced endothelial cell migration in tumor/endothelial cell cocultures; and suppressed mitochondrial function (oxygen consumption, ATP turnover, and respiratory reserve capacity), as well as glycolysis. Treatment of nude mice with aminooxyacetic acid attenuated the growth of patient-derived colon cancer xenografts and reduced tumor blood flow. Similarly, CBS silencing of the tumor cells decreased xenograft growth and suppressed neovessel density, suggesting a role for endogenous H2S in tumor angiogenesis. In contrast to CBS, silencing of cystathionine-γ-lyase (the expression of which was unchanged in colon cancer) did not affect tumor growth or bioenergetics. In conclusion, H2S produced from CBS serves to (i) maintain colon cancer cellular bioenergetics, thereby supporting tumor growth and proliferation, and (ii) promote angiogenesis and vasorelaxation, consequently providing the tumor with blood and nutritients. The current findings identify CBS-derived H2S as a tumor growth factor and anticancer drug target.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Hydrogen sulfide-based therapeutics: exploiting a unique but ubiquitous gasotransmitter.

            Hydrogen sulfide (H2S) has become recognized as an important signalling molecule throughout the body, contributing to many physiological and pathological processes. In recent years, improved methods for measuring H2S levels and the availability of a wider range of H2S donors and more selective inhibitors of H2S synthesis have helped to more accurately identify the many biological effects of this highly reactive gaseous mediator. Animal studies of several H2S-releasing drugs have demonstrated considerable promise for the safe treatment of a wide range of disorders. Several such drugs are now in clinical trials.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Gasotransmitters in cancer: from pathophysiology to experimental therapy.

              The three endogenous gaseous transmitters - nitric oxide (NO), carbon monoxide (CO) and hydrogen sulfide (H2S) - regulate a number of key biological functions. Emerging data have revealed several new mechanisms for each of these three gasotransmitters in tumour biology. It is now appreciated that they show bimodal pharmacological character in cancer, in that not only the inhibition of their biosynthesis but also elevation of their concentration beyond a certain threshold can exert anticancer effects. This Review discusses the role of each gasotransmitter in cancer and the effects of pharmacological agents - some of which are in early-stage clinical studies - that modulate the levels of each gasotransmitter. A clearer understanding of the pharmacological character of these three gases and the mechanisms underlying their biological effects is expected to guide further clinical translation.
                Bookmark

                Author and article information

                Journal
                Oxidative Medicine and Cellular Longevity
                Oxidative Medicine and Cellular Longevity
                Hindawi Limited
                1942-0900
                1942-0994
                2018
                2018
                : 2018
                : 1-15
                Affiliations
                [1 ]The First Affiliated Hospital of Henan University, Kaifeng, Henan 475001, China
                [2 ]School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan 475004, China
                [3 ]Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, Henan 475004, China
                [4 ]College of Pharmacy, Henan University, Kaifeng, Henan 475004, China
                [5 ]Huaihe Hospital of Henan University, Kaifeng, Henan 475000, China
                [6 ]Brain Research Laboratory, College of Life Sciences, Henan University, Kaifeng, Henan 475004, China
                [7 ]Department of Microbiology and Immunology, School of Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA
                [8 ]The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, China
                [9 ]Henan Provincial People’s Hospital Affiliated to Henan University, Zhengzhou, Henan 450003, China
                Article
                10.1155/2018/9647809
                29977458
                e9e42343-ad94-403b-b4f5-b729951c34e2
                © 2018

                http://creativecommons.org/licenses/by/4.0/

                History

                Comments

                Comment on this article