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

      Sulforaphane exposure impairs contractility and mitochondrial function in three-dimensional engineered heart tissue

      research-article

      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

          Sulforaphane (SFN) is a phytochemical compound extracted from cruciferous plants, like broccoli or cauliflower. Its isothiocyanate group renders SFN reactive, thus allowing post-translational modification of cellular proteins to regulate their function with the potential for biological and therapeutic actions. SFN and stabilized variants recently received regulatory approval for clinical studies in humans for the treatment of neurological disorders and cancer. Potential unwanted side effects of SFN on heart function have not been investigated yet. The present study characterizes the impact of SFN on cardiomyocyte contractile function in cardiac preparations from neonatal rat, adult mouse and human induced-pluripotent stem cell-derived cardiomyocytes. This revealed a SFN-mediated negative inotropic effect, when administered either acutely or chronically, with an impairment of the Frank-Starling response to stretch activation. A direct effect of SFN on myofilament function was excluded in chemically permeabilized mouse trabeculae. However, SFN pretreatment increased lactate formation and enhanced the mitochondrial production of reactive oxygen species accompanied by a significant reduction in the mitochondrial membrane potential. Transmission electron microscopy revealed disturbed sarcomeric organization and inflated mitochondria with whorled membrane shape in response to SFN exposure. Interestingly, administration of the alternative energy source l-glutamine to the medium that bypasses the uptake route of pyruvate into the mitochondrial tricarboxylic acid cycle improved force development in SFN-treated EHTs, suggesting indeed mitochondrial dysfunction as a contributor of SFN-mediated contractile dysfunction. Taken together, the data from the present study suggest that SFN might impact negatively on cardiac contractility in patients with cardiovascular co-morbidities undergoing SFN supplementation therapy. Therefore, cardiac function should be monitored regularly to avoid the onset of cardiotoxic side effects.

          Graphical abstract

          Summary scheme of the functional effects of SFN exerted in cardiomyocytes: decline in force, elevation of diastolic tension and alteration of mitochondrial function and metabolism. MM: mitochondrial membrane potential; MPC: mitochondrial pyruvate carrier; mito: mitochondria; PC: pyruvate carboxylase; PDH: pyruvate dehydrogenase; Mitochondrium adapted from Servier Medical ART: SMART ( smart.servier.com).

          Highlights

          • Sulforaphane has negative inotropic effects and increases diastolic tension.

          • Sulforaphane exposure increases lactate levels and mitochondrial ROS production and reduces mitochondrial membrane potential.

          • l-glutamine supplementation rescues the sulforaphane-mediated reduction in force development.

          • Sulforaphane plasma levels and cardiac function should be monitored to avoid unwanted cardiac side effects in patients.

          Related collections

          Most cited references39

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

          The Warburg Effect: How Does it Benefit Cancer Cells?

          Cancer cells rewire their metabolism to promote growth, survival, proliferation, and long-term maintenance. The common feature of this altered metabolism is the increased glucose uptake and fermentation of glucose to lactate. This phenomenon is observed even in the presence of completely functioning mitochondria and, together, is known as the 'Warburg Effect'. The Warburg Effect has been documented for over 90 years and extensively studied over the past 10 years, with thousands of papers reporting to have established either its causes or its functions. Despite this intense interest, the function of the Warburg Effect remains unclear. Here, we analyze several proposed explanations for the function of Warburg Effect, emphasize their rationale, and discuss their controversies.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Energy metabolic phenotype of the cardiomyocyte during development, differentiation, and postnatal maturation.

            Dramatic maturational changes occur in cardiac energy metabolism during cardiac development, differentiation, and postnatal growth. These changes in energy metabolism have important impacts on the ability of the cardiomyocyte to proliferate during early cardiac development, as well as when cardiomyocytes terminally differentiate during later development. During early cardiac development, glycolysis is a major source of energy for proliferating cardiomyocytes. As cardiomyocytes mature and become terminally differentiated, mitochondrial oxidative capacity increases, with fatty acid beta-oxidation becoming a major source of energy for the heart. The increase in mitochondrial oxidative capacity seems to coincide with a decrease in the proliferative ability of the cardiomyocyte. The switch from glycolysis to mitochondrial oxidative metabolism during cardiac development includes both alterations in the transcriptional control and acute alterations in the control of each pathway. Interestingly, if a hypertrophic stress is placed on the adult heart, cardiac energy metabolism switches to a more fetal phenotype, which includes an increase in glycolysis and decrease in mitochondrial fatty acid beta-oxidation. In this article, we review the impact of alterations in energy substrate metabolism on cardiomyocyte proliferation, differentiation, and postnatal maturation.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              The Warburg effect: 80 years on

              Influential research by Warburg and Cori in the 1920s ignited interest in how cancer cells' energy generation is different from that of normal cells. They observed high glucose consumption and large amounts of lactate excretion from cancer cells compared with normal cells, which oxidised glucose using mitochondria. It was therefore assumed that cancer cells were generating energy using glycolysis rather than mitochondrial oxidative phosphorylation, and that the mitochondria were dysfunctional. Advances in research techniques since then have shown the mitochondria in cancer cells to be functional across a range of tumour types. However, different tumour populations have different bioenergetic alterations in order to meet their high energy requirement; the Warburg effect is not consistent across all cancer types. This review will discuss the metabolic reprogramming of cancer, possible explanations for the high glucose consumption in cancer cells observed by Warburg, and suggest key experimental practices we should consider when studying the metabolism of cancer.
                Bookmark

                Author and article information

                Contributors
                Journal
                Redox Biol
                Redox Biol
                Redox Biology
                Elsevier
                2213-2317
                31 March 2021
                May 2021
                31 March 2021
                : 41
                : 101951
                Affiliations
                [a ]Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
                [b ]DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
                [c ]Institute of Experimental Pharmacology and Toxicology, University of Würzburg, Versbacher Str., 9 97078, Würzburg, Germany
                [d ]Department of Morphology and Electron Microscopy, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
                [e ]Functional Proteomics, Faculty of Medicine, Goethe University Frankfurt, 60590, Frankfurt, Germany
                [f ]Leibniz-Institut für Analytische Wissenschaften − ISAS e.V., Bunsen-Kirchhoff-Str. 11, 44139, Dortmund, Germany
                Author notes
                []Corresponding author. Institute of Experimental Pharmacology and Toxicology; University Medical Center Hamburg-Eppendorf, Martinistrasse 52; 20246, Hamburg, Germany. f.cuello@ 123456uke.de
                [∗∗ ]Corresponding author. Institute of Experimental Pharmacology and Toxicology; University Medical Center Hamburg-Eppendorf, Martinistrasse 52; 20246, Hamburg, Germany. dralexandra.rhoden@ 123456gmail.com
                Article
                S2213-2317(21)00099-9 101951
                10.1016/j.redox.2021.101951
                8056268
                33831709
                03fb5911-bdfd-4336-93d2-c3c222b2ef39
                © 2021 The Author(s)

                This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

                History
                : 18 February 2021
                : 16 March 2021
                : 16 March 2021
                Categories
                Research Paper

                sulforaphane,engineered heart tissue,contractile and mitochondrial function

                Comments

                Comment on this article