32
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      NFAT5 expression in bone marrow-derived cells enhances atherosclerosis and drives macrophage migration

      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

          Objective: We have previously shown that the transcription factor, nuclear factor of activated T-cells 5 (NFAT5), regulates vascular smooth muscle cell phenotypic modulation, but the role of NFAT5 in atherosclerosis is unknown. Our main objective was to determine if NFAT5 expression in bone marrow (BM)-derived cells altered atherosclerotic development and macrophage function. Methods and Results: NFAT5 +/−ApoE −/− mice were generated for in vivo atherosclerosis studies. Following high fat diet feeding, en face analysis of the thoracic aorta established that genome-wide NFAT5 haploinsufficiency reduced atherosclerotic lesion formation by 73%. BM transplant studies revealed that transplantation of NFAT5 +/−ApoE −/− marrow into NFAT5 +/+ApoE −/− mice resulted in a similar 86% reduction in lesion formation. In vitro functional analysis of BM-derived macrophages demonstrated that NFAT5 is required for macrophage migration, which is a key event in the propagation of atherosclerosis. Conclusion: We have identified NFAT5 in BM-derived cells as a positive regulator of atherosclerotic lesion formation and macrophage function in the vasculature.

          Related collections

          Most cited references14

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

          The role of NFAT transcription factors in integrin-mediated carcinoma invasion.

          Integrins, receptors for extracellular matrix ligands, are critical regulators of the invasive phenotype. Specifically, the alpha(6)beta(4) integrin has been linked with epithelial cell motility, cellular survival and carcinoma invasion, hallmarks of metastatic tumours. Previous studies have also shown that antagonists of the NFAT (nuclear factor of activated T-cells) family of transcription factors exhibit strong anti-tumour-promoting activity. This suggests that NFAT may function in tumour metastasis. Here, we investigate the involvement of NFAT in promoting carcinoma invasion downstream of the alpha(6)beta(4) integrin. We provide evidence that both NFAT1, and the recently identified NFAT5 isoform, are expressed in invasive human ductal breast carcinomas and participate in promoting carcinoma invasion using cell lines derived from human breast and colon carcinomas. NFAT1 and NFAT5 activity correlates with the expression of the alpha(6)beta(4) integrin. In addition, the transcriptional activity of NFAT5 is induced by alpha(6)beta(4) clustering in the presence of chemo-attractants, resulting in enhanced cell migration. These observations show that NFATs are targets of alpha(6)beta(4) integrin signalling and are involved in promoting carcinoma invasion, highlighting a novel function for this family of transcription factors in human cancer.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            NFAT5/TonEBP mutant mice define osmotic stress as a critical feature of the lymphoid microenvironment.

            Osmotic stress responses are critical not only to the survival of unicellular organisms but also to the normal function of the mammalian kidney. However, the extent to which cells outside the kidney rely on osmotic stress responses in vivo remains unknown. Nuclear factor of activated T cells 5 (NFAT5)/tonicity enhancer binding protein (TonEBP), the only known osmosensitive mammalian transcription factor, is expressed most abundantly in the thymus and is induced upon lymphocyte activation. Here we report that NFAT5/TonEBP is not only essential for normal cell proliferation under hyperosmotic conditions but also necessary for optimal adaptive immunity. Targeted deletion of exons 6 and 7 of the Nfat5 gene, which encode a critical region of the DNA-binding domain, gave rise to a complete loss of function in the homozygous state and a partial loss of function in the heterozygous state. Complete loss of function resulted in late gestational lethality. Furthermore, hypertonicity-induced NFAT5/TonEBP transcriptional activity and hsp70.1 promoter function were completely eliminated, and cell proliferation under hyperosmotic culture conditions was markedly impaired. Partial loss of NFAT5/TonEBP function resulted in lymphoid hypocellularity and impaired antigen-specific antibody responses in viable heterozygous animals. In addition, lymphocyte proliferation ex vivo was reduced under hypertonic, but not isotonic, culture conditions. Direct measurement of tissue osmolality further revealed lymphoid tissues to be hyperosmolar. These results indicate that lymphocyte-mediated immunity is contingent on adaptation to physiologic osmotic stress, thus providing insight into the lymphoid microenvironment and the importance of the NFAT5/TonEBP osmotic stress response pathway in vivo.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Tonicity-responsive enhancer binding protein, a rel-like protein that stimulates transcription in response to hypertonicity.

              Hypertonicity (most often present as high salinity) is stressful to the cells of virtually all organisms. Cells survive in a hypertonic environment by increasing the transcription of genes whose products catalyze cellular accumulation of compatible osmolytes. In mammals, the kidney medulla is normally hypertonic because of the urinary concentrating mechanism. Cellular accumulation of compatible osmolytes in the renal medulla is catalyzed by the sodium/myo-inositol cotransporter (SMIT), the sodium/chloride/betaine cotransporter, and aldose reductase (synthesis of sorbitol). The importance of compatible osmolytes is underscored by the necrotic injury of the renal medulla and subsequent renal failure that results from the inhibition of SMIT in vivo by administration of a specific inhibitor. Tonicity-responsive enhancers (TonE) play a key role in hypertonicity-induced transcriptional stimulation of SMIT, sodium/chloride/betaine cotransporter, and aldose reductase. We report the cDNA cloning of human TonE binding protein (TonEBP), a transcription factor that stimulates transcription through its binding to TonE sequences via a Rel-like DNA binding domain. Western blot and immunohistochemical analyses of cells cultured in hypertonic medium reveal that exposure to hypertonicity elicits slow activation of TonEBP, which is the result of an increase in TonEBP amount and translocation to the nucleus.
                Bookmark

                Author and article information

                Journal
                Front Physiol
                Front Physiol
                Front. Physio.
                Frontiers in Physiology
                Frontiers Media S.A.
                1664-042X
                03 July 2012
                03 August 2012
                2012
                : 3
                : 313
                Affiliations
                [1] 1simpleDepartment of Pharmacology, University of Virginia, Charlottesville VA, USA
                [2] 2simpleThe Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville VA, USA
                [3] 3simpleDepartment of Medicine, University of Maryland School of Medicine, Baltimore MD, USA
                [4] 4simpleUlsan National Institute of Science and Technology Ulsan, South Korea
                [5] 5simpleDepartment of Medicine, Cardiovascular Division, University of Virginia, Charlottesville VA, USA
                Author notes

                Edited by: John D. Imig, Medical College of Wisconsin, USA

                Reviewed by: Brenda Lilly, The Ohio State University, USA; Craig R. Lee, University of North Carolina at Chapel Hill, USA; Ahmed A. Elmarakby, Georgia Health Science University, USA

                *Correspondence: Julia A. Halterman, Robert M. Berne Cardiovascular Research Center, University of Virginia, 409 Lane Road, RM 6022, Charlottesville, VA 22908, USA. e-mail: juliahalterman@ 123456gmail.com

                This article was submitted to Frontiers in Vascular Physiology, a specialty of Frontiers in Physiology.

                Article
                10.3389/fphys.2012.00313
                3429083
                22934063
                a91e96cb-82dd-4a83-a36f-0837c720e46e
                Copyright © 2012 Halterman, Kwon, Leitinger and Wamhoff.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in other forums, provided the original authors and source are credited and subject to any copyright notices concerning any third-party graphics etc.

                History
                : 07 June 2012
                : 17 July 2012
                Page count
                Figures: 5, Tables: 0, Equations: 0, References: 20, Pages: 7, Words: 4223
                Categories
                Physiology
                Original Research Article

                Anatomy & Physiology
                tonebp,macrophage,bone marrow,migration,atherosclerosis
                Anatomy & Physiology
                tonebp, macrophage, bone marrow, migration, atherosclerosis

                Comments

                Comment on this article