6
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      P53 is Subjected to Lipoteichoic Acid-Induced Phosphorylation in the Lungs

      letter

      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

          P53 is a transcription factor protecting the cells against malignancies via modulation of multifarious regulatory signaling cascades. Those activities may result to either cellular repair, or to the elimination of the irreversible damaged tissue components. Recent evidence suggest that this endothelium defender (P53) exerts strong anti-inflammatory activities in the lungs. 1 P53 protects the endothelium cells against the lipopolysaccharide (LPS)-induced endothelial hyperpermeability by reducing the generation of the reactive oxygen species, 2 by suppressing the inflammatory RhoA/MLC2 pathway, 3 and by inducing the repairing activities of the unfolded protein response in the lungs. 4 5 Lung endothelial barrier dysfunction is both a cause and a consequence of severe lung inflammatory disease, including the lethal acute respiratory distress syndrome (ARDS). 1 Indeed, P53 expression levels are crucial for the integrity of the lung microvasculature, since P53 reduction due to LPS-induced P53 phosphorylation or small interfering ribonucleic acid has been previously shown to be related to the collapse of the lung barrier function. 6 Lipoteichoic acid (LTA) contributes in ARDS. 7 Fig. 1A demonstrates by Western blotting in bovine pulmonary arterial endothelial cells purchased from Genlantis (PB30205) (San Diego, California, United States) that LTA induces the phosphorylation of P53 and suppresses its expression levels. The LTA from Staphylococcus aureus (L2515) was purchased from Sigma-Aldrich (St. Louis, Missouri, United States). The densitometric analysis performed with Image J software indicated that this toxin, which is a major constituent of the cell wall of Gram-positive bacteria, increases the expression of pP53 ser392 ( Fig. 1B ), pP53 ser46 ( Fig. 1C ), pP53 ser15 ( Fig. 1D ), and pP53 ser33 ( Fig. 1E ), and reduces P53 ( Fig. 1F ). Interestingly, Hsp90 inhibitors are anticancer agents, which have been shown to counteract the LPS-induced P53 degradation, and deliver protective effects in the inflamed lungs. 3 Although those compounds were initially developed to stochastically eliminate cancers, it now appears ( Fig. 1G ) that they do not affect the viability of human lung microvascular cells (HuLEC-5a) (CRL-3244), which were obtained from the American Type Culture Collection (Manassas, Virginia, United States). Details regarding cell cultures and Western blotting have been previously reported. 2 4 Fig. 1 ( A – F ) Western blot analysis of phosphorylated P53 (pP53 ser392 , pP53 ser46 , pP53 ser15 , pP53 ser33 ) and total P53 expression after treatment of bovine pulmonary artery endothelial cell (BPAEC) with either lipoteichoic acid (LTA) (10 µg/mL) or vehicle (VEH) (phosphate-buffered saline [PBS]) for 2, 4, and 6 hours. The blots shown are representative of three independent experiments. The signal intensity of the protein bands was analyzed by densitometry. Protein levels of phosphorylated P53 and P53 were normalized to P53 and β-actin, respectively. * p  < 0.05, ** p  < 0.01 vs. VEH. Means ± standard error of mean (SEM). ( G ) Effects of the Hsp90 inhibitor AUY-922 in the viability of HuLEC-5a. Cells were treated with either VEH (0.1% dimethyl sulfoxide [DMSO]) or AUY-922 (10 −3 , 10 −2 , 10 −1 , 1.0, 5.0, 10.0, 25.0, 50.0, 100 µM) for 24 hours. Cell viability was evaluated by employing the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. ** p  < 0.01 vs. VEH, n  = 3. Means ± SEM. Treatment of HuLEC-5a cells with moderate concentrations of AUY-922 (101756–820) ( Fig. 1G ) from VWR (Radnor, Pennsylvania, United States) did not affect the viability of those cells, as measured with the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Briefly, the cells were seeded in 96-well culture plates (10,000 cells/well) in complete growth media and were treated with AUY-922 (0–100 µM). After 24 hours, the media was replaced with fresh media containing 0.5 mg/mL MTT. After 3.5 hours of incubation, dimethyl sulfoxide (100 μL/well) was added to dissolve the MTT crystals, and 15 minutes later the absorbance was measured at 570 nm in a Synergy H1 Hybrid Multi-Mode Reader from Biotek (Winooski, Vermont, United States). In all cases, GraphPad Prism (version 5.01) was used to analyze the data, and the values are expressed as the mean ± standard error of mean. Values of p less than 0.05 were considered as an indication of statistical significance, and the number of experimental repeats is indicated by the letter n . In conclusion, the present letter aims to substantiate our hypothesis that P53 is a target of the “inflammatory storm”-induced ARDS. Thus, pharmacological induction of P53 due to treatments with Hsp90 inhibitors, or growth hormone releasing hormone antagonists 8 may deliver a promising approach against the severe lung inflammatory disease. 9

          Related collections

          Most cited references7

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

          Lung inflammation induced by lipoteichoic acid or lipopolysaccharide in humans.

          Recognition of pathogen-associated molecular patterns by Toll-like receptors (TLRs) is considered to be important for an appropriate immune response against pathogens that enter the lower airways. We studied the effects of two different TLR agonists relevant for respiratory infections in the human lung: lipoteichoic acid (LTA; TLR2 agonist, component of gram-positive bacteria) and lipopolysaccharide (LPS; TLR4-agonist, component of gram-negative bacteria). Fifteen healthy subjects were given LPS or LTA: by bronchoscope, sterile saline was instilled into a lung segment followed by instillation of LTA or LPS into the contralateral lung. After 6 hours, a bronchoalveolar lavage was performed and inflammatory parameters were determined. Isolated RNA from purified alveolar macrophages was analyzed by multiplex ligation-dependent probe amplification. In addition, spontaneous cytokine release by alveolar macrophages was measured. Marked differences were detected between LTA- and LPS-induced lung inflammation. Whereas both elicited neutrophil recruitment, only LPS instillation was associated with activation of neutrophils (CD11b surface expression, degranulation product levels) and consistent rises of chemo-/cytokine levels. Moreover, LPS but not LTA activated alveolar macrophages, as reflected by enhanced expression of 10 different mRNAs encoding proinflammatory mediators and increased spontaneous cytokine release upon incubation ex vivo. Remarkably, only LTA induced C5a release. This is the first study to report the in vivo effects of LTA in men and to compare inflammation induced by LTA and LPS in the human lung. Our data suggest that stimulation of TLR2 or TLR4 results in differential pulmonary inflammation, which may be of relevance for understanding pathogenic mechanisms at play during gram-positive and gram-negative respiratory tract infection.
            Bookmark
            • Record: found
            • Abstract: not found
            • Article: not found

            Hsp90 inhibitors induce the unfolded protein response in bovine and mice lung cells

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

              p53 protects against LPS-induced lung endothelial barrier dysfunction.

              New therapies toward heart and blood vessel disorders may emerge from the development of Hsp90 inhibitors. Several independent studies suggest potent anti-inflammatory activities of those agents in human tissues. The molecular mechanisms responsible for their protective effects in the vasculature remain unclear. The present study demonstrates that the transcription factor p53, an Hsp90 client protein, is crucial for the maintenance of vascular integrity, protects again LPS-induced endothelial barrier dysfunction, and is involved in the mediation of the anti-inflammatory activity of Hsp90 inhibitors in lung tissues. p53 silencing by siRNA decreased transendothelial resistance (a measure of endothelial barrier function). A similar effect was induced by the p53 inhibitor pifithrin, which also potentiated the LPS-induced hyperpermeability in human lung microvascular endothelial cells (HLMVEC). On the other hand, p53 induction by nutlin suppressed the LPS-induced vascular barrier dysfunction. LPS decreased p53 expression in lung tissues and that effect was blocked by pretreatment with Hsp90 inhibitors both in vivo and in vitro. Furthermore, the Hsp90 inhibitor 17-allyl-amino-demethoxy-geldanamycin suppressed the LPS-induced overexpression of the p53 negative regulator MDMX as well as p53 and MDM2 (another p53 negative regulator) phosphorylation in HLMVEC. Both negative p53 regulators were downregulated by LPS in vivo. Chemically induced p53 overexpression resulted in the suppression of LPS-induced RhoA activation and MLC2 phosphorylation, whereas p53 suppression caused the opposite effects. These observations reveal new mechanisms for the anti-inflammatory actions of Hsp90 inhibitors, i.e., the induction of the transcription factor p53, which in turn can orchestrate robust vascular anti-inflammatory responses both in vivo and in vitro.
                Bookmark

                Author and article information

                Journal
                TH Open
                TH Open
                10.1055/s-00033990
                TH Open: Companion Journal to Thrombosis and Haemostasis
                Georg Thieme Verlag KG (Stuttgart · New York )
                2567-3459
                2512-9465
                July 2020
                20 August 2020
                : 4
                : 3
                : e173-e174
                Affiliations
                [1 ]School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, United States
                Author notes
                Address for correspondence Nektarios Barabutis, MSc, PhD School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe 1800 Bienville Drive, Monroe, LA 71201United States barabutis@ 123456ulm.edu
                Author information
                http://orcid.org/0000-0001-8366-5099
                Article
                200036
                10.1055/s-0040-1714695
                7440968
                37319608-ec12-4c5f-b7ea-6937e819694a

                This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

                History
                : 02 June 2020
                : 25 June 2020
                Funding
                Funded by: Board of Regents Support Fund
                Award ID: LEQSF(2019-22)-RD-A-26
                Funding This research is supported by the R&D, Research Competitiveness Subprogram (RCS) of the Louisiana Board of Regents through the Board of Regents Support Fund (LEQSF(2019-22)-RD-A-26) (PI: N.B.).
                Categories
                Letter to the Editor

                Comments

                Comment on this article