1
|
Grand K, Stoltz M, Rizzo L, Röck R, Kaminski MM, Salinas G, Getwan M, Naert T, Pichler R, Lienkamp SS. HNF1B Alters an Evolutionarily Conserved Nephrogenic Program of Target Genes. J Am Soc Nephrol 2023; 34:412-432. [PMID: 36522156 PMCID: PMC10103355 DOI: 10.1681/asn.2022010076] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 10/11/2022] [Accepted: 11/09/2022] [Indexed: 12/23/2022] Open
Abstract
SIGNIFICANCE STATEMENT Mutations in hepatocyte nuclear factor-1 β ( HNF1B ) are the most common monogenic causes of congenital renal malformations. HNF1B is necessary to directly reprogram fibroblasts to induced renal tubule epithelial cells (iRECs) and, as we demonstrate, can induce ectopic pronephric tissue in Xenopus ectodermal organoids. Using these two systems, we analyzed the effect of HNF1B mutations found in patients with cystic dysplastic kidney disease. We found cross-species conserved targets of HNF1B, identified transcripts that are differentially regulated by the patient-specific mutant protein, and functionally validated novel HNF1B targets in vivo . These results highlight evolutionarily conserved transcriptional mechanisms and provide insights into the genetic circuitry of nephrogenesis. BACKGROUND Hepatocyte nuclear factor-1 β (HNF1B) is an essential transcription factor during embryogenesis. Mutations in HNF1B are the most common monogenic causes of congenital cystic dysplastic renal malformations. The direct functional consequences of mutations in HNF1B on its transcriptional activity are unknown. METHODS Direct reprogramming of mouse fibroblasts to induced renal tubular epithelial cells was conducted both with wild-type HNF1B and with patient mutations. HNF1B was expressed in Xenopus ectodermal explants. Transcriptomic analysis by bulk RNA-Seq identified conserved targets with differentially regulated expression by the wild-type or R295C mutant. CRISPR/Cas9 genome editing in Xenopus embryos evaluated transcriptional targets in vivo . RESULTS HNF1B is essential for reprogramming mouse fibroblasts to induced renal tubular epithelial cells and induces development of ectopic renal organoids from pluripotent Xenopus cells. The mutation R295C retains reprogramming and inductive capacity but alters the expression of specific sets of downstream target genes instead of diminishing overall transcriptional activity of HNF1B. Surprisingly, targets associated with polycystic kidney disease were less affected than genes affected in congenital renal anomalies. Cross-species-conserved transcriptional targets were dysregulated in hnf1b CRISPR-depleted Xenopus embryos, confirming their dependence on hnf1b . CONCLUSIONS HNF1B activates an evolutionarily conserved program of target genes that disease-causing mutations selectively disrupt. These findings provide insights into the renal transcriptional network that controls nephrogenesis.
Collapse
Affiliation(s)
- Kelli Grand
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Martine Stoltz
- The University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ludovica Rizzo
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Ruth Röck
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Michael M. Kaminski
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | | | - Maike Getwan
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Thomas Naert
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Roman Pichler
- The University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Soeren S. Lienkamp
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
- The University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
2
|
Core promoter mutation of nucleotides A1762T and G1764A of hepatitis B virus increases core promoter transactivation by hepatocyte nuclear factor 1. J Microbiol 2022; 60:1039-1047. [DOI: 10.1007/s12275-022-1675-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 08/10/2022] [Accepted: 08/30/2022] [Indexed: 11/26/2022]
|
3
|
Piedrafita A, Balayssac S, Casemayou A, Saulnier-Blache JS, Lucas A, Iacovoni JS, Breuil B, Chauveau D, Decramer S, Malet-Martino M, Schanstra JP, Faguer S. Hepatocyte nuclear factor-1β shapes the energetic homeostasis of kidney tubule cells. FASEB J 2021; 35:e21931. [PMID: 34653285 DOI: 10.1096/fj.202100782rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 08/28/2021] [Accepted: 09/02/2021] [Indexed: 12/17/2022]
Abstract
Energetic metabolism controls key steps of kidney development, homeostasis, and epithelial repair following acute kidney injury (AKI). Hepatocyte nuclear factor-1β (HNF-1β) is a master transcription factor that controls mitochondrial function in proximal tubule (PT) cells. Patients with HNF1B pathogenic variant display a wide range of kidney developmental abnormalities and progressive kidney fibrosis. Characterizing the metabolic changes in PT cells with HNF-1β deficiency may help to identify new targetable molecular hubs involved in HNF1B-related kidney phenotypes and AKI. Here, we combined 1 H-NMR-based metabolomic analysis in a murine PT cell line with CrispR/Cas9-induced Hnf1b invalidation (Hnf1b-/- ), clustering analysis, targeted metabolic assays, and datamining of published RNA-seq and ChIP-seq dataset to identify the role of HNF-1β in metabolism. Hnf1b-/- cells grown in normoxic conditions display intracellular ATP depletion, increased cytosolic lactate concentration, increased lipid droplet content, failure to use pyruvate for energetic purposes, increased levels of tricarboxylic acid (TCA) cycle intermediates and oxidized glutathione, and a reduction of TCA cycle byproducts, all features consistent with mitochondrial dysfunction and an irreversible switch toward glycolysis. Unsupervised clustering analysis showed that Hnf1b-/- cells mimic a hypoxic signature and that they cannot furthermore increase glycolysis-dependent energetic supply during hypoxic challenge. Metabolome analysis also showed alteration of phospholipid biosynthesis in Hnf1b-/- cells leading to the identification of Chka, the gene coding for choline kinase α, as a new putative target of HNF-1β. HNF-1β shapes the energetic metabolism of PT cells and HNF1B deficiency in patients could lead to a hypoxia-like metabolic state precluding further adaptation to ATP depletion following AKI.
Collapse
Affiliation(s)
- Alexis Piedrafita
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France.,Université Paul Sabatier - Toulouse 3, Toulouse, France.,Département de Néphrologie et Transplantation d'Organes, Centre de Référence des Maladies Rénales Rares, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Stéphane Balayssac
- Groupe de RMN Biomédicale, Laboratoire SPCMIB, UMR CNRS 5068, Université Paul Sabatier, Centre National de la Recherche Scientifique, Toulouse, France.,Laboratoire des Interaction Moléculaires et Réactivité Chimique et Photochimique (IMRCP), UMR 5623, Toulouse, France
| | - Audrey Casemayou
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France.,Université Paul Sabatier - Toulouse 3, Toulouse, France.,Département de Néphrologie et Transplantation d'Organes, Centre de Référence des Maladies Rénales Rares, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Jean-Sébastien Saulnier-Blache
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France.,Université Paul Sabatier - Toulouse 3, Toulouse, France
| | - Alexandre Lucas
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France
| | - Jason S Iacovoni
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France
| | - Benjamin Breuil
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France
| | - Dominique Chauveau
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France.,Université Paul Sabatier - Toulouse 3, Toulouse, France.,Département de Néphrologie et Transplantation d'Organes, Centre de Référence des Maladies Rénales Rares, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Stéphane Decramer
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France.,Université Paul Sabatier - Toulouse 3, Toulouse, France.,Service de Néphrologie, Médecine interne et Hypertension artérielle, Hôpital des Enfants, Centre de Référence des Maladies Rénales Rares, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Myriam Malet-Martino
- Groupe de RMN Biomédicale, Laboratoire SPCMIB, UMR CNRS 5068, Université Paul Sabatier, Centre National de la Recherche Scientifique, Toulouse, France
| | - Joost P Schanstra
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France.,Université Paul Sabatier - Toulouse 3, Toulouse, France
| | - Stanislas Faguer
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France.,Université Paul Sabatier - Toulouse 3, Toulouse, France.,Département de Néphrologie et Transplantation d'Organes, Centre de Référence des Maladies Rénales Rares, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| |
Collapse
|
4
|
Megalin-mediated albumin endocytosis in renal proximal tubules is involved in the antiproteinuric effect of angiotensin II type 1 receptor blocker in a subclinical acute kidney injury animal model. Biochim Biophys Acta Gen Subj 2021; 1865:129950. [PMID: 34144121 DOI: 10.1016/j.bbagen.2021.129950] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 06/07/2021] [Accepted: 06/10/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Tubule-interstitial injury (TII) is one of the mechanisms involved in the progression of renal diseases with progressive proteinuria. Angiotensin II (Ang II) type 1 receptor blockers (ARBs) have been successfully used to treat renal diseases. However, the mechanism correlating treatment with ARBs and proteinuria is not completely understood. The hypothesis that the anti-proteinuric effect of losartan is associated with the modulation of albumin endocytosis in PT epithelial cells (PTECs) was assessed. METHODS We used a subclinical acute kidney injury animal model (subAKI) and LLC-PK1 cells, a model of PTECs. RESULTS In subAKI, PT albumin overload induced TII development, measured by: (1) increase in urinary lactate dehydrogenase and γ-glutamyltranspeptidase activity; (2) proteinuria associated with impairment in megalin-mediated albumin reabsorption; (3) increase in luminal and interstitial space in tubular cortical segments. These effects were avoided by treating the animals with losartan, an ARB. Using LLC-PK1 cells, we observed that: (1) 20 mg/mL albumin increased the secretion of Ang II and decreased megalin-mediated albumin endocytosis; (2) the effects of Ang II and albumin were abolished by 10-8 M losartan; (3) MEK/ERK pathway is the molecular mechanism underlying the Ang II-mediated inhibitory effect of albumin on PT albumin endocytosis. CONCLUSION Our results show that PT megalin-mediated albumin endocytosis is a possible target during the treatment of renal diseases patients with ARB. GENERAL SIGNIFICANCE The findings obtained in the present work represents a step forward to the current knowledge on about the role of ARBs in the treatment of renal disease.
Collapse
|
5
|
Chandra S, Srinivasan S, Batra J. Hepatocyte nuclear factor 1 beta: A perspective in cancer. Cancer Med 2021; 10:1791-1804. [PMID: 33580750 PMCID: PMC7940219 DOI: 10.1002/cam4.3676] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/27/2020] [Accepted: 11/30/2020] [Indexed: 12/17/2022] Open
Abstract
Hepatocyte nuclear factor 1 beta (HNF1 β/B) exists as a homeobox transcription factor having a vital role in the embryonic development of organs mainly liver, kidney and pancreas. Initially described as a gene causing maturity‐onset diabetes of the young (MODY), HNF1β expression deregulation and single nucleotide polymorphisms in HNF1β have now been associated with several tumours including endometrial, prostate, ovarian, hepatocellular, renal and colorectal cancers. Its function has been studied either as homodimer or heterodimer with HNF1α. In this review, the role of HNF1B in different cancers will be discussed along with the role of its splice variants, and its emerging role as a potential biomarker in cancer.
Collapse
Affiliation(s)
- Shubhra Chandra
- Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Australian Prostate Cancer Research Centre-Queensland, Queensland University of Technology, Brisbane, QLD, Australia.,Translational Research Institute, Woolloongabba, QLD, Australia
| | - Srilakshmi Srinivasan
- Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Australian Prostate Cancer Research Centre-Queensland, Queensland University of Technology, Brisbane, QLD, Australia.,Translational Research Institute, Woolloongabba, QLD, Australia
| | - Jyotsna Batra
- Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Australian Prostate Cancer Research Centre-Queensland, Queensland University of Technology, Brisbane, QLD, Australia.,Translational Research Institute, Woolloongabba, QLD, Australia
| |
Collapse
|
6
|
Zhang Y, Hao J, Du Z, Li P, Hu J, Ruan M, Li S, Ma Y, Lou Q. Inhibition of hepatocyte nuclear factor 1β contributes to cisplatin nephrotoxicity via regulation of nf-κb pathway. J Cell Mol Med 2021; 25:2861-2871. [PMID: 33512774 PMCID: PMC7957194 DOI: 10.1111/jcmm.16316] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/06/2021] [Accepted: 01/11/2021] [Indexed: 12/12/2022] Open
Abstract
Cisplatin nephrotoxicity has been considered as serious side effect caused by cisplatin‐based chemotherapy. Recent evidence indicates that renal tubular cell apoptosis and inflammation contribute to the progression of cisplatin‐induced acute kidney injury (AKI). Hepatocyte nuclear factor 1β (HNF1β) has been reported to regulate the development of kidney cystogenesis, diabetic nephrotoxicity, etc However, the regulatory mechanism of HNF1β in cisplatin nephrotoxicity is largely unknown. In the present study, we examined the effects of HNF1β deficiency on the development of cisplatin‐induced AKI in vitro and in vivo. HNF1β down‐regulation exacerbated cisplatin‐induced RPTC apoptosis by indirectly inducing NF‐κB p65 phosphorylation and nuclear translocation. HNF1β knockdown C57BL/6 mice were constructed by injecting intravenously with HNF1β‐interfering shRNA and PEI. The HNF1β scramble and knockdown mice were treated with 30 mg/kg cisplatin for 3 days to induce acute kidney injury. Cisplatin treatment caused increased caspase 3 cleavage and p65 phosphorylation, elevated serum urea nitrogen and creatinine, and obvious histological damage of kidney such as fractured tubules in control mice, which were enhanced in HNF1β knockdown mice. These results suggest that HNF1β may ameliorate cisplatin nephrotoxicity in vitro and in vivo, probably through regulating NF‐κB signalling pathway.
Collapse
Affiliation(s)
- Yan Zhang
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, China
| | - Jielu Hao
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Zijun Du
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, China
| | - Peiyao Li
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, China
| | - Jinghua Hu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Mengna Ruan
- Department of Nephrology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Shulian Li
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, China
| | - Yuanfang Ma
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, China
| | - Qiang Lou
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, China
| |
Collapse
|
7
|
Barker H, Parkkila S. Bioinformatic characterization of angiotensin-converting enzyme 2, the entry receptor for SARS-CoV-2. PLoS One 2020; 15:e0240647. [PMID: 33112891 PMCID: PMC7592753 DOI: 10.1371/journal.pone.0240647] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/30/2020] [Indexed: 12/15/2022] Open
Abstract
The World Health Organization declared the COVID-19 epidemic a public health emergency of international concern on March 11th, 2020, and the pandemic is rapidly spreading worldwide. COVID-19 is caused by a novel coronavirus SARS-CoV-2, which enters human target cells via angiotensin converting enzyme 2 (ACE2). We used a number of bioinformatics tools to computationally characterize ACE2 by determining its cell-specific expression in trachea, lung, and small intestine, derive its putative functions, and predict transcriptional regulation. The small intestine expressed higher levels of ACE2 mRNA than any other organ. By immunohistochemistry, duodenum, kidney and testis showed strong signals, whereas the signal was weak in the respiratory tract. Single cell RNA-Seq data from trachea indicated positive signals along the respiratory tract in key protective cell types including club, goblet, proliferating, and ciliary epithelial cells; while in lung the ratio of ACE2-expressing cells was low in all cell types (<2.6%), but was highest in vascular endothelial and goblet cells. Gene ontology analysis suggested that, besides its classical role in the renin-angiotensin system, ACE2 may be functionally associated with angiogenesis/blood vessel morphogenesis. Using a novel tool for the prediction of transcription factor binding sites we identified several putative binding sites within two tissue-specific promoters of the ACE2 gene as well as a new putative short form of ACE2. These include several interferon-stimulated response elements sites for STAT1, IRF8, and IRF9. Our results also confirmed that age and gender play no significant role in the regulation of ACE2 mRNA expression in the lung.
Collapse
Affiliation(s)
- Harlan Barker
- Faculty of Medicine and Health Technology, Tampere University and Fimlab Ltd, Tampere University Hospital, Tampere, Finland
| | - Seppo Parkkila
- Faculty of Medicine and Health Technology, Tampere University and Fimlab Ltd, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
8
|
Ferrè S, Igarashi P. New insights into the role of HNF-1β in kidney (patho)physiology. Pediatr Nephrol 2019; 34:1325-1335. [PMID: 29961928 PMCID: PMC6312759 DOI: 10.1007/s00467-018-3990-7] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/24/2018] [Accepted: 05/25/2018] [Indexed: 12/14/2022]
Abstract
Hepatocyte nuclear factor-1β (HNF-1β) is an essential transcription factor that regulates the development and function of epithelia in the kidney, liver, pancreas, and genitourinary tract. Humans who carry HNF1B mutations develop heterogeneous renal abnormalities, including multicystic dysplastic kidneys, glomerulocystic kidney disease, renal agenesis, renal hypoplasia, and renal interstitial fibrosis. In the embryonic kidney, HNF-1β is required for ureteric bud branching, initiation of nephrogenesis, and nephron segmentation. Ablation of mouse Hnf1b in nephron progenitors causes defective tubulogenesis, whereas later inactivation in elongating tubules leads to cyst formation due to downregulation of cystic disease genes, including Umod, Pkhd1, and Pkd2. In the adult kidney, HNF-1β controls the expression of genes required for intrarenal metabolism and solute transport by tubular epithelial cells. Tubular abnormalities observed in HNF-1β nephropathy include hyperuricemia with or without gout, hypokalemia, hypomagnesemia, and polyuria. Recent studies have identified novel post-transcriptional and post-translational regulatory mechanisms that control HNF-1β expression and activity, including the miRNA cluster miR17 ∼ 92 and the interacting proteins PCBD1 and zyxin. Further understanding of the molecular mechanisms upstream and downstream of HNF-1β may lead to the development of new therapeutic approaches in cystic kidney disease and other HNF1B-related renal diseases.
Collapse
Affiliation(s)
- Silvia Ferrè
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, Texas, USA,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Peter Igarashi
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA. .,Department of Medicine, University of Minnesota Medical School, 420 Delaware St. SE, MMC 194, Minneapolis, MN, 55455, USA.
| |
Collapse
|
9
|
Ferrè S, Deng Y, Huen SC, Lu CY, Scherer PE, Igarashi P, Moe OW. Renal tubular cell spliced X-box binding protein 1 (Xbp1s) has a unique role in sepsis-induced acute kidney injury and inflammation. Kidney Int 2019; 96:1359-1373. [PMID: 31601454 DOI: 10.1016/j.kint.2019.06.023] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 06/04/2019] [Accepted: 06/27/2019] [Indexed: 12/15/2022]
Abstract
Sepsis is a systemic inflammatory state in response to infection, and concomitant acute kidney injury (AKI) increases mortality significantly. Endoplasmic reticulum stress is activated in many cell types upon microbial infection and modulates inflammation. The role of endoplasmic reticulum signaling in the kidney during septic AKI is unknown. Here we tested the role of the spliced X-box binding protein 1 (Xbp1s), a key component of the endoplasmic reticulum stress-activated pathways, in the renal response to sepsis in the lipopolysaccharide (LPS) model. Xbp1s was increased in the kidneys of mice treated with LPS but not in other models of AKI, or several chronic kidney disease models. The functional significance of Xbp1s induction was examined by genetic manipulation in renal tubules. Renal tubule-specific overexpression of Xbp1s caused severe tubule dilation and vacuolation with expression of the injury markers Kim1 and Ngal, the pro-inflammatory molecules interleukin-6 (Il6) and Toll-like receptor 4 (Tlr4), decreased kidney function and 50% mortality in five days. Renal tubule-specific genetic ablation of Xbp1 had no phenotype at baseline. However, after LPS, Xbp1 knockdown mice displayed lower renal NGAL, pro-apoptotic factor CHOP, serum creatinine levels, and a tendency towards lower Tlr4 compared to LPS-treated mice with intact Xbp1s. LPS treatment in Xbp1s-overexpressing mice caused a mild increase in NGAL and CHOP compared to LPS-treated mice without genetic Xbp1s overexpression. Thus, increased Xbp1s signaling in renal tubules is unique to sepsis-induced AKI and contributes to renal inflammation and injury. Inhibition of this pathway may be a potential portal to alleviate injury.
Collapse
Affiliation(s)
- Silvia Ferrè
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| | - Yingfeng Deng
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Sarah C Huen
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Christopher Y Lu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Philipp E Scherer
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Peter Igarashi
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Orson W Moe
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| |
Collapse
|
10
|
Abstract
Hepatocyte nuclear factor 1β (HNF1β) is a transcription factor belonging to the HNF-1 family and has been implicated in a number of cancers, but its role in Wilms' tumor (nephroblastoma) has not been addressed. Here, we compared its expression between Wilms' tumor patient kidney tissue and adjacent tissue based on the Oncomine database ( www.oncomine.com ). Cell proliferation, apoptosis, migration, and HNF1β expression level were analyzed in Wilms' tumor-derived G401 cells. Using a variety of mouse tissues (lung, heart, kidney, etc.), we found that HNF1β is the highest expression in the kidneys. Oncomine analysis further demonstrated that HNF1β has a lower expression in Wilms' tumor tissue than in paracancerous tissues. Overexpression of HNF1β decreased cell proliferation and migration, but promoted cell apoptosis. Knockdown of HNF1β produced the opposite results. These results indicated that HNF1β may play important roles in kidney development and function, and its activation may negatively regulate Wilms' tumor progression.
Collapse
Affiliation(s)
- Yamin Liu
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University , Chongqing , P. R. China
| | - Quist Kanyomse
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University , Chongqing , P. R. China
| | - Yajun Xie
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University , Chongqing , P. R. China
| |
Collapse
|
11
|
Shen Z, Lin J, Teng J, Zhuang Y, Zhang H, Wang C, Zhang Y, Ding X, Zhang X. Association of urinary ionomic profiles and acute kidney injury and mortality in patients after cardiac surgery. J Thorac Cardiovasc Surg 2019; 159:918-926.e5. [PMID: 31351778 DOI: 10.1016/j.jtcvs.2019.02.095] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 01/28/2019] [Accepted: 02/14/2019] [Indexed: 10/27/2022]
Abstract
OBJECTIVE The rarity of sensitive biomarkers for acute kidney injury (AKI) has impeded the timely therapy of AKI. Emerging evidence suggests that ion homeostasis may play pertinent roles in AKI. We aimed to screen out representative urinary ions and build a cardiac surgery-associated AKI indication model. METHODS We performed urinary ionomic analysis from patients undergoing cardiac surgeries in Zhongshan Hospital, Fudan University, Shanghai, China (N = 261). By bioinformatics analysis, we identified differentially changed elements and established the AKI indication model we named the urinary ion index (UII). Follow-ups were performed to evaluate 30-day survival. RESULTS The concentrations of most ions dynamically changed whether a patient developed AKI or not. A significant number of differentially changed elements between AKI and non-AKI groups were detected, especially at 2 hours after cardiac surgery, based on which we generated UII, with the area under the curve of 0.815 ± 0.006 and a cut-off value of 1.24. UII was associated with need for renal replacement therapy, with an area under the curve of 0.83 at a cutoff value of 1.62. Kaplan-Meier and log-rank methods, as well as Cox proportional hazards model, reflected that patients in the UII > 1.24 group had significantly higher risk of mortality within 30 days after surgery (hazard ratio, 5.15; P = .0097 and hazard ratio, 3.56; P = .033) than the UII ≤ 1.24 group. CONCLUSIONS Our data demonstrate that UII appears to be a novel and valid index of early cardiac surgery-associated AKI. UII > 1.24 at 2 hours after surgery indicates high risk of AKI and less 30-day survival.
Collapse
Affiliation(s)
- Ziyan Shen
- Kidney and Dialysis Institute of Shanghai, Shanghai, China; Kidney and Blood Purification Laboratory of Shanghai, Shanghai, China; Department of Nephrology, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jie Lin
- College of Life Sciences & Oceanography, Shenzhen University, Shenzhen, Guangdong Province, China
| | - Jie Teng
- Kidney and Dialysis Institute of Shanghai, Shanghai, China; Kidney and Blood Purification Laboratory of Shanghai, Shanghai, China; Department of Nephrology, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yamin Zhuang
- Department of Cardiac Surgery, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Han Zhang
- Kidney and Dialysis Institute of Shanghai, Shanghai, China; Kidney and Blood Purification Laboratory of Shanghai, Shanghai, China; Department of Nephrology, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chunsheng Wang
- Department of Cardiac Surgery, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan Zhang
- College of Life Sciences & Oceanography, Shenzhen University, Shenzhen, Guangdong Province, China
| | - Xiaoqiang Ding
- Kidney and Dialysis Institute of Shanghai, Shanghai, China; Kidney and Blood Purification Laboratory of Shanghai, Shanghai, China; Department of Nephrology, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, China
| | - Xiaoyan Zhang
- Kidney and Dialysis Institute of Shanghai, Shanghai, China; Kidney and Blood Purification Laboratory of Shanghai, Shanghai, China; Department of Nephrology, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
12
|
The molecular functions of hepatocyte nuclear factors - In and beyond the liver. J Hepatol 2018; 68:1033-1048. [PMID: 29175243 DOI: 10.1016/j.jhep.2017.11.026] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 11/16/2017] [Accepted: 11/20/2017] [Indexed: 12/27/2022]
Abstract
The hepatocyte nuclear factors (HNFs) namely HNF1α/β, FOXA1/2/3, HNF4α/γ and ONECUT1/2 are expressed in a variety of tissues and organs, including the liver, pancreas and kidney. The spatial and temporal manner of HNF expression regulates embryonic development and subsequently the development of multiple tissues during adulthood. Though the HNFs were initially identified individually based on their roles in the liver, numerous studies have now revealed that the HNFs cross-regulate one another and exhibit synergistic relationships in the regulation of tissue development and function. The complex HNF transcriptional regulatory networks have largely been elucidated in rodent models, but less so in human biological systems. Several heterozygous mutations in these HNFs were found to cause diseases in humans but not in rodents, suggesting clear species-specific differences in mutational mechanisms that remain to be uncovered. In this review, we compare and contrast the expression patterns of the HNFs, the HNF cross-regulatory networks and how these liver-enriched transcription factors serve multiple functions in the liver and beyond, extending our focus to the pancreas and kidney. We also summarise the insights gained from both human and rodent studies of mutations in several HNFs that are known to lead to different disease conditions.
Collapse
|
13
|
Yu Y, Cui H, Chen C, Wen G, Xu J, Zheng B, Zhang J, Wang C, Chai Y, Mei J. Hypoxia-inducible Factor-1α directs renal regeneration induced by decellularized scaffolds. Biomaterials 2018; 165:48-55. [PMID: 29501969 DOI: 10.1016/j.biomaterials.2018.02.045] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 02/19/2018] [Accepted: 02/22/2018] [Indexed: 01/09/2023]
Abstract
Although mammalian kidney regeneration has been reported to occur throughout life, mature kidneys in mammals are not thought to regenerate sufficiently, particularly glomeruli. In our previous work, we found that renal regeneration could be enhanced by decellularized renal scaffolds after partial nephrectomy. In this study, we verified that the enhanced renal regeneration mediated by decellularized scaffolds could be attributed to regenerated glomeruli, which were counted both indirectly and directly under a microscope. Using the isobaric tag for relative and absolute quantitation, we performed proteomics analysis and found that hypoxia-inducible factor (HIF)-1α may be a key factor involved in induced renal regeneration. Dimethyloxyallyl glycine (DMOG), a propyl hydroxylase inhibitor, was applied to stabilize constitutive expression of HIF-1α protein, and small interfering RNA was used to inhibit gene expression. Administration of DMOG to decellularized scaffold-grafted rats improved the induced renal regeneration, whereas siHif1α transfection decreased the regeneration capacity. These findings revealed the critical role of HIF-1α in renal regeneration and provided important insights into our understanding of kidney development and the treatment of various kidney diseases.
Collapse
Affiliation(s)
- Yaling Yu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China; Anatomy Department, Wenzhou Medical University, Wenzhou, 325035, China
| | - Haomin Cui
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Chuan Chen
- Department of Hand Surgery, Ningbo No.6 Hospital, Ningbo, 315040, China
| | - Gen Wen
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Jia Xu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Binbin Zheng
- Anatomy Department, Wenzhou Medical University, Wenzhou, 325035, China
| | - Jianse Zhang
- Anatomy Department, Wenzhou Medical University, Wenzhou, 325035, China
| | - Chunyang Wang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Yimin Chai
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| | - Jin Mei
- Anatomy Department, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
14
|
Casemayou A, Fournel A, Bagattin A, Schanstra J, Belliere J, Decramer S, Marsal D, Gillet M, Chassaing N, Huart A, Pontoglio M, Knauf C, Bascands JL, Chauveau D, Faguer S. Hepatocyte Nuclear Factor-1 β Controls Mitochondrial Respiration in Renal Tubular Cells. J Am Soc Nephrol 2017; 28:3205-3217. [PMID: 28739648 DOI: 10.1681/asn.2016050508] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 05/18/2017] [Indexed: 12/19/2022] Open
Abstract
AKI is a frequent condition that involves renal microcirculation impairment, infiltration of inflammatory cells with local production of proinflammatory cytokines, and subsequent epithelial disorders and mitochondrial dysfunction. Peroxisome proliferator-activated receptor γ coactivator 1-α (PPARGC1A), a coactivator of the transcription factor PPAR-γ that controls mitochondrial biogenesis and function, has a pivotal role in the early dysfunction of the proximal tubule and the subsequent renal repair. Here, we evaluated the potential role of hepatocyte nuclear factor-1β (HNF-1β) in regulating PPARGC1A expression in AKI. In mice, endotoxin injection to induce AKI also induced early and transient inflammation and PPARGC1A inhibition, which overlapped with downregulation of the HNF-1β transcriptional network. In vitro, exposure of proximal tubule cells to the inflammatory cytokines IFN-γ and TNF-α led to inhibition of HNF-1β transcriptional activity. Moreover, inhibition of HNF-1β significantly reduced PPARGC1A expression and altered mitochondrial morphology and respiration in proximal tubule cells. Chromatin immunoprecipitation assays and PCR analysis confirmed HNF-1β binding to the Ppargc1a promoter in mouse kidneys. We also demonstrated downregulation of renal PPARGC1A expression in a patient with an HNF1B germinal mutation. Thus, we propose that HNF-1β links extracellular inflammatory signals to mitochondrial dysfunction during AKI partly via PPARGC1A signaling. Our findings further strengthen the view of HNF1B-related nephropathy as a mitochondrial disorder in adulthood.
Collapse
Affiliation(s)
- Audrey Casemayou
- Institut National de la Santé et de la Recherche Médicale, U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France.,University Toulouse III Paul-Sabatier, Toulouse, France
| | - Audren Fournel
- University Toulouse III Paul-Sabatier, Toulouse, France.,Institut National de la Santé et de la Recherche Médicale U1220, Institut de Recherche en Santé Digestive (IRSD), CHU Purpan-BP3028, 31024 Toulouse Cedex 3
| | - Alessia Bagattin
- Laboratoire d'Expression Génique, Développement et Maladies, Département Développement, Reproduction et Cancer, Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France
| | - Joost Schanstra
- Institut National de la Santé et de la Recherche Médicale, U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France.,University Toulouse III Paul-Sabatier, Toulouse, France
| | - Julie Belliere
- Institut National de la Santé et de la Recherche Médicale, U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France.,University Toulouse III Paul-Sabatier, Toulouse, France.,Department of Nephrology and Organ Transplantation, Center for Rare Renal Diseases, University Hospital of Toulouse, Toulouse, France
| | - Stéphane Decramer
- Institut National de la Santé et de la Recherche Médicale, U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France.,University Toulouse III Paul-Sabatier, Toulouse, France.,Department of Nephrology, Internal Medicine and Hypertension, Center for Rare Renal Diseases, Children' Hospital, University Hospital of Toulouse, Toulouse, France
| | - Dimitri Marsal
- Institut National de la Santé et de la Recherche Médicale, U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France.,University Toulouse III Paul-Sabatier, Toulouse, France
| | - Marion Gillet
- Institut National de la Santé et de la Recherche Médicale, U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France.,University Toulouse III Paul-Sabatier, Toulouse, France
| | - Nicolas Chassaing
- Department of Medical Genetics, Hôpital Purpan, University Hospital of Toulouse, Toulouse, France; and
| | - Antoine Huart
- Department of Nephrology and Organ Transplantation, Center for Rare Renal Diseases, University Hospital of Toulouse, Toulouse, France
| | - Marco Pontoglio
- Laboratoire d'Expression Génique, Développement et Maladies, Département Développement, Reproduction et Cancer, Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France
| | - Claude Knauf
- University Toulouse III Paul-Sabatier, Toulouse, France.,Institut National de la Santé et de la Recherche Médicale U1220, Institut de Recherche en Santé Digestive (IRSD), CHU Purpan-BP3028, 31024 Toulouse Cedex 3
| | - Jean-Loup Bascands
- Institut National de la Santé et de la Recherche Médicale, U1188, DéTROI (Diabète Athérothrombose Thérapies Réunion Océan Indien), University of La Réunion
| | - Dominique Chauveau
- Institut National de la Santé et de la Recherche Médicale, U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France.,University Toulouse III Paul-Sabatier, Toulouse, France.,Department of Nephrology and Organ Transplantation, Center for Rare Renal Diseases, University Hospital of Toulouse, Toulouse, France
| | - Stanislas Faguer
- Institut National de la Santé et de la Recherche Médicale, U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France; .,University Toulouse III Paul-Sabatier, Toulouse, France.,Department of Nephrology and Organ Transplantation, Center for Rare Renal Diseases, University Hospital of Toulouse, Toulouse, France
| |
Collapse
|
15
|
Chen Q, Cui Y, Ding G, Jia Z, Zhang Y, Zhang A, Huang S. PEA3 protects against gentamicin nephrotoxicity: role of mitochondrial dysfunction. Am J Transl Res 2017; 9:2153-2162. [PMID: 28559968 PMCID: PMC5446500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 04/10/2017] [Indexed: 06/07/2023]
Abstract
Toxin-induced nephrotoxicity is one of the major causes leading to the acute kidney injury (AKI). Among these nephrotoxic toxins, gentamicin can induce AKI with elusive mechanisms. Emerging evidence demonstrated that PEA3 (polyomavirus enhancer activator 3) contributed to the nephrogenesis, while its role in AKI remains unknown. Thus, this study was to investigate the role of PEA3 in gentamicin nephrotoxicity, as well as the underlying mechanisms. In rats, gentamicin treatment (200 mg/kg twice per day) for two days induced remarkable kidney injury with a peak damage on day 5 evaluated by the tubular injury score, proteinuria, and tubular injury markers of NGAL and KIM-1. In parallel with the tubular injury, PEA3 protein and mRNA expressions were significantly upregulated by gentamicin and peaked on day 5. To define the role of PEA3 in gentamicin nephrotoxicity, proximal tubule cells were transfected with PEA3 plasmid with or without gentamicin treatment (1 mg/ml). Notably, overexpression of PEA3 attenuated gentamicin-induced cell injury shown by the ameliorated cell apoptosis and NGAL and KIM-1 upregulation. Meantime, gentamicin caused severe mitochondrial dysfunction, which was largely normalized by PEA3 overexpression. In contrast, silencing PEA3 by a siRNA strategy further deteriorated gentamicin-induced cell apoptosis and mitochondrial dysfunction. In sum, PEA3 protected against gentamicin nephrotoxicity possibly via a mitochondrial mechanism.
Collapse
Affiliation(s)
- Qiuxia Chen
- Department of Nephrology, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, Jiangsu Province, P. R. China
- Jiangsu Key Laboratory of PediatricsNanjing 210029, Jiangsu Province, P. R. China
| | - Yiyun Cui
- Department of Nephrology, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, Jiangsu Province, P. R. China
- Jiangsu Key Laboratory of PediatricsNanjing 210029, Jiangsu Province, P. R. China
| | - Guixia Ding
- Department of Nephrology, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, Jiangsu Province, P. R. China
- Jiangsu Key Laboratory of PediatricsNanjing 210029, Jiangsu Province, P. R. China
| | - Zhanjun Jia
- Department of Nephrology, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, Jiangsu Province, P. R. China
- Jiangsu Key Laboratory of PediatricsNanjing 210029, Jiangsu Province, P. R. China
| | - Yue Zhang
- Jiangsu Key Laboratory of PediatricsNanjing 210029, Jiangsu Province, P. R. China
| | - Aihua Zhang
- Department of Nephrology, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, Jiangsu Province, P. R. China
- Jiangsu Key Laboratory of PediatricsNanjing 210029, Jiangsu Province, P. R. China
| | - Songming Huang
- Department of Nephrology, Children’s Hospital of Nanjing Medical UniversityNanjing 210008, Jiangsu Province, P. R. China
- Jiangsu Key Laboratory of PediatricsNanjing 210029, Jiangsu Province, P. R. China
| |
Collapse
|
16
|
Hao J, Lou Q, Wei Q, Mei S, Li L, Wu G, Mi QS, Mei C, Dong Z. MicroRNA-375 Is Induced in Cisplatin Nephrotoxicity to Repress Hepatocyte Nuclear Factor 1-β. J Biol Chem 2017; 292:4571-4582. [PMID: 28119452 DOI: 10.1074/jbc.m116.754929] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 01/19/2017] [Indexed: 12/25/2022] Open
Abstract
Nephrotoxicity is a major adverse effect of cisplatin-mediated chemotherapy in cancer patients. The pathogenesis of cisplatin-induced nephrotoxicity remains largely unclear, making it difficult to design effective renoprotective approaches. Here, we have examined the role of microRNAs (miRNAs) in cisplatin-induced nephrotoxicity. We show that cisplatin nephrotoxicity was not affected by overall depletion of both beneficial and detrimental miRNAs from kidney proximal tubular cells in mice in which the miRNA-generating enzyme Dicer had been conditionally knocked out. To identify miRNAs involved in cisplatin nephrotoxicity, we used microarray analysis to profile miRNA expression and identified 47 up-regulated microRNAs and 20 down-regulated microRNAs in kidney cortical tissues. One up-regulated miRNA was miR-375, whose expression was also induced in cisplatin-treated renal tubular cells. Interestingly, inhibition of miR-375 decreased cisplatin-induced apoptosis, suggesting that miR-375 is a cell-damaging or pro-apoptotic agent. Blockade of P53 or NF-κB attenuated cisplatin-induced miR-375 expression, supporting a role of P53 and NF-κB in miR-375 induction. We also identified hepatocyte nuclear factor 1 homeobox B (HNF-1β) as a key downstream target of miR-375. Of note, we further demonstrated that HNF-1β protected renal cells against cisplatin-induced apoptosis. Together, these results suggest that upon cisplatin exposure, P53 and NF-κB collaboratively induce miR-375 expression, which, in turn, represses HNF-1β activity, resulting in renal tubular cell apoptosis and nephrotoxicity.
Collapse
Affiliation(s)
- Jielu Hao
- From the Department of Nephrology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China.,the Department of Cellular Biology and Anatomy and
| | - Qiang Lou
- the Department of Cellular Biology and Anatomy and.,the Antibody Drug Engineering Laboratory of Henan Province, Henan University School of Medicine, Kaifeng, Henan 475004, China
| | - Qingqing Wei
- the Department of Cellular Biology and Anatomy and
| | - Shuqin Mei
- From the Department of Nephrology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China.,the Department of Cellular Biology and Anatomy and
| | - Lin Li
- From the Department of Nephrology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China.,the Department of Cellular Biology and Anatomy and
| | - Guangyu Wu
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia 30912
| | - Qing-Sheng Mi
- the Departments of Dermatology and Internal Medicine, Henry Ford Health System, Detroit, Michigan 48202, and
| | - Changlin Mei
- From the Department of Nephrology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China,
| | - Zheng Dong
- the Department of Cellular Biology and Anatomy and .,the Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
17
|
Regenerative pharmacology for the treatment of acute kidney injury: Skeletal muscle stem/progenitor cells for renal regeneration? Pharmacol Res 2016; 113:802-807. [PMID: 27001227 DOI: 10.1016/j.phrs.2016.03.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Revised: 02/25/2016] [Accepted: 03/13/2016] [Indexed: 01/01/2023]
Abstract
Regenerative pharmacology and advanced therapy medicinal products is a relatively new and challenging field in drug development. Acute kidney injury (AKI) is a common clinical condition in nephrology with increasing incidence and high mortality rate. During the last few decades, researchers have been eagerly trying to find novel therapeutic strategies for AKI treatment, including advanced pharmacological therapies using mesenchymal stem cells (MSCs). Several types of MSCs have been thoroughly investigated, including bone marrow, adipose derived and umbilical cord blood MSCs and shown promising results in kidney repair. Research has demonstrated, that MSCs exert their effect through reduction of apoptosis, increased production of growth factors, suppression of oxidative stress and inflammatory processes, promotion of renal tubular cell proliferation, as well as by migration and direct incorporation into the renal tissue. Skeletal muscle-derived stem/progenitor cells (MDSPCs) are mesenchymal stem cell lineage of multipotent cells, demonstrating long-term proliferation, high self-renewal capacities, and ability to enhance endogenous tissue repair. The capacity of MDSPCs to regenerate a variety of different tissues following acute injury or destructive tissue diseases have been demonstrated in preclinical and clinical studies. MDSPCs were also reported to promote endogenous tissue repair via paracrine pathway. Considering advantageous properties of MDSPCs, the administration of these cells might be considered as a potential strategy for the treatment of AKI. However, to date, the therapeutic effect of MDSPCs for renal regeneration has not been investigated. This review reflects the current development in AKI treatment using different types of MSCs and the pilot results of the experimental study in vivo using a novel type of stem cells - MDSPCs for the treatment of gentamicin-induced AKI.
Collapse
|
18
|
Mandai M, Amano Y, Yamaguchi K, Matsumura N, Baba T, Konishi I. Ovarian clear cell carcinoma meets metabolism; HNF-1β confers survival benefits through the Warburg effect and ROS reduction. Oncotarget 2015; 6:30704-14. [PMID: 26375553 PMCID: PMC4741562 DOI: 10.18632/oncotarget.5228] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 08/10/2015] [Indexed: 12/21/2022] Open
Abstract
Ovarian clear cell carcinoma (OCCC) constitutes one of the subtypes of ovarian cancers, but it has unique clinical, histological and biological characteristics, one of which is chemo-resistance. It is also known to develop from endometriotic cyst, a benign ovarian tumor, at relatively high frequency. Recently, it is becoming well known that most of OCCCs express HNF1β, a transcription factor, which is closely associated with the development of liver, pancreas and kidney, as well as occurrence of familial forms of type 2 diabetes. Expression of HNF1β is now regarded as a hallmark of this tumor. Nevertheless, exact biological function of this gene in OCCC has not been clarified. We have shown in previous studies that microenvironment in endometriotic cysts contains severe oxidative stress and OCCC develops under such stressful environment as stress-resistant tumor, which may lead to chemo-resistance. We also showed that increased expression of HNF1β facilitates glucose uptake and glycolysis, which is known as Warburg effect. In the previous issue of this journal, by using comprehensive metabolome analysis, we report that HNF1β actually reduces and protects themselves from internal oxidative stress by dramatically changing cellular metabolism. In this article, we review the relevance and significance of cancer-specific metabolism and how they are associated with biological characteristics of OCCC via expression of HNF1β, along with future clinical implications of targeting cancer-specific metabolism.
Collapse
Affiliation(s)
- Masaki Mandai
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Obstetrics and Gynecology, Kinki University Faculty of Medicine, Osaka-Sayama, Japan
| | - Yasuaki Amano
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ken Yamaguchi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Noriomi Matsumura
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tsukasa Baba
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ikuo Konishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
19
|
Sordi R, Chiazza F, Johnson FL, Patel NSA, Brohi K, Collino M, Thiemermann C. Inhibition of IκB Kinase Attenuates the Organ Injury and Dysfunction Associated with Hemorrhagic Shock. Mol Med 2015; 21:563-75. [PMID: 26101953 DOI: 10.2119/molmed.2015.00049] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 06/16/2015] [Indexed: 12/21/2022] Open
Abstract
Nuclear factor-kappa B (NF-κB) activation is widely implicated in multiple organ failure (MOF); however, a direct inhibitor of IκB kinase (IKK), which plays a pivotal role in the activation of NF-κB, has not been investigated in shock. Thus, the aim of the present work was to investigate the effects of an IKK inhibitor on the MOF associated with hemorrhagic shock (HS). Therefore, rats were subjected to HS and were resuscitated with the shed blood. Rats were treated with the inhibitor of IKK or vehicle at resuscitation. Four hours later, blood and organs were assessed for organ injury and signaling events involved in the activation of NF-κB. Additionally, survival following serum deprivation was assessed in HK-2 cells treated with the inhibitor of IKK. HS resulted in renal dysfunction, lung, liver and muscular injury, and increases in serum inflammatory cytokines. Kidney and liver tissue from HS rats revealed increases in phosphorylation of IKKαβ and IκBα, nuclear translocation of NF-κB and expression of inducible isoform of nitric oxide synthase (iNOS). IKK16 treatment upon resuscitation attenuated NF-κB activation and activated the Akt survival pathway, leading to a significant attenuation of all of the above parameters. Furthermore, IKK16 exhibited cytoprotective effects in human kidney cells. In conclusion, the inhibitor of IKK complex attenuated the MOF associated with HS. This effect may be due to the inhibition of the NF-κB pathway and activation of the survival kinase Akt. Thus, the inhibition of the IKK complex might be an effective strategy for the prevention of MOF associated with HS.
Collapse
Affiliation(s)
- Regina Sordi
- The William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, United Kingdom.,Capes Foundation, Ministry of Education of Brazil, Brasilia, DF, Brazil
| | - Fausto Chiazza
- University of Turin, Department of Drug Science and Technology, Turin, Italy
| | - Florence L Johnson
- The William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, United Kingdom
| | - Nimesh S A Patel
- The William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, United Kingdom
| | - Karim Brohi
- Blizard Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, United Kingdom
| | - Massimo Collino
- University of Turin, Department of Drug Science and Technology, Turin, Italy
| | - Christoph Thiemermann
- The William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
20
|
Oral delivery of prolyl hydroxylase inhibitor: AKB-4924 promotes localized mucosal healing in a mouse model of colitis. Inflamm Bowel Dis 2015; 21:267-75. [PMID: 25545377 DOI: 10.1097/mib.0000000000000277] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Pharmacological induction of hypoxia-inducible factor (HIF), a global transcriptional regulator of the hypoxic response, by prolyl hydroxylase inhibitors (PHDi) is protective in murine models of colitis, and epithelial cells are critical for the observed therapeutic efficacy. Because systemic HIF activation may lead to potentially negative off-target effects, we hypothesized that targeting epithelial HIF through oral delivery of PHDi would be sufficient to protect against colitis in a mouse model. METHODS Using a chemically induced trinitrobenzene sulfonic acid murine model of colitis, we compared the efficacy of oral and intraperitoneal (i.p.) delivery of the PHDi; AKB-4924 in preventing colitis, as measured by endoscopy, histology, barrier integrity, and immune profiling. Furthermore, we measured potential off-target effects, examining HIF and HIF target genes in the heart and kidney, as well as erythropoietin and hematocrit levels. RESULTS Oral administration of AKB-4924 exhibited mucosal protection comparable i.p. dosing. Oral delivery of PHDi led to reduced colonic epithelial HIF stabilization compared with i.p. delivery, but this was still sufficient to induce transcription of downstream HIF targets. Furthermore, oral delivery of PHDi led to reduced stabilization of HIF and activation of HIF targets in extraintestinal organs. CONCLUSIONS Oral delivery of PHDi therapies to this intestinal mucosa protects against colitis in animal models and represents a potential therapeutic strategy for inflammatory bowel disease, which also precludes unwanted extraintestinal effects.
Collapse
|
21
|
Schanstra JP, Zürbig P, Alkhalaf A, Argiles A, Bakker SJL, Beige J, Bilo HJG, Chatzikyrkou C, Dakna M, Dawson J, Delles C, Haller H, Haubitz M, Husi H, Jankowski J, Jerums G, Kleefstra N, Kuznetsova T, Maahs DM, Menne J, Mullen W, Ortiz A, Persson F, Rossing P, Ruggenenti P, Rychlik I, Serra AL, Siwy J, Snell-Bergeon J, Spasovski G, Staessen JA, Vlahou A, Mischak H, Vanholder R. Diagnosis and Prediction of CKD Progression by Assessment of Urinary Peptides. J Am Soc Nephrol 2015; 26:1999-2010. [PMID: 25589610 DOI: 10.1681/asn.2014050423] [Citation(s) in RCA: 196] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 09/30/2014] [Indexed: 01/13/2023] Open
Abstract
Progressive CKD is generally detected at a late stage by a sustained decline in eGFR and/or the presence of significant albuminuria. With the aim of early and improved risk stratification of patients with CKD, we studied urinary peptides in a large cross-sectional multicenter cohort of 1990 individuals, including 522 with follow-up data, using proteome analysis. We validated that a previously established multipeptide urinary biomarker classifier performed significantly better in detecting and predicting progression of CKD than the current clinical standard, urinary albumin. The classifier was also more sensitive for identifying patients with rapidly progressing CKD. Compared with the combination of baseline eGFR and albuminuria (area under the curve [AUC]=0.758), the addition of the multipeptide biomarker classifier significantly improved CKD risk prediction (AUC=0.831) as assessed by the net reclassification index (0.303±-0.065; P<0.001) and integrated discrimination improvement (0.058±0.014; P<0.001). Correlation of individual urinary peptides with CKD stage and progression showed that the peptides that associated with CKD, irrespective of CKD stage or CKD progression, were either fragments of the major circulating proteins, suggesting failure of the glomerular filtration barrier sieving properties, or different collagen fragments, suggesting accumulation of intrarenal extracellular matrix. Furthermore, protein fragments associated with progression of CKD originated mostly from proteins related to inflammation and tissue repair. Results of this study suggest that urinary proteome analysis might significantly improve the current state of the art of CKD detection and outcome prediction and that identification of the urinary peptides allows insight into various ongoing pathophysiologic processes in CKD.
Collapse
Affiliation(s)
- Joost P Schanstra
- Institute of Cardiovascular and Metabolic Disease, French Institute of Health and Medical Research U1048, Toulouse, France; Paul Sabatier University (Toulouse III), Toulouse, France
| | | | - Alaa Alkhalaf
- University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | | | - Stephan J L Bakker
- University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Joachim Beige
- KfH Renal Unit, Department Nephrology, Leipzig and Martin-Luther-University, Halle/Wittenberg, Germany
| | - Henk J G Bilo
- University Medical Center Groningen and University of Groningen, Groningen, The Netherlands; Diabetes Centre, Isala Clinics, Zwolle, The Netherlands
| | - Christos Chatzikyrkou
- Department of Nephrology and Hypertension, University Hospital of Magdeburg, Magdeburg, Germany
| | | | - Jesse Dawson
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Christian Delles
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Hermann Haller
- Department of Nephrology and Hypertension, Medical School of Hanover, Hanover, Germany
| | - Marion Haubitz
- Department of Nephrology, Klinikum Fulda gAG, Fulda, Germany
| | - Holger Husi
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research, RWTH Aachen University Hospital, Aachen, Germany; Department of Internal Medicine IV, Charity Medical University of Berlin, Berlin, Germany
| | - George Jerums
- Austin Health, University of Melbourne, Heidelberg, Australia
| | - Nanne Kleefstra
- University Medical Center Groningen and University of Groningen, Groningen, The Netherlands; Diabetes Centre, Isala Clinics, Zwolle, The Netherlands
| | - Tatiana Kuznetsova
- Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium; Faculty of Health, University of Copenhagen, Copenhagen, Denmark
| | - David M Maahs
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado
| | - Jan Menne
- Department of Nephrology and Hypertension, Medical School of Hanover, Hanover, Germany
| | - William Mullen
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Alberto Ortiz
- School of Medicine, Jimenez Diaz Foundation Institute for Health Research, Autonomous University of Madrid, Madrid, Spain
| | | | - Peter Rossing
- Steno Diabetes Center, Gentofte, Denmark; Faculty of Health, University of Aarhus, Aarhus, Denmark; Faculty of Health, University of Copenhagen, Copenhagen, Denmark
| | | | - Ivan Rychlik
- Second Department of Internal Medicine, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Andreas L Serra
- Division of Nephrology, University Hospital, and Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Switzerland
| | - Justyna Siwy
- mosaiques diagnostics GmbH, Hanover, Germany; Department of Internal Medicine IV, Charity Medical University of Berlin, Berlin, Germany
| | - Janet Snell-Bergeon
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado
| | - Goce Spasovski
- University Department of Nephrology, Medical Faculty, University of Skopje, Skopje, Macedonia
| | - Jan A Staessen
- Faculty of Health, University of Copenhagen, Copenhagen, Denmark
| | - Antonia Vlahou
- Division of Biotechnology, Biomedical Research Foundation, Academy of Athens, Athens, Greece; School of Biomedical and Healthcare Sciences, Plymouth University, Plymouth, United Kingdom; and
| | - Harald Mischak
- Faculty of Health, University of Copenhagen, Copenhagen, Denmark
| | - Raymond Vanholder
- Nephrology Section, Department of Internal Medicine, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|