12
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Farnesoid X receptor is essential for the survival of renal medullary collecting duct cells under hypertonic stress.

      Read this article at

      ScienceOpenPublisherPubMed
          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

          Hypertonicity in renal medulla is critical for the kidney to produce concentrated urine. Renal medullary cells have to survive high medullary osmolarity during antidiuresis. Previous study reported that farnesoid X receptor (FXR), a nuclear receptor transcription factor activated by endogenous bile acids, increases urine concentrating ability by up-regulating aquaporin 2 expression in medullary collecting duct cells (MCDs). However, whether FXR is also involved in the maintenance of cell survival of MCDs under dehydration condition and hypertonic stress remains largely unknown. In the present study, we demonstrate that 24-hours water restriction selectively up-regulated renal medullary expression of FXR with little MCD apoptosis in wild-type mice. In contrast, water deprivation caused a massive apoptosis of MCDs in both global FXR gene-deficient mice and collecting duct-specific FXR knockout mice. In vitro studies showed that hypertonicity significantly increased FXR and tonicity response enhancer binding protein (TonEBP) expression in mIMCD3 cell line and primary cultured MCDs. Activation and overexpression of FXR markedly increased cell viability and decreased cell apoptosis under hyperosmotic conditions. In addition, FXR can increase gene expression and nuclear translocation of TonEBP. We conclude that FXR protects MCDs from hypertonicity-induced cell injury very likely via increasing TonEBP expression and nuclear translocation. This study provides insights into the molecular mechanism by which FXR enhances urine concentration via maintaining cell viability of MCDs under hyperosmotic condition.

          Related collections

          Most cited references20

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

          Targeted disruption of the nuclear receptor FXR/BAR impairs bile acid and lipid homeostasis.

          Mice lacking the nuclear bile acid receptor FXR/BAR developed normally and were outwardly identical to wild-type littermates. FXR/BAR null mice were distinguished from wild-type mice by elevated serum bile acid, cholesterol, and triglycerides, increased hepatic cholesterol and triglycerides, and a proatherogenic serum lipoprotein profile. FXR/BAR null mice also had reduced bile acid pools and reduced fecal bile acid excretion due to decreased expression of the major hepatic canalicular bile acid transport protein. Bile acid repression and induction of cholesterol 7alpha-hydroxylase and the ileal bile acid binding protein, respectively, did not occur in FXR/BAR null mice, establishing the regulatory role of FXR/BAR for the expression of these genes in vivo. These data demonstrate that FXR/BAR is critical for bile acid and lipid homeostasis by virtue of its role as an intracellular bile acid sensor.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Bile acids in glucose metabolism in health and disease

            Bile acids were recently shown to regulate glucose homeostasis through diverse mechanisms involving the host and its microbiome. Herein, Shapiro et al. discuss the impact of bile acids on normal and impaired glycemic responses, including potential therapeutic implications in treating hyperglycemia and diabetes.
              Bookmark
              • Record: found
              • Abstract: not found
              • Article: not found

              Thiazolidinediones expand body fluid volume through PPARgamma stimulation of ENaC-mediated renal salt absorption.

              Thiazolidinediones (TZDs) are widely used to treat type 2 diabetes mellitus; however, their use is complicated by systemic fluid retention. Along the nephron, the pharmacological target of TZDs, peroxisome proliferator-activated receptor-gamma (PPARgamma, encoded by Pparg), is most abundant in the collecting duct. Here we show that mice treated with TZDs experience early weight gain from increased total body water. Weight gain was blocked by the collecting duct-specific diuretic amiloride and was also prevented by deletion of Pparg from the collecting duct, using Pparg (flox/flox) mice. Deletion of collecting duct Pparg decreased renal Na(+) avidity and increased plasma aldosterone. Treating cultured collecting ducts with TZDs increased amiloride-sensitive Na(+) absorption and Scnn1g mRNA (encoding the epithelial Na(+) channel ENaCgamma) expression through a PPARgamma-dependent pathway. These studies identify Scnn1g as a PPARgamma target gene in the collecting duct. Activation of this pathway mediates fluid retention associated with TZDs, and suggests amiloride might provide a specific therapy.
                Bookmark

                Author and article information

                Journal
                Proc. Natl. Acad. Sci. U.S.A.
                Proceedings of the National Academy of Sciences of the United States of America
                Proceedings of the National Academy of Sciences
                1091-6490
                0027-8424
                May 22 2018
                : 115
                : 21
                Affiliations
                [1 ] Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044 Liaoning, China.
                [2 ] Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104.
                [3 ] Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Dalian Medical University, Dalian, 116044 Liaoning, China.
                [4 ] Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044 Liaoning, China; guanyf@dmu.edu.cn jgustafs@central.uh.edu zhangxy@dmu.edu.cn.
                [5 ] Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77204; guanyf@dmu.edu.cn jgustafs@central.uh.edu zhangxy@dmu.edu.cn.
                [6 ] Center for Biosciences, Department of Biosciences and Nutrition, Karolinska Institutet, Novum, 14186 Stockholm, Sweden.
                Article
                1803945115
                10.1073/pnas.1803945115
                29739889
                54d23f9a-245f-435d-909a-7d3d8a50dfd6
                History

                cell viability,hypertonicity,osmoprotection,NFAT5,bile acid receptor

                Comments

                Comment on this article