1
|
Mistry HD, Klossner R, Scaife PJ, Eisele N, Kurlak LO, Kallol S, Albrecht C, Gennari-Moser C, Briggs LV, Broughton Pipkin F, Mohaupt MG. Alterations of Placental Sodium in Preeclampsia: Trophoblast Responses. Hypertension 2024; 81:1924-1934. [PMID: 38966986 PMCID: PMC11319085 DOI: 10.1161/hypertensionaha.124.23001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 06/19/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND Evidence suggests that increasing salt intake in pregnancy lowers blood pressure, protecting against preeclampsia. We hypothesized that sodium (Na+) evokes beneficial placental signals that are disrupted in preeclampsia. METHODS Blood and urine were collected from nonpregnant women of reproductive age (n=26) and pregnant women with (n=50) and without (n=55) preeclampsia, along with placental biopsies. Human trophoblast cell lines and primary human trophoblasts were cultured with varying Na+ concentrations. RESULTS Women with preeclampsia had reduced placental and urinary Na+ concentrations, yet increased urinary angiotensinogen and reduced active renin, aldosterone concentrations, and osmotic response signal TonEBP (tonicity-responsive enhancer binding protein) expression. In trophoblast cell cultures, TonEBP was consistently increased upon augmented Na+ exposure. Mechanistically, inhibiting Na+/K+-ATPase or adding mannitol evoked the TonEBP response, whereas inhibition of cytoskeletal signaling abolished it. CONCLUSIONS Enhanced Na+ availability induced osmotic gradient-dependent cytoskeletal signals in trophoblasts, resulting in proangiogenic responses. As placental salt availability is compromised in preeclampsia, adverse systemic responses are thus conceivable.
Collapse
Affiliation(s)
- Hiten D. Mistry
- Department of Women and Children’s Health, School of Life Course and Population Health Sciences, King’s College London, United Kingdom (H.D.M.)
| | - Rahel Klossner
- Teaching Hospital Internal Medicine, Lindenhofgruppe, Switzerland (R.K., M.G.M.)
- Department of Nephrology and Hypertension (R.K., N.E., C.G.-M., M.G.M.), University of Bern, Switzerland
- Department for BioMedical Research (R.K., N.E., C.G.-M., M.G.M.), University of Bern, Switzerland
| | - Paula J. Scaife
- Clinical, Metabolic and Molecular Physiology (P.J.S.), University of Nottingham, United Kingdom
| | - Nicole Eisele
- Department of Nephrology and Hypertension (R.K., N.E., C.G.-M., M.G.M.), University of Bern, Switzerland
- Department for BioMedical Research (R.K., N.E., C.G.-M., M.G.M.), University of Bern, Switzerland
| | - Lesia O. Kurlak
- Stroke Trials Unit (School of Medicine) (L.O.K.), University of Nottingham, United Kingdom
| | - Sampada Kallol
- Institute for Biochemistry and Molecular Medicine, University of Bern, Switzerland (S.K., C.A.)
| | - Christiane Albrecht
- Institute for Biochemistry and Molecular Medicine, University of Bern, Switzerland (S.K., C.A.)
| | - Carine Gennari-Moser
- Department of Nephrology and Hypertension (R.K., N.E., C.G.-M., M.G.M.), University of Bern, Switzerland
- Department for BioMedical Research (R.K., N.E., C.G.-M., M.G.M.), University of Bern, Switzerland
| | - Louise V. Briggs
- Advanced Material Research Group, Faculty of Engineering (L.V.B.), University of Nottingham, United Kingdom
| | | | - Markus G. Mohaupt
- Teaching Hospital Internal Medicine, Lindenhofgruppe, Switzerland (R.K., M.G.M.)
- Department of Nephrology and Hypertension (R.K., N.E., C.G.-M., M.G.M.), University of Bern, Switzerland
- Department for BioMedical Research (R.K., N.E., C.G.-M., M.G.M.), University of Bern, Switzerland
| |
Collapse
|
2
|
Davis KC, Raizen DM. A mechanism for sickness sleep: lessons from invertebrates. J Physiol 2017; 595:5415-5424. [PMID: 28028818 DOI: 10.1113/jp273009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Accepted: 12/16/2016] [Indexed: 11/08/2022] Open
Abstract
During health, animal sleep is regulated by an internal clock and by the duration of prior wakefulness. During sickness, sleep is regulated by cytokines released from either peripheral cells or from cells within the nervous system. These cytokines regulate central nervous system neurons to induce sleep. Recent research in the invertebrates Caenorhabditis elegans and Drosophila melanogaster has led to new insights into the mechanism of sleep during sickness. Sickness is triggered by exposure to environments such as infection, heat, or ultraviolet light irradiation, all of which cause cellular stress. Epidermal growth factor is released from stressed cells and signals to activate central neuroendocrine cell(s). These neuron(s) release neuropeptides including those containing an amidated arginine(R)-phenylalanine(F) motif at their C-termini (RFamide peptides). Importantly, mechanisms regulating sickness sleep are partially distinct from those regulating healthy sleep. We will here review key findings that have elucidated the central neuroendocrine mechanism of sleep during sickness. Adaptive mechanisms employed in the control of sickness sleep may play a role in correcting cellular homeostasis after various insults. We speculate that these mechanisms may play a maladaptive role in human pathological conditions such as in the fatigue and anorexia associated with autoimmune diseases, with major depression, and with unexplained chronic fatigue.
Collapse
Affiliation(s)
- Kristen C Davis
- Department of Neurology, Centre for Sleep and Neurobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - David M Raizen
- Department of Neurology, Centre for Sleep and Neurobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
3
|
Protective role of fructokinase blockade in the pathogenesis of acute kidney injury in mice. Nat Commun 2017; 8:14181. [PMID: 28194018 PMCID: PMC5316807 DOI: 10.1038/ncomms14181] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 12/07/2016] [Indexed: 01/10/2023] Open
Abstract
Acute kidney injury is associated with high mortality, especially in intensive care unit patients. The polyol pathway is a metabolic route able to convert glucose into fructose. Here we show the detrimental role of endogenous fructose production by the polyol pathway and its metabolism through fructokinase in the pathogenesis of ischaemic acute kidney injury (iAKI). Consistent with elevated urinary fructose in AKI patients, mice undergoing iAKI show significant polyol pathway activation in the kidney cortex characterized by high levels of aldose reductase, sorbitol and endogenous fructose. Wild type but not fructokinase knockout animals demonstrate severe kidney injury associated with ATP depletion, elevated uric acid, oxidative stress and inflammation. Interestingly, both the renal injury and dysfunction in wild-type mice undergoing iAKI is significantly ameliorated when exposed to luteolin, a recently discovered fructokinase inhibitor. This study demonstrates a role for fructokinase and endogenous fructose as mediators of acute renal disease. The polyol pathway, which converts glucose into sorbitol and fructose, is active in chronic conditions like hepatic steatosis and chronic kidney disease. Here, Andres-Hernando et al. show that fructose production promotes renal injury and fructokinase inhibition protects against kidney damage during ischaemic acute kidney disease.
Collapse
|
4
|
Lai KP, Li JW, Gu J, Chan TF, Tse WKF, Wong CKC. Transcriptomic analysis reveals specific osmoregulatory adaptive responses in gill mitochondria-rich cells and pavement cells of the Japanese eel. BMC Genomics 2015; 16:1072. [PMID: 26678671 PMCID: PMC4683740 DOI: 10.1186/s12864-015-2271-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 12/03/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Homeostasis of ions and water is important for the maintenance of cellular functions. The regulation of the homeostasis is particularly important in euryhaline fish that migrate between freshwater (FW) and seawater (SW) environments. The fish gill, the major tissue that forms an interface separating the extracellular fluids and external water environment, has an effective transport system to maintain and regulate a constant body osmolality. In fish gills, the two major epithelial cells, pavement cells (PVCs) and mitochondria-rich cells (MRCs), are known to play key and complementary roles in ion transport at the interface. Discovering the robust mechanisms underlying the two cell types' response to osmotic stress would benefit our understanding of the fundamental mechanism allowing PVCs and MRCs to handle osmotic stress. Owing to the limited genomic data available on estuarine species, existing knowledge in this area is slim. In this study, transcriptome analyses were conducted using PVCs and MRCs isolated from Japanese eels adapted to FW or SW environments to provide a genome-wide molecular study to unravel the fundamental processes at work. RESULTS The study identified more than 12,000 transcripts in the gill cells. Interestingly, remarkable differential expressed genes (DEGs) were identified in PVCs (970 transcripts) instead of MRCs (400 transcripts) in gills of fish adapted to FW or SW. Since PVCs cover more than 90 % of the gill epithelial surface, the greater change in gene expression patterns in PVCs in response to external osmolality is anticipated. In the integrity pathway analysis, 19 common biological functions were identified in PVCs and MRCs. In the enriched signaling pathways analysis, most pathways differed between PVCs and MRCs; 14 enriched pathways were identified in PVCs and 12 in MRCs. The results suggest that the osmoregulatory responses in PVCs and MRCs are cell-type specific, which supports the complementary functions of the cells in osmoregulation. CONCLUSIONS This is the first study to provide transcriptomic analysis of PVCs and MRCs in gills of eels adapted to FW or SW environments. It describes the cell-type specific transcriptomic network in different tonicity. The findings consolidate the known osmoregulatory pathways and provide molecular insight in osmoregulation. The presented data will be useful for researchers to select their targets for further studies.
Collapse
Affiliation(s)
- Keng Po Lai
- School of Biological Sciences, Kadoorie Biological Sciences Building, The University of Hong Kong, Pokfulam Road, Pok Fu Lam, Hong Kong
| | - Jing-Woei Li
- State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, Hong Kong.,Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Je Gu
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Ting-Fung Chan
- State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - William Ka Fai Tse
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Hong Kong.
| | - Chris Kong Chu Wong
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Hong Kong. .,Croucher Institute for Environmental Sciences, Hong Kong Baptist University, Pok Fu Lam, Hong Kong.
| |
Collapse
|
5
|
Kitterer D, Latus J, Ulmer C, Fritz P, Biegger D, Ott G, Alscher MD, Witowski J, Kawka E, Jörres A, Seeger H, Segerer S, Braun N. Activation of nuclear factor of activated T cells 5 in the peritoneal membrane of uremic patients. Am J Physiol Renal Physiol 2015; 308:F1247-58. [DOI: 10.1152/ajprenal.00617.2014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 03/25/2015] [Indexed: 11/22/2022] Open
Abstract
Peritoneal inflammation and fibrosis are responses to the uremic milieu and exposure to hyperosmolar dialysis fluids in patients on peritoneal dialysis. Cells respond to high osmolarity via the transcription factor nuclear factor of activated T cells (NFAT5). In the present study, the response of human peritoneal fibroblasts to glucose was analyzed in vitro. Expression levels of NFAT5 and chemokine (C-C motif) ligand (CCL2) mRNA were quantified in peritoneal biopsies of five nonuremic control patients, five uremic patients before PD (pPD), and eight patients on PD (oPD) using real-time PCR. Biopsies from 5 control patients, 25 pPD patients, and 25 oPD patients were investigated using immunohistochemistry to detect the expression of NFAT5, CCL2, NF-κB p50, NF-κB p65, and CD68. High glucose concentrations led to an early, dose-dependent induction of NFAT5 mRNA in human peritoneal fibroblasts. CCL2 mRNA expression was upregulated by high concentrations of glucose after 6 h, but, most notably, a concentration-dependent induction of CCL2 was present after 96 h. In human peritoneal biopsies, NFAT5 mRNA levels were increased in uremic patients compared with nonuremic control patients. No significant difference was found between the pPD group and oPD group. CCL2 mRNA expression was higher in the oPD group. Immunohistochemistry analysis was consistent with the results of mRNA analysis. CD68-positive cells were significantly increased in the oPD group. In conclusion, uremia results in NFAT5 induction, which might promote early changes of the peritoneum. Upregulation of NFAT5 in PD patients is associated with NFκB induction, potentially resulting in the recruitment of macrophages.
Collapse
Affiliation(s)
- Daniel Kitterer
- Division of Nephrology, Department of Internal Medicine, Robert-Bosch-Hospital, Stuttgart, Germany
| | - Joerg Latus
- Division of Nephrology, Department of Internal Medicine, Robert-Bosch-Hospital, Stuttgart, Germany
| | - Christoph Ulmer
- Department of General, Visceral, and Trauma Surgery, Robert-Bosch-Hospital, Stuttgart, Germany
| | - Peter Fritz
- Department of Diagnostic Medicine, Division of Pathology, Robert-Bosch Hospital, Stuttgart, Germany
| | - Dagmar Biegger
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, University of Tuebingen, Stuttgart, Germany
| | - German Ott
- Department of Diagnostic Medicine, Division of Pathology, Robert-Bosch Hospital, Stuttgart, Germany
| | - M. Dominik Alscher
- Division of Nephrology, Department of Internal Medicine, Robert-Bosch-Hospital, Stuttgart, Germany
| | - Janusz Witowski
- Department of Pathophysiology, University of Medical Sciences, Poznan, Poland
| | - Edyta Kawka
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Achim Jörres
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Harald Seeger
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland; and
- Institute of Physiology and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Stephan Segerer
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland; and
- Institute of Physiology and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Niko Braun
- Division of Nephrology, Department of Internal Medicine, Robert-Bosch-Hospital, Stuttgart, Germany
| |
Collapse
|
6
|
Küper C, Beck FX, Neuhofer W. NFAT5-mediated expression of S100A4 contributes to proliferation and migration of renal carcinoma cells. Front Physiol 2014; 5:293. [PMID: 25152734 PMCID: PMC4126233 DOI: 10.3389/fphys.2014.00293] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 07/21/2014] [Indexed: 12/13/2022] Open
Abstract
The osmosensitive transcription factor nuclear factor of activated T-cells (NFAT) 5, also known as tonicity enhancer binding protein (TonEBP), has been associated with the development of a variety of tumor entities, among them breast cancer, colon carcinoma, and melanoma. The aim of the present study was to determine whether NFAT5 is also involved in the development of renal cell carcinoma (RCC). The most common type of RCC, the clear cell RCC, originates from the proximal convoluted tubule. We tested our hypothesis in the clear cell RCC cell line CaKi-1 and the non-cancerous proximal tubule cell line HK-2, as control. Basal expression of NFAT5 and NFAT5 activity in CaKi-1 cells was several times higher than in HK-2 cells. Osmotic stress induced an increased NFAT5 activity in both CaKi-1 and HK-2 cells, again with significantly higher activities in CaKi-1 cells. Analysis of NFAT5-regulating signaling pathways in CaKi-1 cells revealed that inhibition of the MAP kinases p38, c-Jun-terminal kinase (JNK) and extracellular regulated kinase (ERK) and of the focal adhesion kinase (FAK) partially blunted NFAT5 activity. FAK and ERK were both constitutively active, even under isotonic conditions, which may contribute to the high basal expression and activity of NFAT5 in CaKi-1 cells. In contrast, the MAP kinases p38 and JNK were inactive under isotonic conditions and became activated under osmotic stress conditions, indicating that p38 and JNK mediate upregulation of NFAT5 activity under these conditions. siRNA-mediated knockdown of NFAT5 in CaKi-1 cells reduced the expression of S100A4, a member of the S100 family of proteins, which promotes metastasis. Knockdown of NFAT5 was accompanied by a significant decrease in proliferation and migration activity. Taken together, our results indicate that NFAT5 induces S100A4 expression in CaKi-1 cells, thereby playing an important role in RCC proliferation and migration.
Collapse
Affiliation(s)
- Christoph Küper
- Department of Physiology, University of Munich Munich, Germany
| | | | - Wolfgang Neuhofer
- Department of Internal Medicine, Medical Faculty of Mannheim, Medical Clinic V, University of Heidelberg Mannheim, Germany
| |
Collapse
|
7
|
Fernández-Martínez AB, Lucio Cazaña FJ. Prostaglandin E2 induces retinoic acid receptor-β up-regulation through MSK1. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:1997-2004. [PMID: 24953041 DOI: 10.1016/j.bbamcr.2014.05.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 05/05/2014] [Accepted: 05/23/2014] [Indexed: 01/30/2023]
Abstract
The pharmacological modulation of putative renoprotective factors hypoxia-inducible factor-1α (HIF-1α) and HIF-1α-regulated vascular endothelial growth factor-A (VEGF-A) in the kidney has therapeutic interest. In human renal proximal tubular HK2 cells, prostaglandin E2 (PGE2) up-regulates HIF-1α and VEGF-A through epidermal growth factor receptor (EGFR)-dependent up-regulation of retinoic acid receptor-β (RARβ). Here we studied the role of mitogen-activated protein kinases (MAPKs) ERK1/2 and p38 and their target kinase, mitogen- and stress activated kinase-1 (MSK1), in the signaling cascade. Treatment of HK2 cells with PGE2 resulted in increased phosphorylation of EGFR, the three studied kinases and the histone H3 (Ser10) at the RARβ gene promoter (the latter has been proposed as a molecular signature of the activated RARβ gene promoter). Prevention of the phosphorylation of EGFR, ERK1/2, p38 MAPK or MSK1 is by incubating, respectively, with AG1478, PD98059, SB203580 or H89 allowed to elucidate the precise phosphorylation order in the signaling cascade triggered by PGE2: first, EGFR; then, ERK1/2 and p38 MAPK and, finally, MSK1. Phosphorylation of MSK1 led to that of Ser10 in histone H3 and to activation of RARβ gene transcription (and the consequent increase in the expression of HIF-1α and VEGF-A), which was suppressed by H89 or by transfecting cells with a vector encoding for a dominant-negative mutant of MSK1. These results highlight the relevance of MSK1 in the up-regulation of RARβ by PGE2. They also may contribute to new therapeutic approaches based upon the pharmacological control of HIF-1α/VEGF-A in the proximal tubule through the modulation of the PGE2/EGFR/MAPK/MSK1/RARβ pathway.
Collapse
|
8
|
Jantsch J, Binger KJ, Müller DN, Titze J. Macrophages in homeostatic immune function. Front Physiol 2014; 5:146. [PMID: 24847274 PMCID: PMC4017126 DOI: 10.3389/fphys.2014.00146] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 03/27/2014] [Indexed: 01/20/2023] Open
Abstract
Macrophages are not only involved in inflammatory and anti-infective processes, but also play an important role in maintaining tissue homeostasis. In this review, we summarize recent evidence investigating the role of macrophages in controlling angiogenesis, metabolism as well as salt and water balance. Particularly, we summarize the importance of macrophage tonicity enhancer binding protein (TonEBP, also termed nuclear factor of activated T-cells 5 [NFAT5]) expression in the regulation of salt and water homeostasis. Further understanding of homeostatic macrophage function may lead to new therapeutic approaches to treat ischemia, hypertension and metabolic disorders.
Collapse
Affiliation(s)
- Jonathan Jantsch
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität Erlangen-Nürnberg Erlangen, Germany
| | - Katrina J Binger
- Experimental and Clinical Research Center (ECRC), Max-Delbrück Center for Molecular Medicine, Charité Medical Faculty Berlin, Germany
| | - Dominik N Müller
- Experimental and Clinical Research Center (ECRC), Max-Delbrück Center for Molecular Medicine, Charité Medical Faculty Berlin, Germany
| | - Jens Titze
- Interdisciplinary Center for Clinical Research and Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg Erlangen, Germany ; Divison of Clinical Pharmacology, Vanderbilt University School of Medicine Nashville, TN, USA
| |
Collapse
|
9
|
Cheung CY, Ko BC. NFAT5 in cellular adaptation to hypertonic stress - regulations and functional significance. J Mol Signal 2013; 8:5. [PMID: 23618372 PMCID: PMC3655004 DOI: 10.1186/1750-2187-8-5] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 04/11/2013] [Indexed: 12/22/2022] Open
Abstract
The Nuclear Factor of Activated T Cells-5 (NFAT5), also known as OREBP or TonEBP, is a member of the nuclear factors of the activated T cells family of transcription factors. It is also the only known tonicity-regulated transcription factor in mammals. NFAT5 was initially known for its role in the hypertonic kidney inner medulla for orchestrating a genetic program to restore the cellular homeostasis. Emerging evidence, however, suggests that NFAT5 might play a more diverse functional role, including a pivotal role in blood pressure regulation and the development of autoimmune diseases. Despite the growing significance of NFAT5 in physiology and diseases, our understanding of how its activity is regulated remains very limited. Furthermore, how changes in tonicities are converted into functional outputs via NFAT5 remains elusive. Therefore, this review aims to summarize our current knowledge on the functional roles of NFAT5 in osmotic stress adaptation and the signaling pathways that regulate its activity.
Collapse
Affiliation(s)
- Chris Yk Cheung
- Department of Anatomical and Cellular Pathology, and The State Key Laboratory in Oncology in South China, The Chinese University of Hong Kong, The Prince of Wales Hospital, Rm 38019, Clinical Sciences Building, Shatin, Hong Kong, China.
| | | |
Collapse
|
10
|
The role of osmotic polysorbitol-based transporter in RNAi silencing via caveolae-mediated endocytosis and COX-2 expression. Biomaterials 2012; 33:8868-80. [DOI: 10.1016/j.biomaterials.2012.08.049] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 08/22/2012] [Indexed: 12/11/2022]
|
11
|
NFAT5 is activated by hypoxia: role in ischemia and reperfusion in the rat kidney. PLoS One 2012; 7:e39665. [PMID: 22768306 PMCID: PMC3388090 DOI: 10.1371/journal.pone.0039665] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Accepted: 05/27/2012] [Indexed: 12/13/2022] Open
Abstract
The current hypothesis postulates that NFAT5 activation in the kidney's inner medulla is due to hypertonicity, resulting in cell protection. Additionally, the renal medulla is hypoxic (10–18 mmHg); however there is no information about the effect of hypoxia on NFAT5. Using in vivo and in vitro models, we evaluated the effect of reducing the partial pressure of oxygen (PO2) on NFAT5 activity. We found that 1) Anoxia increased NFAT5 expression and nuclear translocation in primary cultures of IMCD cells from rat kidney. 2) Anoxia increased transcriptional activity and nuclear translocation of NFAT5 in HEK293 cells. 3) The dose-response curve demonstrated that HIF-1α peaked at 2.5% and NFAT5 at 1% of O2. 4) At 2.5% of O2, the time-course curve of hypoxia demonstrated earlier induction of HIF-1α gene expression than NFAT5. 5) siRNA knockdown of NFAT5 increased the hypoxia-induced cell death. 6) siRNA knockdown of HIF-1α did not affect the NFAT5 induction by hypoxia. Additionally, HIF-1α was still induced by hypoxia even when NFAT5 was knocked down. 7) NFAT5 and HIF-1α expression were increased in kidney (cortex and medulla) from rats subjected to an experimental model of ischemia and reperfusion (I/R). 7) Experimental I/R increased the NFAT5-target gene aldose reductase (AR). 8) NFAT5 activators (ATM and PI3K) were induced in vitro (HEK293 cells) and in vivo (I/R kidneys) with the same timing of NFAT5. 8) Wortmannin, which inhibits ATM and PI3K, reduces hypoxia-induced NFAT5 transcriptional activation in HEK293 cells. These results demonstrate for the first time that NFAT5 is induced by hypoxia and could be a protective factor against ischemic damage.
Collapse
|
12
|
Zhuang S, Duan M, Yan Y. Src family kinases regulate renal epithelial dedifferentiation through activation of EGFR/PI3K signaling. J Cell Physiol 2012; 227:2138-44. [PMID: 21780115 DOI: 10.1002/jcp.22946] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Dedifferentiation, a process by which differentiated cells become mesenchymal-like proliferating cells, is the first step in renal epithelium repair and occurs in vivo after acute kidney injury and in vitro in primary culture. However, the underlying mechanism remains poorly understood. In this report, we studied the signaling events that mediate dedifferentiation of proximal renal tubular cells (RPTC) in primary culture. RPTC dedifferentiation characterized by increased expression of vimentin concurrent with decreased expression of cytokeratin-18 was observed at 24 h after the initial plating of freshly isolated proximal tubules and persisted for 72 h. At 96 h, RPTC started to redifferentiate as revealed by reciprocal expression of cytokeratin-18 and vimentin and completed at 120 h. Phosphorylation levels of Src, epidermal growth factor receptor (EGFR), AKT (a target of phosphoinositide-3-kinase (PI3K)), and ERK1/2 were increased in the early time course of culture (<72 h). Inhibition of Src family kinases (SFKs) with PP1 blocked EGFR, AKT, and ERK1/2 phosphorylation, as well as RPTC dedifferentiation. Inhibition of EGFR with AG1478 also blocked AKT and ERK1/2 phosphorylation and RPTC dedifferentiation. Although inactivation of the PI3K/AKT pathway with LY294002 inhibited RPTC dedifferentiation, blocking the ERK1/2 pathway with U0126 did not show such an effect. Moreover, inhibition of SFKs, EGFR, PI3K/AKT, but not ERK1/2 pathways abrogated RPTC outgrowth and SFK inhibition decreased RPTC proliferation and migration. These findings demonstrate a critical role of SFKs in mediating RPTC dedifferentiation through activation of the EGFR/PI3K signaling pathway.
Collapse
Affiliation(s)
- Shougang Zhuang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.
| | | | | |
Collapse
|
13
|
NFAT5 contributes to osmolality-induced MCP-1 expression in mesothelial cells. Mediators Inflamm 2012; 2012:513015. [PMID: 22619484 PMCID: PMC3350971 DOI: 10.1155/2012/513015] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 01/28/2012] [Indexed: 02/03/2023] Open
Abstract
Increased expression of the C-C chemokine monocyte chemoattractant protein-1 (MCP-1) in mesothelial cells in response to high glucose concentrations and/or high osmolality plays a crucial role in the development of peritoneal fibrosis during continuous ambulatory peritoneal dialysis (CAPD). Recent studies suggest that in kidney cells osmolality-induced MCP-1 upregulation is mediated by the osmosensitive transcription factor, nuclear factor of activated T cells 5 (NFAT5). The present study addressed the question of whether activation of NFAT5 by hyperosmolality, as present in PD fluids, contributes to MCP-1 expression in the mesothelial cell line Met5A. Hyperosmolality, induced by addition of glucose, NaCl, or mannitol to the growth medium, increased NFAT5 activity and stimulated MCP-1 expression in Met5A cells. siRNA-mediated knockdown of NFAT5 attenuated osmolality-induced MCP-1 upregulation substantially. Hyperosmolality also induced activation of nuclear factor-κB (NF-κB). Accordingly, pharmacological inhibition of NF-κB significantly decreased osmolality-induced MCP-1 expression. Taken together, these results indicate that high osmolalities activate the transcription factor NFAT5 in mesothelial cells. NFAT5 in turn upregulates MCP-1, likely in combination with NF-κB, and thus may participate in the development of peritoneal fibrosis during CAPD.
Collapse
|
14
|
Küper C, Beck FX, Neuhofer W. Toll-like receptor 4 activates NF-κB and MAP kinase pathways to regulate expression of proinflammatory COX-2 in renal medullary collecting duct cells. Am J Physiol Renal Physiol 2012; 302:F38-46. [DOI: 10.1152/ajprenal.00590.2010] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Binding of bacterial LPS to the Toll-like receptor 4 (TLR4) complex of inner medullary collecting duct (IMCD) cells plays a central role in recognition of ascending bacterial infections and activation of proinflammatory responses. Since proinflammatory cyclooxygenase (COX)-2 is induced in IMCD cells upon LPS exposure, the present study addressed the question of whether TLR4 mediates COX-2 induction in IMCD cells and characterized the underlying signaling mechanisms. Enhanced COX-2 expression and activity in the presence of LPS was diminished by TLR4 inhibition. LPS induced a TLR4-dependent stimulation of NF-κB and the MAPKs p38, ERK1/2, and JNK. Activation of NF-κB was under negative control of JNK, as inhibition of JNK increased NF-κB activity and COX-2 expression. Phosphorylation of p38 and ERK1/2 required TLR4-dependent release of TGF-α with subsequent activation of the epidermal growth factor receptor (EGFR), whereas JNK activation was EGFR independent. Inhibition of p38 or ERK1/2 had no significant effect on LPS-induced NF-κB activation, nor on activator protein 1-, cAMP response element-, or serum response element-driven reporter constructs. However, the transcriptional regulator SP-1 appears to contribute to COX-2 expression after LPS exposure. In conclusion, these results propose that LPS mediates enhanced COX-2 expression in IMCD cells by 1) TLR4-mediated activation of the NF-κB signaling pathway, 2) TLR4-dependent release of TGF-α with subsequent activation of the EGFR and downstream MAPKs p38 and ERK1/2, and 3) TLR4-mediated, EGFR-independent activation of JNK that negatively regulates NF-κB activation.
Collapse
Affiliation(s)
| | | | - Wolfgang Neuhofer
- Departments of Physiology and
- Nephrology, University of Munich, Munich, Germany
| |
Collapse
|
15
|
Islam MA, Yun CH, Choi YJ, Shin JY, Arote R, Jiang HL, Kang SK, Nah JW, Park IK, Cho MH, Cho CS. Accelerated gene transfer through a polysorbitol-based transporter mechanism. Biomaterials 2011; 32:9908-24. [DOI: 10.1016/j.biomaterials.2011.09.013] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Accepted: 09/06/2011] [Indexed: 10/17/2022]
|
16
|
Neuhofer W. Role of NFAT5 in inflammatory disorders associated with osmotic stress. Curr Genomics 2011; 11:584-90. [PMID: 21629436 PMCID: PMC3078683 DOI: 10.2174/138920210793360961] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Revised: 10/05/2010] [Accepted: 10/11/2010] [Indexed: 12/25/2022] Open
Abstract
Nuclear factor of activated T cells 5 (NFAT5) is the most recently described member of the Rel family of transcription factors, including NF-κB and NFAT1-4, which play central roles in inducible gene expression during the immune response. NFAT5 was initially described to drive osmoprotective gene expression in renal medullary cells, which are routinely faced by high extracellular osmolalities. Recent data however indicate profound biological importance of the mammalian osmotic stress response in view of NFAT5 dependent gene regulation in non-renal tissues. In mononuclear cells and epithelial cells, NFAT5 stimulates the expression of various pro-inflammatory cytokines during elevated ambient tonicity. Accordingly, compared to plasma, the interstitial tonicity of lymphoid organs like spleen and thymus and that of liver is substantially hypertonic under physiological conditions. In addition, anisotonic disorders (hypernatremia, diabetes mellitus, dehydration) entail systemic hyperosmolality, and, in inflammatory disorders, the skin, intestine, and cornea are sites of local hyperosmolality. This article summarizes the current knowledge regarding systemic and local osmotic stress in anisotonic and inflammatory disorders in view of NFAT5 activation and regulation, and NFAT5 dependent cytokine production.
Collapse
Affiliation(s)
- Wolfgang Neuhofer
- Departments of Nephrology and Physiology, Inner City Campus, University of Munich, Munich, Germany
| |
Collapse
|
17
|
Roth I, Leroy V, Kwon HM, Martin PY, Féraille E, Hasler U. Osmoprotective transcription factor NFAT5/TonEBP modulates nuclear factor-kappaB activity. Mol Biol Cell 2010; 21:3459-74. [PMID: 20685965 PMCID: PMC2947481 DOI: 10.1091/mbc.e10-02-0133] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Tonicity responsive binding protein (TonEBP) is a transcription factor that plays a key role in osmoprotection. Here, we demonstrate enhanced activity of prosurvival NF-κB—at the onset of hypertonic challenge that depends on p38 kinase—and Akt-dependent formation of p65-TonEBP complexes that bind to elements of NF-κB-responsive genes. Tonicity-responsive binding-protein (TonEBP or NFAT5) is a widely expressed transcription factor whose activity is regulated by extracellular tonicity. TonEBP plays a key role in osmoprotection by binding to osmotic response element/TonE elements of genes that counteract the deleterious effects of cell shrinkage. Here, we show that in addition to this “classical” stimulation, TonEBP protects cells against hypertonicity by enhancing nuclear factor-κB (NF-κB) activity. We show that hypertonicity enhances NF-κB stimulation by lipopolysaccharide but not tumor necrosis factor-α, and we demonstrate overlapping protein kinase B (Akt)-dependent signal transduction pathways elicited by hypertonicity and transforming growth factor-α. Activation of p38 kinase by hypertonicity and downstream activation of Akt play key roles in TonEBP activity, IκBα degradation, and p65 nuclear translocation. TonEBP affects neither of these latter events and is itself insensitive to NF-κB signaling. Rather, we reveal a tonicity-dependent interaction between TonEBP and p65 and show that NF-κB activity is considerably enhanced after binding of NF-κB-TonEBP complexes to κB elements of NF-κB–responsive genes. We demonstrate the key roles of TonEBP and Akt in renal collecting duct epithelial cells and in macrophages. These findings reveal a novel role for TonEBP and Akt in NF-κB activation on the onset of hypertonic challenge.
Collapse
Affiliation(s)
- Isabelle Roth
- Department of Cellular Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | | | | | | | | | | |
Collapse
|