1
|
Bantounas I, Rooney KM, Lopes FM, Tengku F, Woods S, Zeef LAH, Lin IH, Kuba SY, Bates N, Hummelgaard S, Hillman KA, Cereghini S, Woolf AS, Kimber SJ. Human pluripotent stem cell-derived kidney organoids reveal tubular epithelial pathobiology of heterozygous HNF1B-associated dysplastic kidney malformations. Stem Cell Reports 2024; 19:859-876. [PMID: 38788724 PMCID: PMC11297557 DOI: 10.1016/j.stemcr.2024.04.011] [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: 02/14/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
Hepatocyte nuclear factor 1B (HNF1B) encodes a transcription factor expressed in developing human kidney epithelia. Heterozygous HNF1B mutations are the commonest monogenic cause of dysplastic kidney malformations (DKMs). To understand their pathobiology, we generated heterozygous HNF1B mutant kidney organoids from CRISPR-Cas9 gene-edited human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) reprogrammed from a family with HNF1B-associated DKMs. Mutant organoids contained enlarged malformed tubules displaying deregulated cell turnover. Numerous genes implicated in Mendelian kidney tubulopathies were downregulated, and mutant tubules resisted the cyclic AMP (cAMP)-mediated dilatation seen in controls. Bulk and single-cell RNA sequencing (scRNA-seq) analyses indicated abnormal Wingless/Integrated (WNT), calcium, and glutamatergic pathways, the latter hitherto unstudied in developing kidneys. Glutamate ionotropic receptor kainate type subunit 3 (GRIK3) was upregulated in malformed mutant nephron tubules and prominent in HNF1B mutant fetal human dysplastic kidney epithelia. These results reveal morphological, molecular, and physiological roles for HNF1B in human kidney tubule differentiation and morphogenesis illuminating the developmental origin of mutant-HNF1B-causing kidney disease.
Collapse
Affiliation(s)
- Ioannis Bantounas
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, and the Manchester Academic Health Science Centre, Manchester, UK
| | - Kirsty M Rooney
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, and the Manchester Academic Health Science Centre, Manchester, UK
| | - Filipa M Lopes
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, and the Manchester Academic Health Science Centre, Manchester, UK
| | - Faris Tengku
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, and the Manchester Academic Health Science Centre, Manchester, UK
| | - Steven Woods
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, and the Manchester Academic Health Science Centre, Manchester, UK
| | - Leo A H Zeef
- Bioinformatics Core Facility, University of Manchester, Manchester, UK
| | - I-Hsuan Lin
- Bioinformatics Core Facility, University of Manchester, Manchester, UK
| | - Shweta Y Kuba
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, and the Manchester Academic Health Science Centre, Manchester, UK
| | - Nicola Bates
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, and the Manchester Academic Health Science Centre, Manchester, UK
| | - Sandra Hummelgaard
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, and the Manchester Academic Health Science Centre, Manchester, UK; Department of Biomedicine, Aarhus University, Denmark
| | - Katherine A Hillman
- Manchester Institute of Nephrology and Transplantation, Manchester University NHS Foundation Trust, Manchester, UK
| | - Silvia Cereghini
- Sorbonne Université, CNRS, Institut de Biologie Paris Seine, Laboratorial de Biologie du Développement, IBPS, UMR7622, F-75005 Paris, France
| | - Adrian S Woolf
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, and the Manchester Academic Health Science Centre, Manchester, UK; Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester, UK.
| | - Susan J Kimber
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, and the Manchester Academic Health Science Centre, Manchester, UK.
| |
Collapse
|
2
|
Kelam J, Kelam N, Filipović N, Komić L, Racetin A, Komić D, Kostić S, Kuzmić Prusac I, Vukojević K. Expression of Congenital Anomalies of the Kidney and Urinary Tract (CAKUT) Candidate Genes EDA2R, PCDH9, and TRAF7 in Normal Human Kidney Development and CAKUT. Genes (Basel) 2024; 15:702. [PMID: 38927638 PMCID: PMC11203332 DOI: 10.3390/genes15060702] [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: 05/07/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Approximately half of the cases of chronic kidney disease (CKD) in childhood are caused by congenital anomalies of the kidney and urinary tract (CAKUT). Specific genes were identified as having significant importance in regard to the underlying genetic factors responsible for the CAKUT phenotype, and in our research, we focused on analyzing and comparing the expression levels of ectodysplasin A2 receptor (EDA2R), protocadherin9 (PCDH9), and TNF receptor-associated factor 7 (TRAF7) proteins in the cortex and medulla of healthy control kidneys during developmental phases 2, 3, and 4. We also performed an analysis of the area percentages of the mentioned proteins in the cortical and medullary sections of healthy embryonic and fetal kidneys compared to those affected by CAKUT, including duplex kidneys (DK), horseshoe kidneys (HK), hypoplastic kidneys (HYP), and dysplastic kidneys (DYS). We found that the CAKUT candidate gene proteins EDA2R, PCDH9, and TRAF7 are all expressed during normal human kidney development stages. In DYS, the expression of EDA2R was higher than in normal kidneys, likely due to EDA2R's role in apoptosis, which was upregulated in specific cases and could possibly contribute to the formation of DYS. The expression of PCDH9 was lower in HK, which can be attributed to the possible role of PCDH9 in cell migration suppression. Decreased PCDH9 expression is linked to increased cell migration, potentially contributing to the development of HK. The level of TRAF7 expression was reduced in all examined kidney disorders compared to normal kidneys, suggesting that this reduction might be attributed to the crucial role of TRAF7 in the formation of endothelium and ciliogenesis, both of which are essential for normal kidney development. Further research is required to ascertain the function of these proteins in both the typical development of the kidney and in CAKUT.
Collapse
Affiliation(s)
- Jelena Kelam
- Department of Family Medicine, Split-Dalmatia County Health Center, 21000 Split, Croatia; (J.K.); (L.K.)
| | - Nela Kelam
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, 21000 Split, Croatia; (N.K.); (N.F.); (A.R.); (D.K.); (S.K.)
| | - Natalija Filipović
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, 21000 Split, Croatia; (N.K.); (N.F.); (A.R.); (D.K.); (S.K.)
| | - Luka Komić
- Department of Family Medicine, Split-Dalmatia County Health Center, 21000 Split, Croatia; (J.K.); (L.K.)
| | - Anita Racetin
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, 21000 Split, Croatia; (N.K.); (N.F.); (A.R.); (D.K.); (S.K.)
| | - Dora Komić
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, 21000 Split, Croatia; (N.K.); (N.F.); (A.R.); (D.K.); (S.K.)
| | - Sandra Kostić
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, 21000 Split, Croatia; (N.K.); (N.F.); (A.R.); (D.K.); (S.K.)
| | - Ivana Kuzmić Prusac
- Department of Pathology, University Hospital Center Split, 21000 Split, Croatia;
| | - Katarina Vukojević
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, 21000 Split, Croatia; (N.K.); (N.F.); (A.R.); (D.K.); (S.K.)
- Department of Anatomy, School of Medicine, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
- Center for Translational Research in Biomedicine, School of Medicine, University of Split, 21000 Split, Croatia
| |
Collapse
|
3
|
Nowak KL, Edelstein CL. Apoptosis and autophagy in polycystic kidney disease (PKD). Cell Signal 2019; 68:109518. [PMID: 31881325 DOI: 10.1016/j.cellsig.2019.109518] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/20/2019] [Accepted: 12/21/2019] [Indexed: 02/08/2023]
Abstract
Apoptosis in the cystic epithelium is observed in most rodent models of polycystic kidney disease (PKD) and in human autosomal dominant PKD (ADPKD). Apoptosis inhibition decreases cyst growth, whereas induction of apoptosis in the kidney of Bcl-2 deficient mice increases proliferation of the tubular epithelium and subsequent cyst formation. However, alternative evidence indicates that both induction of apoptosis as well as increased overall rates of apoptosis are associated with decreased cyst growth. Autophagic flux is suppressed in cell, zebra fish and mouse models of PKD and suppressed autophagy is known to be associated with increased apoptosis. There may be a link between apoptosis and autophagy in PKD. The mammalian target of rapamycin (mTOR), B-cell lymphoma 2 (Bcl-2) and caspase pathways that are known to be dysregulated in PKD, are also known to regulate both autophagy and apoptosis. Induction of autophagy in cell and zebrafish models of PKD results in suppression of apoptosis and reduced cyst growth supporting the hypothesis autophagy induction may have a therapeutic role in decreasing cyst growth, perhaps by decreasing apoptosis and proliferation in PKD. Future research is needed to evaluate the effects of direct autophagy inducers on apoptosis in rodent PKD models, as well as the cause and effect relationship between autophagy, apoptosis and cyst growth in PKD.
Collapse
Affiliation(s)
- Kristen L Nowak
- Division of Renal Diseases and Hypertension, Univ. of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Charles L Edelstein
- Division of Renal Diseases and Hypertension, Univ. of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
4
|
Woolf AS. Growing a new human kidney. Kidney Int 2019; 96:871-882. [PMID: 31399199 PMCID: PMC6856720 DOI: 10.1016/j.kint.2019.04.040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 03/01/2019] [Accepted: 04/01/2019] [Indexed: 12/17/2022]
Abstract
There are 3 reasons to generate a new human kidney. The first is to learn more about the biology of the developing and mature organ. The second is to generate tissues with which to model congenital and acquired kidney diseases. In particular, growing human kidneys in this manner ultimately should help us understand the mechanisms of common chronic kidney diseases such as diabetic nephropathy and others featuring fibrosis, as well as nephrotoxicity. The third reason is to provide functional kidney tissues that can be used directly in regenerative medicine therapies. The second and third reasons to grow new human kidneys are especially compelling given the millions of persons worldwide whose lives depend on a functioning kidney transplant or long-term dialysis, as well as those with end-stage renal disease who die prematurely because they are unable to access these treatments. As shown in this review, the aim to create healthy human kidney tissues has been partially realized. Moreover, the technology shows promise in terms of modeling genetic disease. In contrast, barely the first steps have been taken toward modeling nongenetic chronic kidney diseases or using newly grown human kidney tissue for regenerative medicine therapies.
Collapse
Affiliation(s)
- Adrian S Woolf
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, United Kingdom; Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom.
| |
Collapse
|
5
|
Kimber SJ, Woolf AS. From human pluripotent stem cells to functional kidney organoids and models of renal disease. Stem Cell Investig 2018; 5:20. [PMID: 30148153 DOI: 10.21037/sci.2018.07.02] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 07/09/2018] [Indexed: 01/16/2023]
Affiliation(s)
- Susan J Kimber
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - Adrian S Woolf
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK.,Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
6
|
Frazier KS. Species Differences in Renal Development and Associated Developmental Nephrotoxicity. Birth Defects Res 2017; 109:1243-1256. [DOI: 10.1002/bdr2.1088] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 06/14/2017] [Accepted: 06/20/2017] [Indexed: 12/19/2022]
|
7
|
Sarı F, Yalçın AD, Genç GE, Sarıkaya M, Bisgin A, Çetinkaya R, Gümüşlü S. Autosomal Dominant Polycystic Disease is Associated with Depressed Levels of Soluble Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand. Balkan Med J 2016; 33:512-516. [PMID: 27761278 PMCID: PMC5056653 DOI: 10.5152/balkanmedj.2016.150685] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 05/07/2016] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Autosomal dominant polycystic kidney disease (ADPKD) is characterized by multiple, large renal cysts and impaired kidney function. Although the reason for the development of kidney cysts is unknown, ADPKD is associated with cell cycle arrest and abundant apoptosis of renal tubular epithelial cells. AIMS We asked whether serum-soluble TNF-related apoptosis-inducing ligand (sTRAIL) might underlie ADPKD. STUDY DESIGN Case-control study. METHODS Serum sTRAIL levels were measured in 44 patients with ADPKD and 18 healthy volunteers. The human soluble TRAIL/Apo2L ELISA kit was used for the in vitro quantitative determination of sTRAIL in serum samples. RESULTS Mean serum sTRAIL levels were lower in patients with ADPKD as compared to the control group (446.9±103.1 and 875.9±349.6 pg/mL, p<0.001). Serum sTRAIL levels did not differ among stages of renal failure in patients with ADPKD. There was no correlation between serum sTRAIL levels and estimated glomerular filtration rate in patients with ADPKD (p>0.05). CONCLUSION Our results show that ADPKD patients have depressed sTRAIL levels, indicating apoptosis unrelated to the stage of chronic renal failure.
Collapse
Affiliation(s)
- Funda Sarı
- Division of Nephrology, Department of Internal Medicine, Antalya Training and Research Hospital, Antalya, Turkey
| | - Arzu Didem Yalçın
- Department of Internal Medicine, Antalya Training and Research Hospital, Antalya, Turkey
| | - Gizem Esra Genç
- Department of Biochemistry, Akdeniz University School of Medicine, Antalya, Turkey
| | - Metin Sarıkaya
- Division of Nephrology, Department of Internal Medicine, Antalya Training and Research Hospital, Antalya, Turkey
| | - Atıl Bisgin
- Department of Clinical and Experimental Medicine, University of Linköping School of Health Sciences, Linköping, Sweden
| | - Ramazan Çetinkaya
- Division of Nephrology, Department of Internal Medicine, Antalya Training and Research Hospital, Antalya, Turkey
| | - Saadet Gümüşlü
- Department of Biochemistry, Akdeniz University School of Medicine, Antalya, Turkey
| |
Collapse
|
8
|
He X, Xie Z, Dong Q, Chen P, Hu J, Wang T. Apoptosis in the kidneys of rats that experienced intrauterine growth restriction. Nephrology (Carlton) 2014; 20:34-9. [PMID: 25243775 DOI: 10.1111/nep.12340] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2014] [Indexed: 01/21/2023]
Affiliation(s)
- Xiaori He
- Department of Neonatology; Second Xiangya Hospital; Central South University; Changsha Hunan China
| | - Zongde Xie
- Department of Neonatology; Second Xiangya Hospital; Central South University; Changsha Hunan China
| | - Qingyi Dong
- Department of Neonatology; Second Xiangya Hospital; Central South University; Changsha Hunan China
| | - Pingyang Chen
- Department of Neonatology; Second Xiangya Hospital; Central South University; Changsha Hunan China
| | - Jingtao Hu
- Department of Neonatology; Second Xiangya Hospital; Central South University; Changsha Hunan China
| | - Tao Wang
- Department of Neonatology; Second Xiangya Hospital; Central South University; Changsha Hunan China
| |
Collapse
|
9
|
Liu J, Qin J, Mei W, Zhang H, Yuan Q, Peng Z, Luo R, Yuan X, Huang L, Tao L. Expression of Niban in renal interstitial fibrosis. Nephrology (Carlton) 2014; 19:479-89. [PMID: 24750539 DOI: 10.1111/nep.12266] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2014] [Indexed: 12/30/2022]
Affiliation(s)
- Jishi Liu
- Division of Nephrology; Xiangya Hospital; Central South University; Changsha China
| | - Jiao Qin
- Division of Nephrology; Xiangya Hospital; Central South University; Changsha China
| | - Wenjuan Mei
- Division of Nephrology; Xiangya Hospital; Central South University; Changsha China
| | - Hao Zhang
- Division of Nephrology; The Third Xiangya Hospital; Central South University; Changsha China
| | - Qiongjing Yuan
- Division of Nephrology; Xiangya Hospital; Central South University; Changsha China
| | - Zhangzhe Peng
- Division of Nephrology; Xiangya Hospital; Central South University; Changsha China
| | - Renna Luo
- Division of Nephrology; Xiangya Hospital; Central South University; Changsha China
| | - Xiangning Yuan
- Division of Nephrology; Xiangya Hospital; Central South University; Changsha China
| | - Ling Huang
- Division of Nephrology; Xiangya Hospital; Central South University; Changsha China
| | - Lijian Tao
- Division of Nephrology; Xiangya Hospital; Central South University; Changsha China
- State Key Laboratory of Medical Genetics of China; Central South University; Changsha China
| |
Collapse
|
10
|
Experimental renal progenitor cells: repairing and recreating kidneys? Pediatr Nephrol 2014; 29:665-72. [PMID: 24221350 DOI: 10.1007/s00467-013-2667-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 10/14/2013] [Accepted: 10/15/2013] [Indexed: 01/07/2023]
Abstract
Strategies to facilitate repair or generate new nephrons are exciting prospects for acute and chronic human renal disease. Repair of kidney injury involves not just local mechanisms but also mobilisation of progenitor/stem cells from intrarenal niches, including papillary, tubular and glomerular locations. Diverse markers characterise these unique cells, often including CD24 and CD133. Extrarenal stem cells may also contribute to repair, with proposed roles in secreting growth factors, transfer of microvesicles and exosomes and immune modulation. Creating new nephrons from stem cells is beginning to look feasible in mice in which kidneys can be dissociated into single cells and will then generate mature renal structures when recombined. The next step is to identify the correct human markers for progenitor cells from the fetus or mature kidney with similar potential to form new kidneys. Intriguingly, development can continue in vivo: whole foetal kidneys and recombined organs engraft, develop a blood supply and grow when xenotransplanted, and there are new advances in decellularised scaffolds to promote differentiation. This is an exciting time for human kidney repair and regeneration. Many of the approaches and techniques are in their infancy and based on animal rather than human work, but there is a rapid pace of discovery, and we predict that therapies based on advances in this field will come into clinical practice in the next decade.
Collapse
|
11
|
Anders C, Ashton N, Ranjzad P, Dilworth MR, Woolf AS. Ex vivo modeling of chemical synergy in prenatal kidney cystogenesis. PLoS One 2013; 8:e57797. [PMID: 23554868 PMCID: PMC3595278 DOI: 10.1371/journal.pone.0057797] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 01/25/2013] [Indexed: 01/08/2023] Open
Abstract
Cyclic adenosine monophosphate (cAMP) drives genetic polycystic kidney disease (PKD) cystogenesis. Yet within certain PKD families, striking differences in disease severity exist between affected individuals, and genomic and/or environmental modifying factors have been evoked to explain these observations. We hypothesized that PKD cystogenesis is accentuated by an aberrant fetal milieu, specifically by glucocorticoids. The extent and nature of cystogenesis was assessed in explanted wild-type mouse embryonic metanephroi, using 8-Br-cAMP as a chemical to mimic genetic PKD and the glucocorticoid dexamethasone as the environmental modulator. Cysts and glomeruli were quantified by an observer blinded to culture conditions, and tubules were phenotyped using specific markers. Dexamethasone or 8-Br-cAMP applied on their own produced cysts predominantly arising in proximal tubules and descending limbs of loops of Henle. When applied together, however, dexamethasone over a wide concentration range synergized with 8-Br-cAMP to generate a more severe, glomerulocystic, phenotype; we note that prominent glomerular cysts have been reported in autosomal dominant PKD fetal kidneys. Our data support the idea that an adverse antenatal environment exacerbates renal cystogenesis.
Collapse
Affiliation(s)
- Corina Anders
- Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Manchester Academic Health Science Centre and St Mary's and Manchester Children's Hospital, Manchester, United Kingdom.
| | | | | | | | | |
Collapse
|
12
|
Mekahli D, Parys JB, Bultynck G, Missiaen L, De Smedt H. Polycystins and cellular Ca2+ signaling. Cell Mol Life Sci 2012; 70:2697-712. [PMID: 23076254 PMCID: PMC3708286 DOI: 10.1007/s00018-012-1188-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 10/01/2012] [Accepted: 10/02/2012] [Indexed: 12/19/2022]
Abstract
The cystic phenotype in autosomal dominant polycystic kidney disease is characterized by a profound dysfunction of many cellular signaling patterns, ultimately leading to an increase in both cell proliferation and apoptotic cell death. Disturbance of normal cellular Ca2+ signaling seems to be a primary event and is clearly involved in many pathways that may lead to both types of cellular responses. In this review, we summarize the current knowledge about the molecular and functional interactions between polycystins and multiple components of the cellular Ca2+-signaling machinery. In addition, we discuss the relevant downstream responses of the changed Ca2+ signaling that ultimately lead to increased proliferation and increased apoptosis as observed in many cystic cell types.
Collapse
Affiliation(s)
- D. Mekahli
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven, Campus Gasthuisberg O/N-I, B-802, Herestraat 49, 3000 Leuven, Belgium
| | - Jan B. Parys
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven, Campus Gasthuisberg O/N-I, B-802, Herestraat 49, 3000 Leuven, Belgium
| | - G. Bultynck
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven, Campus Gasthuisberg O/N-I, B-802, Herestraat 49, 3000 Leuven, Belgium
| | - L. Missiaen
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven, Campus Gasthuisberg O/N-I, B-802, Herestraat 49, 3000 Leuven, Belgium
| | - H. De Smedt
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven, Campus Gasthuisberg O/N-I, B-802, Herestraat 49, 3000 Leuven, Belgium
| |
Collapse
|
13
|
Majumdar K, Chakrabarti I, Giri A. Pediatric hydronephrotic segmental renal dysplasia with ipsilateral ureterovesical obstruction--rare coincidence or a consequence? Fetal Pediatr Pathol 2012; 31:202-9. [PMID: 22414265 DOI: 10.3109/15513815.2011.648721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Obstruction in the developing urinary tract during embryonic life is one of the factors promoting disordered metanephric development in renal dysplasia. Dysplastic kidneys usually undergo involution during the first 5 years of life. Herein, we narrate a rare case of hydronephrotic segmental dysplasia co-existing with ipsilateral ureterovesical obstruction, in a 7-year-old male child, masquerading as hydronephrosis. Etiopathogenesis of segmental dysplasia presenting at this age is not clearly understood, and could be a consequence of intrauterine obstructive uropathy. Histologic evidence of dysplasia in a hydronephrotic kidney should warrant a close follow-up for pathologic changes in the contralateral kidney or urinary tract.
Collapse
|
14
|
Abstract
Congenital anomalies of the kidney and urinary tract anatomy (CAKUT) are common in children and represent approximately 30% of all prenatally diagnosed malformations. CAKUT is phenotypically variable and can affect the kidney(s) alone and/or the lower urinary tract. The spectrum includes more common anomalies such as vesicoureteral reflux and, rarely, more severe malformations such as bilateral renal agenesis. In young children, congenital anomalies are the leading cause of kidney failure and for kidney transplantation or dialysis. CAKUT can also lead to significant renal problems in adulthood and may present itself with hypertension and/or proteinuria. Congenital renal anomalies can be sporadic or familial, syndromic (also affecting nonrenal or non-urinary tract tissues), or nonsyndromic. Genetic causes have been identified for the syndromic forms and have shed some light into the molecular mechanisms of kidney development in human beings. The genetic causes for the more common nonsyndromic forms of CAKUT are unknown. The role of prenatal interventions and postnatal therapies as well as the benefits of screening affected individuals and their family members are not clear.
Collapse
Affiliation(s)
- Hakan R Toka
- Renal Division, Brigham and Women's Hospital, Boston, MA 02115, USA.
| | | | | | | |
Collapse
|
15
|
Goilav B. Apoptosis in polycystic kidney disease. Biochim Biophys Acta Mol Basis Dis 2011; 1812:1272-80. [PMID: 21241798 DOI: 10.1016/j.bbadis.2011.01.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Revised: 01/05/2011] [Accepted: 01/07/2011] [Indexed: 12/20/2022]
Abstract
Apoptosis is the process of programmed cell death. It is a ubiquitous, controlled process consuming cellular energy and designed to avoid cytokine release despite activation of local immune cells, which clear the cell fragments. The process occurs during organ development and in maintenance of homeostasis. Abnormalities in any step of the apoptotic process are associated with autoimmune diseases and malignancies. Polycystic kidney disease (PKD) is the most common inherited kidney disease leading to end-stage renal disease (ESRD). Cyst formation requires multiple mechanisms and apoptosis is considered one of them. Abnormalities in apoptotic processes have been described in various murine and rodent models of PKD as well as in human PKD kidneys. The purpose of this review is to outline the role of apoptosis in progression of PKD as well as to describe the mechanisms involved. This article is part of a Special Issue entitled: Polycystic Kidney Disease.
Collapse
|
16
|
Fujita H, Hida M, Kanemoto K, Fukuda K, Nagata M, Awazu M. Cyclic stretch induces proliferation and TGF-β1-mediated apoptosis via p38 and ERK in ureteric bud cells. Am J Physiol Renal Physiol 2010; 299:F648-55. [DOI: 10.1152/ajprenal.00402.2009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We previously reported that p38 mitogen-activated protein kinase (p38) and phosphorylated ERK are upregulated in cyst epithelium of human renal dysplasia and obstructive uropathy in fetal lambs (Omori S, Fukuzawa R, Hida M, Awazu M. Kidney Int 61: 899–906, 2002; Omori S, Kitagawa H, Koike J, Fujita H, Hida M, Pringle KC, Awazu M. Kidney Int 73: 1031–1037, 2008). Dysplastic epithelium is characterized by proliferation, apoptosis, and upregulation of Pax2 and transforming growth factor (TGF)-β1. In the present study, we investigated whether cyclic mechanical stretching of ureteric bud cells, a mimic of the hydrodynamic derangement after fetal urinary tract obstruction, reproduces events seen in vivo. Cyclic stretch activated p38 and ERK and upregulated Pax2 expression in a time-dependent manner in ureteric bud cells. Stretch-stimulated Pax2 expression was suppressed by a p38 inhibitor, SB203580, or a MEK inhibitor, PD98059. 5-Deoxyuridine incorporation was increased by stretch at 24 h, which was also abolished by SB203580 or PD98059. On the other hand, apoptosis was not induced at 24 h by stretch but was significantly increased at 48 h. TGF-β1 secretion was increased by stretch at 24 h, which was inhibited by SB203580 or PD98059. Inhibition of p38 or ERK as well as anti-TGF-β antibody abolished the stretch-induced apoptosis. Finally, exogenous TGF-β1 induced apoptosis of ureteric bud cells, which was inhibited by SB203580 and PD98059. In conclusion, cyclic stretch induces Pax2 upregulation, proliferation, and TGF-β1-mediated apoptosis, features characteristic of dysplastic epithelium, via p38 and ERK in ureteric bud cells.
Collapse
Affiliation(s)
| | | | - Katsuyoshi Kanemoto
- Department of Molecular Pathology, Institute of Basic Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Keiichi Fukuda
- Regenerative Medicine and Advanced Cardiac Therapeutics, Keio University School of Medicine, Tokyo; and
| | - Michio Nagata
- Department of Molecular Pathology, Institute of Basic Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | | |
Collapse
|
17
|
Expression of transforming growth factor-beta1 limits renal ischemia-reperfusion injury. Transplantation 2010; 89:1320-7. [PMID: 20458271 DOI: 10.1097/tp.0b013e3181d8e9dc] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Renal ischemia-reperfusion injury (IRI) largely contributes to kidney transplant dysfunction and acute kidney injury, but its pathogenesis is not fully understood. In this study, the role of transforming growth factor (TGF)-beta1 in renal IRI is investigated using TGF-beta1 deficient mice. METHOD Human renal tubular epithelial cells (TEC) line (HK-2) was used as an in vitro model, and cell apoptosis was determined by flow cytometric analysis. Renal IRI was induced in mice by clamping renal vein and artery for 45 min at 32 degrees C. RESULTS Here, we showed that in cultures of HK-2 cells, TGF-beta1 expression was up-regulated by tumor necrosis factor (TNF)-alpha. Neutralization of TGF-beta1 activity increased both spontaneous and TNF-alpha-mediated apoptosis, and knockdown of TGF-beta1 expression increased the sensitivity of cell apoptosis to TNF-alpha. In a mouse model of renal IRI, a deficiency in TGF-beta1 expression increased the severity of renal injury, as indicated by more severe renal tubular damage, higher levels of serum creatinine or blood urea nitrogen in TGF-beta1 deficient mice as compared with those in wild-type controls. Further experiments showed that the antiapoptosis of TGF-beta1 correlated with up-regulation of Bcl-2 in kidney cells. CONCLUSION Expression of TGF-beta1 in TECs, potentially induced by proinflammatory TNF-alpha, renders TECs resistance to cell death. In mice, TGF-beta1 deficiency results in more prone to IRI. These data imply that TGF-beta1 may act as a feedback survival factor in the resistance to kidney injury and maintenance of epithelium homeostasis.
Collapse
|
18
|
Mechanisms of renal injury and progression of renal disease in congenital obstructive nephropathy. Pediatr Nephrol 2010; 25:687-97. [PMID: 19844747 DOI: 10.1007/s00467-009-1316-5] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2009] [Revised: 07/27/2009] [Accepted: 07/28/2009] [Indexed: 12/21/2022]
Abstract
Congenital obstructive nephropathy accounts for the greatest fraction of chronic kidney disease in children. Genetic and nongenetic factors responsible for the lesions are largely unidentified, and attention has been focused on minimizing obstructive renal injury and optimizing long-term outcomes. The cellular and molecular events responsible for obstructive injury to the developing kidney have been elucidated from animal models. These have revealed nephron loss through cellular phenotypic transition and cell death, leading to the formation of atubular glomeruli and tubular atrophy. Altered renal expression of growth factors and cytokines, including angiotensin, transforming growth factor-beta, and adhesion molecules, modulate cell death by apoptosis or phenotypic transition of glomerular, tubular, and vascular cells. Mediators of cellular injury include hypoxia, ischemia, and reactive oxygen species, while fibroblasts undergo myofibroblast transformation with increased deposition of extracellular matrix. Progression of the lesions involves interstitial inflammation and interstitial fibrosis, both of which impair growth of the obstructed kidney and result in compensatory growth of the contralateral kidney. The long-term outcome depends on timing and severity of the obstruction and its relief, minimizing ongoing injury, and enhancing remodeling. Advances will depend on new biomarkers to evaluate the severity of obstruction, to determine therapy, and to follow the evolution of lesions.
Collapse
|
19
|
Adalat S, Bockenhauer D, Ledermann SE, Hennekam RC, Woolf AS. Renal malformations associated with mutations of developmental genes: messages from the clinic. Pediatr Nephrol 2010; 25:2247-55. [PMID: 20603712 PMCID: PMC2937138 DOI: 10.1007/s00467-010-1578-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Revised: 05/07/2010] [Accepted: 05/27/2010] [Indexed: 01/30/2023]
Abstract
Renal tract malformations (RTMs) account for about 40% of children with end-stage renal failure. RTMs can be caused by mutations of genes normally active in the developing kidney and lower renal tract. Moreover, some RTMs occur in the context of multi-organ malformation syndromes. For these reasons, and because genetic testing is becoming more widely available, pediatric nephrologists should work closely with clinical geneticists to make genetic diagnoses in children with RTMs, followed by appropriate family counseling. Here we highlight families with renal cysts and diabetes, renal coloboma and Fraser syndromes, and a child with microdeletion of chromosome 19q who had a rare combination of malformations. Such diagnoses provide families with often long-sought answers to the question "why was our child born with kidney disease". Precise genetic diagnoses will also help to define cohorts of children with RTMs for long-term clinical outcome studies.
Collapse
Affiliation(s)
- Shazia Adalat
- UCL Institute of Child Health and Great Ormond Street Hospital NHS Trust, London, UK
| | - Detlef Bockenhauer
- UCL Institute of Child Health and Great Ormond Street Hospital NHS Trust, London, UK
| | - Sarah E. Ledermann
- UCL Institute of Child Health and Great Ormond Street Hospital NHS Trust, London, UK
| | - Raoul C. Hennekam
- Department of Pediatrics, University of Amsterdam, Amsterdam, The Netherlands
| | - Adrian S. Woolf
- University of Manchester and Manchester Children’s Hospital, Manchester, England ,Developmental and Regenerative Medicine Research Group, University of Manchester, Oxford Road, Manchester, M13 9PT UK
| |
Collapse
|
20
|
Sparrow DB, Boyle SC, Sams RS, Mazuruk B, Zhang L, Moeckel GW, Dunwoodie SL, de Caestecker MP. Placental insufficiency associated with loss of Cited1 causes renal medullary dysplasia. J Am Soc Nephrol 2009; 20:777-86. [PMID: 19297558 DOI: 10.1681/asn.2008050547] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
A number of studies have shown that placental insufficiency affects embryonic patterning of the kidney and leads to a decreased number of functioning nephrons in adulthood; however, there is circumstantial evidence that placental insufficiency may also affect renal medullary growth, which could account for cases of unexplained renal medullary dysplasia and for abnormalities in renal function among infants who had experienced intrauterine growth retardation. We observed that mice with late gestational placental insufficiency associated with genetic loss of Cited1 expression in the placenta had renal medullary dysplasia. This was not caused by lower urinary tract obstruction or by defects in branching of the ureteric bud during early nephrogenesis but was associated with decreased tissue oxygenation and increased apoptosis in the expanding renal medulla. Loss of placental Cited1 was required for Cited1 mutants to develop renal dysplasia, and this was not dependent on alterations in embryonic Cited1 expression. Taken together, these findings suggest that renal medullary dysplasia in Cited1 mutant mice is a direct consequence of decreased tissue oxygenation resulting from placental insufficiency.
Collapse
Affiliation(s)
- Duncan B Sparrow
- Developmental Biology Division, Victor Chang Cardiac Research Institute, University of New South Wales, Sydney, Australia
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Edelstein CL. Mammalian target of rapamycin and caspase inhibitors in polycystic kidney disease. Clin J Am Soc Nephrol 2008; 3:1219-26. [PMID: 18587045 DOI: 10.2215/cjn.05611207] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
One of the most important abnormalities of the tubular epithelial cells lining the cysts as well as noncystic tubular epithelium is a disturbance in the balance between tubular cell proliferation and apoptosis. Activation of the mammalian target of rapamycin signaling pathway results in increased cell proliferation. Recent studies suggested abnormalities of the mammalian target of rapamycin signaling pathway in polycystic kidney disease. Mammalian target of rapamycin inhibition with sirolimus or everolimus results in attenuation of cyst formation in rat and mouse models of polycystic kidney disease. Apoptosis is a pathologic feature of most models of polycystic kidney disease, including human polycystic kidneys. Caspases, the major mediators of apoptosis, are increased in polycystic kidney disease kidneys. Both in vitro and in vivo studies suggest that caspase or apoptosis inhibition attenuates cyst formation. This review focuses on mammalian target of rapamycin and apoptosis signaling pathways in polycystic kidney disease and the role of mammalian target of rapamycin inhibitors and apoptosis inhibitors as potential therapies to reduce cyst formation.
Collapse
Affiliation(s)
- Charles L Edelstein
- Division of Renal Diseases and Hypertension, University of Colorado School of Medicine, Box C281, 4200 E. 9th Avenue, Denver, CO 80262, USA.
| |
Collapse
|
22
|
Mure PY, Mouriquand P. Upper urinary tract dilatation: prenatal diagnosis, management and outcome. Semin Fetal Neonatal Med 2008; 13:152-63. [PMID: 17988967 DOI: 10.1016/j.siny.2007.09.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Upper urinary tract dilatation is one of the most common abnormalities detected on prenatal ultrasound scanning. It is commonly due to transient urine flow impairment (UFI) at the level of the pelvi-ureteric and vesico-ureteric junctions, which improves with time in most cases. It is usually in the neonatal period that the diagnosis is confirmed and during the first 18 months of life that the prognosis of the dilatation is defined.
Collapse
Affiliation(s)
- Pierre-Yves Mure
- Department of Paediatric Urology, Claude Bernard University Lyon I, Hôpital Debrousse, 29 Rue Soeur Bouvier, Lyon cedex 05, France.
| | | |
Collapse
|
23
|
Abstract
Dysplastic kidneys are common malformations affecting up to 1 in 1000 of the general population. They are part of the spectrum of Congenital Abnormalities of the Kidney and Urinary Tract (CAKUT) and an increasing number of children are being diagnosed on antenatal ultrasound. In the past, these patients may not have been detected until adulthood following investigation for other illness, or even as incidental findings at post mortem, unless there was severe bilateral dysplasia leading to Potter's sequence or renal failure in childhood. Excluding syndromic cases with defects in other organ systems, features linked to worse prognosis at presentation are: (1) bilateral disease; (2) decreased functional renal mass (which encompasses not just small kidneys but also large ones where cysts replace normal architecture); (3) lower urinary tract obstruction; and (4) anhydramnios or severe oligohydramnios. Dysplasia and renal function are dynamic and can evolve during pregnancy, so repeated assessment is necessary when pathology is expected. Worsening dimensions or decreasing amniotic fluid levels imply poorer prognosis, but there are no proven therapies during pregnancy, though vesicoamniotic shunting may be indicated with obstruction. Postnatal investigations aim to define the anatomy, which helps to estimate risks of infection and kidney function. Management might then involve observation, prophylactic antibiotics, surgery and/or renal support. Risks of renal malignancy and hypertension are low during childhood, but longer-term follow-up is needed, particularly to determine blood pressure and renal function in adulthood and pregnancy. Around 10% of cases have a family history of significant renal/urinary tract malformation. Monogenic causes include mutations in individual genes, such as TCF2/hepatocyte nuclear factor 1ss (HNF1beta), PAX2 and uroplakins, but there are also recent reports of children with compound heterozygote mutations in several renal/urinary tract developmental genes. Effective genetic screening in future may require gene chip or other techniques to assess multiple genes concurrently, but this should not replace a multidisciplinary approach to these often difficult cases.
Collapse
Affiliation(s)
- Paul Winyard
- Institute of Child Health, University College London, 30 Guilford Street, London, UK.
| | | |
Collapse
|
24
|
Bridgewater D, Cox B, Cain J, Lau A, Athaide V, Gill PS, Kuure S, Sainio K, Rosenblum ND. Canonical WNT/β-catenin signaling is required for ureteric branching. Dev Biol 2008; 317:83-94. [DOI: 10.1016/j.ydbio.2008.02.010] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2007] [Revised: 01/30/2008] [Accepted: 02/05/2008] [Indexed: 12/23/2022]
|
25
|
Renal tract malformations: perspectives for nephrologists. ACTA ACUST UNITED AC 2008; 4:312-25. [DOI: 10.1038/ncpneph0807] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2007] [Accepted: 03/05/2008] [Indexed: 01/13/2023]
|
26
|
Tao Y, Zafar I, Kim J, Schrier RW, Edelstein CL. Caspase-3 gene deletion prolongs survival in polycystic kidney disease. J Am Soc Nephrol 2008; 19:749-55. [PMID: 18272845 DOI: 10.1681/asn.2006121378] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Affiliation(s)
- Yunxia Tao
- Division of Renal Diseases and Hypertension, University of Colorado School of Medicine, Box C281, 4200 E. 9th Avenue, Denver, CO 80262, USA
| | | | | | | | | |
Collapse
|
27
|
Activated extracellular signal-regulated kinase correlates with cyst formation and transforming growth factor-beta expression in fetal obstructive uropathy. Kidney Int 2008; 73:1031-7. [PMID: 18272960 DOI: 10.1038/ki.2008.3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Human renal dysplasia is frequently associated with urinary tract obstruction and the abnormal expression of mitogen-activated protein kinase (MAPK). Here, we determined the renal responses and MAPK expression in developing kidneys that were obstructed in fetal lambs. Kidneys were harvested at various times after obstruction (gestation day 60) through normal term (day 145). Dilation of Bowman's capsule and proximal tubules was seen 2 days after obstruction and involved the whole cortex 18 days later, with numerous cysts present throughout the kidney at term. The proliferation marker Ki-67 and transforming growth factor-beta (TGF-beta) were detected 2 days after obstruction and progressively increased in tubules, cysts, and the interstitium. In control kidneys, p38 was expressed in tubules only during the fetal stage, whereas phosphorylated extracellular signal-regulated kinase (P-ERK) was limited to ureteric buds and collecting ducts at all stages examined. However, Jun-N-terminal kinase (JNK) was absent in the fetal kidney but present in tubules at term. In obstructed kidneys, cyst epithelia were positive for p38 and P-ERK but negative for JNK throughout all stages. These studies show that P-ERK correlated spatially and temporally with Ki-67 and TGF-beta expression, which suggests that ERK may contribute to cyst formation and fibrosis in the obstructed fetal kidney.
Collapse
|
28
|
Masoumi A, Reed-Gitomer B, Kelleher C, Schrier RW. Potential pharmacological interventions in polycystic kidney disease. Drugs 2008; 67:2495-510. [PMID: 18034588 DOI: 10.2165/00003495-200767170-00004] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Polycystic kidney diseases (autosomal dominant and autosomal recessive) are progressive renal tubular cystic diseases, which are characterised by cyst expansion and loss of normal kidney structure and function. Autosomal dominant polycystic kidney disease (ADPKD) is the most common life- threatening, hereditary disease. ADPKD is more prevalent than Huntington's disease, haemophilia, sickle cell disease, cystic fibrosis, myotonic dystrophy and Down's syndrome combined. Early diagnosis and treatment of hypertension with inhibitors of the renin-angiotensin-aldosterone system (RAAS) and its potential protective effect on left ventricular hypertrophy has been one of the major therapeutic goals to decrease cardiac complications and contribute to improved prognosis of the disease. Advances in the understanding of the genetics, molecular biology and pathophysiology of the disease are likely to facilitate the improvement of treatments for these diseases. Developments in describing the role of intracellular calcium ([Ca(2+)](i)) and its correlation with cellular signalling systems, Ras/Raf/mitogen extracellular kinase (MEK)/extracellular signal-regulated protein kinase (ERK), and interaction of these pathways with cyclic adenosine monophosphate (cAMP) levels, provide new insights on treatment strategies. Blocking the vasopressin V(2) receptor, a major adenylyl cyclase agonist, demonstrated significant improvements in inhibiting cytogenesis in animal models. Because of activation of the mammalian target of rapamycin (mTOR) pathway, the use of sirolimus (rapamycin) an mTOR inhibitor, markedly reduced cyst formation and decreased polycystic kidney size in several animal models. Caspase inhibitors have been shown to decrease cytogenesis and renal failure in rats with cystic disease. Cystic fluid secretion results in cyst enlargement and somatostatin analogues have been shown to decrease renal cyst progression in patients with ADPKD. The safety and efficacy of these classes of drugs provide potential interventions for experimental and clinical trials.
Collapse
Affiliation(s)
- Amirali Masoumi
- Department of Medicine, Health Sciences Center, University of Colorado School of Medicine, Denver, Colorado, USA
| | | | | | | |
Collapse
|
29
|
Chevalier RL, Forbes MS. Generation and Evolution of Atubular Glomeruli in the Progression of Renal Disorders. J Am Soc Nephrol 2008; 19:197-206. [DOI: 10.1681/asn.2007080862] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
|
30
|
Chronic partial ureteral obstruction and the developing kidney. Pediatr Radiol 2008; 38 Suppl 1:S35-40. [PMID: 18071697 DOI: 10.1007/s00247-007-0585-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2007] [Accepted: 07/10/2007] [Indexed: 10/22/2022]
Abstract
Although congenital urinary tract obstruction is a common disorder, its pathophysiology remains poorly understood and clinical practice is controversial. Animal models have been used to elucidate the mechanisms responsible for obstructive nephropathy, and the models reveal that renal growth and function are impaired in proportion to the severity and duration of obstruction. Ureteral obstruction in the neonatal rat or mouse leads to activation of the renin-angiotensin system, renal infiltration by macrophages, and tubular apoptosis. Nephrons are lost by glomerular sclerosis and the formation of atubular glomeruli, and progressive injury leads to tubular atrophy and interstitial fibrosis. Recovery following release of obstruction depends on the timing, severity, and duration of obstruction. Growth factors and cytokines are produced by the hydronephrotic kidney, including MCP-1 and TGF-beta1, which are excreted in urine and can serve as biomarkers of renal injury. Because MRI can be used to monitor renal morphology, blood flow, and filtration rate, its use might supplant current imaging modalities (ultrasonography and diuretic renography), which have significant drawbacks. Combined use of MRI and new urinary biomarkers should improve our understanding of human congenital obstructive nephropathy and should lead to new approaches to evaluation and management of this challenging group of patients.
Collapse
|
31
|
Polito C, La Manna A, Rambaldi PF, Valentini N, Marte A, Lama G. Long-term evolution of renal damage associated with unilateral vesicoureteral reflux. J Urol 2007; 178:1043-7; discussion 1047. [PMID: 17632145 DOI: 10.1016/j.juro.2007.05.061] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2006] [Indexed: 11/27/2022]
Abstract
PURPOSE We determined the long-term evolution of renal damage associated with vesicoureteral reflux. MATERIALS AND METHODS We retrospectively selected 74 consecutive children with unilateral primary vesicoureteral reflux, ipsilateral renal differential uptake less than 45% at dimercapto-succinic acid scintigraphy performed 4 to 6 months after urinary tract infection (60 patients) or shortly after diagnosis of vesicoureteral reflux investigated for prenatal hydronephrosis (14), and normal ultrasound and scintigraphic imaging of the contralateral nonrefluxing kidney. Average patient age at diagnosis was 3 years. The outcome was assessed via dimercapto-succinic acid scan at 5 to 24 years (mean 8.9). RESULTS In 65 patients (88%) variations of less than 5% in differential uptake were recorded. Three patients (4%) showed an increase of greater than 5% in differential uptake of the refluxing kidney. Six patients (8%) demonstrated a decrease of greater than 5%, of whom 3 had 1 and 3 had no febrile urinary tract infection during followup. A total of 18 patients had a differential uptake of 35% to 45% at the first visit, of whom 3 exhibited a decrease of 5.2% to 27% in differential uptake and had no history of febrile urinary tract infection. CONCLUSIONS In most cases differential uptake of the unilaterally refluxing affected kidney remains stable from early childhood to puberty despite the increase in body mass, which necessitates increasing renal work. In some patients a significant decrease in differential uptake may be observed even in the absence of recurrent febrile urinary tract infections. A mild decrease in differential uptake (35% to 45%) at diagnosis does not exclude the possibility of a subsequent significant decrease, even in the absence of febrile urinary tract infection.
Collapse
Affiliation(s)
- Cesare Polito
- Department of Pediatrics, Second University of Naples, Naples, Italy.
| | | | | | | | | | | |
Collapse
|
32
|
Chiu MG, Johnson TM, Woolf AS, Dahm-Vicker EM, Long DA, Guay-Woodford L, Hillman KA, Bawumia S, Venner K, Hughes RC, Poirier F, Winyard PJD. Galectin-3 associates with the primary cilium and modulates cyst growth in congenital polycystic kidney disease. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 169:1925-38. [PMID: 17148658 PMCID: PMC1762475 DOI: 10.2353/ajpath.2006.060245] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Several lines of evidence implicate the beta-galactoside-binding lectin galectin-3 in development and pathological processes in renal collecting ducts: galectin-3 is expressed in the ureteric bud/collecting duct lineage during nephrogenesis, modulates collecting duct growth/differentiation in vitro, and is expressed in human autosomal recessive polycystic kidney disease in cyst epithelia, almost all of which arise from collecting ducts. Moreover, exogenous galectin-3 restricts growth of cysts generated by Madin-Darby canine kidney collecting duct-derived cells in three-dimensional culture in collagen. Using the cpk mouse model of recessively inherited polycystic kidney disease, we observed widespread galectin-3 mRNA and protein in cyst epithelia. Exogenous galectin-3 reduced cyst formation in suspension culture, and mice-null mutant for galectin-3 had more extensive renal cysts in vivo. Galectin-3 was also detected for the first time in the centrosome/primary cilium, which has been implicated in diverse polycystic kidney disease. Cilia structure/number appeared normal in galectin-3-null mutants. Finally, paclitaxel, a therapy that retards polycystic kidney disease in cpk mice, increased extracellular galectin-3, in which the lectin could potentially interact with cilia. These data raise the possibility that galectin-3 may act as a natural brake on cystogenesis in cpk mice, perhaps via ciliary roles.
Collapse
Affiliation(s)
- Miliyun G Chiu
- Nephro-Urology Unit, UCL Institute of Child Health, 30 Guilford St., London WC1N 1EH, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Woolf AS, Hillman KA. Unilateral renal agenesis and the congenital solitary functioning kidney: developmental, genetic and clinical perspectives. BJU Int 2007; 99:17-21. [PMID: 16956352 DOI: 10.1111/j.1464-410x.2006.06504.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Adrian S Woolf
- Nephro-Urology Unit, UCL Institute of Child Health, and Centre for Nephrology, Royal Free and University College Medical School, London, UK.
| | | |
Collapse
|
34
|
Jenkins D, Woolf AS. Uroplakins: new molecular players in the biology of urinary tract malformations. Kidney Int 2006; 71:195-200. [PMID: 17183244 DOI: 10.1038/sj.ki.5002053] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The uroplakins (UPs) are a family of proteins which associate with each other and form plaques on the apical surface of the urothelium. These plaques contribute to a permeability barrier, preventing the influx of urine from the urinary tract lumen. Urinary tract malformations associated with human and mouse UP mutations, the human fetal expression patterns of UPs and experiments in Xenopus oocytes are collectively revealing new functions for the UPs, forcing us to view these proteins in a new light. Rather than simply being products of the urothelial differentiation program, they may be a group of proteins central to the process of urinary tract differentiation itself.
Collapse
Affiliation(s)
- D Jenkins
- Clinical Genetics Unit, Weatherall Institute of Molecular Medicine, University of Oxford, UK
| | | |
Collapse
|
35
|
Schreuder M, Delemarre-van de Waal H, van Wijk A. Consequences of Intrauterine Growth Restriction for the Kidney. Kidney Blood Press Res 2006; 29:108-25. [PMID: 16837795 DOI: 10.1159/000094538] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Low birth weight due to intrauterine growth restriction is associated with various diseases in adulthood, such as hypertension, cardiovascular disease, insulin resistance and end-stage renal disease. The purpose of this review is to describe the effects of intrauterine growth restriction on the kidney. Nephrogenesis requires a fine balance of many factors that can be disturbed by intrauterine growth restriction, leading to a low nephron endowment. The compensatory hyperfiltration in the remaining nephrons results in glomerular and systemic hypertension. Hyperfiltration is attributed to several factors, including the renin-angiotensin system (RAS), insulin-like growth factor (IGF-I) and nitric oxide. Data from human and animal studies are presented, and suggest a faltering IGF-I and an inhibited RAS in intrauterine growth restriction. Hyperfiltration makes the kidney more vulnerable during additional kidney disease, and is associated with glomerular damage and kidney failure in the long run. Animal studies have provided a possible therapy with blockage of the RAS at an early stage in order to prevent the compensatory glomerular hyperfiltration, but this is far from being applicable to humans. Research is needed to further unravel the effect of intrauterine growth restriction on the kidney.
Collapse
Affiliation(s)
- Michiel Schreuder
- Department of Pediatric Nephrology, VU University Medical Center, Amsterdam, The Netherlands.
| | | | | |
Collapse
|
36
|
Omori S, Hida M, Fujita H, Takahashi H, Tanimura S, Kohno M, Awazu M. Extracellular signal-regulated kinase inhibition slows disease progression in mice with polycystic kidney disease. J Am Soc Nephrol 2006; 17:1604-14. [PMID: 16641154 DOI: 10.1681/asn.2004090800] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The expression of mitogen-activated protein kinases (MAPK) in DBA/2-pcy/pcy (pcy) mice, a murine model of polycystic kidney disease was investigated. Proliferating cell nuclear antigen-positive cells were recognized in cyst epithelium from embryonic day 14.5 to 25 wk of age. Extracellular signal-regulated kinase (ERK) was expressed in the renal tubules of control and pcy mice, but stronger immunostaining was observed in cyst epithelium. Phosphorylated ERK was detected only in pcy mice and was localized predominantly in the cysts. p38 MAPK (p38) was no longer expressed after birth in controls but was detected in the cyst epithelium and in occasional tubular cells of pcy mice at all stages examined. c-Jun N-terminal kinase (JNK) was expressed in all tubular segments of controls after neonatal day 7, whereas in pcy kidneys, tubules became positive for JNK after 8 wk, and the cysts expressed little JNK. Administration of an oral MAP/ERK kinase inhibitor, PD184352, 400 mg/kg per d, to 10-wk-old pcy mice daily for the first week and then every third day for 6 additional weeks significantly decreased BP, kidney weight, serum creatinine level, and water intake and significantly increased urine osmolality. The cystic index and expression of phosphorylated ERK and ERK were significantly lower in PD184352-treated pcy mice. These results demonstrate that the expression of MAPK is dysregulated in cyst epithelium and that inhibition of ERK slowed the progression of renal disease in pcy mice.
Collapse
Affiliation(s)
- Sayu Omori
- Department of Pediatrics, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | | | | | | | | | | | | |
Collapse
|
37
|
Affiliation(s)
- A S Woolf
- Nephro-Urology Unit, Institute of Child Health, University College London, London, UK.
| |
Collapse
|
38
|
Thornhill BA, Burt LE, Chen C, Forbes MS, Chevalier RL. Variable chronic partial ureteral obstruction in the neonatal rat: a new model of ureteropelvic junction obstruction. Kidney Int 2005; 67:42-52. [PMID: 15610226 DOI: 10.1111/j.1523-1755.2005.00052.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Congenital ureteropelvic junction (UPJ) obstruction is a common developmental anomaly. To elucidate the mechanisms underlying the renal consequences of congenital UPJ obstruction, we have developed a new model of variable partial unilateral ureteral obstruction (UUO) in the neonatal rat. METHODS Rat pups were subjected to sham-operation, complete UUO, or variable partial UUO within the first day of life. After 14 or 28 days, the relative number of glomeruli, cell proliferation, tubular apoptosis, tubular atrophy, and interstitial fibrosis were quantitated in histologic sections. Glomerular filtration rate (GFR) was determined after 28 days of partial or complete UUO. RESULTS Following 70% to 75% reduction in ureteral diameter, renal growth from 14 to 28 days was reduced by 60%, and the number of glomeruli decreased by 50%. Renal pelvic diameter increased in proportion to the severity of obstruction following 14 days of partial UUO, and by 28 days, was maximally dilated regardless of the luminal diameter. Renal proliferation was increased, while tubular apoptosis, tubular atrophy, and interstitial fibrosis were less severe 14 days following partial UUO than in complete UUO. GFR was reduced by 80%, and proteinuria developed following 28 days of partial UUO. CONCLUSION Renal function is impaired by chronic ipsilateral partial UUO, which reduces the number of nephrons, and leads to progressive renal pelvic dilatation. Tubular atrophy and interstitial fibrosis develop prior to significant renal pelvic dilatation. Correlation of clinically measurable parameters with renal morphometry or imaging studies in this model may lead to new approaches to the management of congenital UPJ obstruction.
Collapse
Affiliation(s)
- Barbara A Thornhill
- Department of Pediatrics, UVa Children's Hospital, University of Virginia, Charlottesville, Virginia, USA
| | | | | | | | | |
Collapse
|
39
|
Tse HKW, Leung MBW, Woolf AS, Menke AL, Hastie ND, Gosling JA, Pang CP, Shum ASW. Implication of Wt1 in the pathogenesis of nephrogenic failure in a mouse model of retinoic acid-induced caudal regression syndrome. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 166:1295-307. [PMID: 15855632 PMCID: PMC1606386 DOI: 10.1016/s0002-9440(10)62349-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/11/2005] [Indexed: 11/18/2022]
Abstract
Renal malformations are common human birth defects that sometimes occur in the context of the caudal regression syndrome. Here, we found that exposure of pregnant mice to all-trans retinoic acid, at a time when the metanephros has yet to form, causes a failure of kidney development along with caudal regression. Maternal treatment with Am580 (retinoic acid receptor alpha agonist) also induced similar patterns of kidney maldevelopment in the fetus. In metanephroi from retinoic acid-treated pregnancies, renal mesenchyme condensed around the ureteric bud but then failed to differentiate into nephrons, instead undergoing involution by fulminant apoptosis to produce a renal agenesis phenotype. Results of whole organ cultures in serum-free medium, and also tissue recombination experiments, showed that the nephrogenic defect was intrinsic to the kidney and that it resided in the metanephric mesenchyme and not the ureteric bud. Renal mesenchyme from control embryos expressed Wilms' tumor 1 (Wt1), but this transcription factor, which is indispensable for kidney development, failed to express in metanephroi of retinoic acid-exposed embryos. Wt1 expression and organogenesis were both restored, however, when metanephroi from retinoic acid-treated pregnancies were grown in serum-containing media. Our data illuminate the pathobiology of a severe, teratogen-induced kidney malformation.
Collapse
Affiliation(s)
- Herman K W Tse
- Department of Anatomy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, China
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Tao Y, Kim J, Faubel S, Wu JC, Falk SA, Schrier RW, Edelstein CL. Caspase inhibition reduces tubular apoptosis and proliferation and slows disease progression in polycystic kidney disease. Proc Natl Acad Sci U S A 2005; 102:6954-9. [PMID: 15863619 PMCID: PMC1100753 DOI: 10.1073/pnas.0408518102] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
We have previously demonstrated an increase in proapoptotic caspase-3 in the kidney of Han:SPRD rats with polycystic kidney disease (PKD). The aim of the present study was to determine the effect of caspase inhibition on tubular cell apoptosis and proliferation, cyst formation, and renal failure in the Han:SPRD rat model of PKD. Heterozygous (Cy/+) and littermate control (+/+) male rats were weaned at 3 weeks of age and then treated with the caspase inhibitor IDN-8050 (10 mg/kg per day) by means of an Alzet (Palo Alto, CA) minipump or vehicle [polyethylene glycol (PEG 300)] for 5 weeks. The two-kidney/total body weight ratio more than doubled in Cy/+ rats compared with +/+ rats. IDN-8050 significantly reduced the kidney enlargement by 44% and the cyst volume density by 29% in Cy/+ rats. Cy/+ rats with PKD have kidney failure as indicated by a significant increase in blood urea nitrogen. IDN-8050 significantly reduced the increase in blood urea nitrogen in the Cy/+ rats. The number of proliferating cell nuclear antigen-positive tubular cells and apoptotic tubular cells in non-cystic and cystic tubules was significantly reduced in IDN-8050-treated Cy/+ rats compared with vehicle-treated Cy/+ rats. On immunoblot, the active form of caspase-3 (20 kDa) was significantly decreased in IDN-8050-treated Cy/+ rats compared with vehicle-treated Cy/+ rats. In summary, in a rat model of PKD, caspase inhibition with IDN-8050 (i) decreases apoptosis and proliferation in cystic and noncystic tubules; (ii) inhibits renal enlargement and cystogenesis, and (iii) attenuates the loss of kidney function.
Collapse
Affiliation(s)
- Yunxia Tao
- Division of Renal Diseases and Hypertension, University of Colorado Health Sciences Center, Renal Box C281, 4200 East 9th Avenue, Denver, CO 80262, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Welham SJM, Riley PR, Wade A, Hubank M, Woolf AS. Maternal diet programs embryonic kidney gene expression. Physiol Genomics 2005; 22:48-56. [PMID: 15827236 DOI: 10.1152/physiolgenomics.00167.2004] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Human epidemiological data associating birth weight with adult disease suggest that organogenesis is "programmed" by maternal diet. In rats, protein restriction in pregnancy produces offspring with fewer renal glomeruli and higher systemic blood pressures than controls. We tested the hypothesis that maternal diet alters gene expression in the metanephros, the precursor of the definitive mammalian kidney. We demonstrated that maternal low-protein diet initiated when pregnancy starts and maintained to embryonic day 13, when the metanephros consists of mesenchyme surrounding a once-branched ureteric bud, is sufficient to significantly reduce glomerular numbers in offspring by about 20%. As assessed by representational difference analyses and real-time quantitative polymerase chain reactions, low-protein diet modulated gene expression in embryonic day 13 metanephroi. In particular, levels of prox-1, the ortholog of Drosophila transcription factor prospero, and cofilin-1, a regulator of the actin cytoskeleton, were reduced. During normal metanephrogenesis, prox-1 protein was first detected in mesenchymal cells around the ureteric tree and thereafter in nascent nephron epithelia, whereas cofilin-1 immunolocalized to bud derivatives and condensing mesenchyme. Previously, we reported that low-protein diets increased mesenchymal apoptosis cells when metanephrogenesis began and thereafter reduced numbers of precursor cells. Collectively, these studies prove that the maternal diet programs the embryonic kidney, altering cell turnover and gene expression at a time when nephrons and glomeruli have yet to form. The human implication is that the maternal diet ingested between conception and 5- 6-wk gestation contributes to the variation in glomerular numbers that are known to occur between healthy and hypertensive populations.
Collapse
Affiliation(s)
- Simon J M Welham
- Nephro-Urology, Institute of Child Health, University College London, London, United Kingdom.
| | | | | | | | | |
Collapse
|
42
|
Kip SN, Hunter LW, Ren Q, Harris PC, Somlo S, Torres VE, Sieck GC, Qian Q. [Ca2+]i reduction increases cellular proliferation and apoptosis in vascular smooth muscle cells: relevance to the ADPKD phenotype. Circ Res 2005; 96:873-80. [PMID: 15790956 DOI: 10.1161/01.res.0000163278.68142.8a] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cardiovascular complications are the leading cause of morbidity and mortality in autosomal dominant polycystic kidney disease. Pkd2+/- vascular smooth muscle cells (VSMCs) have an abnormal phenotype and defective intracellular Ca2+ ([Ca2+]i) regulation. We examined cAMP content in vascular smooth muscles from Pkd2+/- mice because cAMP is elevated in cystic renal epithelial cells. We found cAMP concentration was significantly increased in Pkd2+/- vessels compared with wild-type vessels. Furthermore, reducing the wild-type VSMC [Ca2+]i by Verapamil or BAPTA-AM significantly increased cellular cAMP concentration (mainly by phosphodiesterase [PDE] inhibition), the rate of VSMC proliferation (determined by direct cell counting, 3H-incorporation, FACS analysis of cells entering S phase, and quantitative Western PCNA and ERK1/2 analyses), and the rate of apoptosis (by Hoechst staining, FACS analysis of the Annexin-V positive cells, and quantitative Western Bax, cytochrome c, and activated caspase 9 and 3 analyses). The low [Ca2+]i induced VSMC proliferation was independent of cAMP/B-Raf signaling, while that of apoptosis was promoted by cAMP. In summary, Pkd2+/- VSMCs have elevated cAMP levels. This elevation can also be induced by reducing [Ca2+]i in wild-type VSMCs. The [Ca2+]i reduction and cAMP accumulation can cause an increase in both cellular proliferation and apoptosis, resembling Pkd mutant phenotype.
Collapse
Affiliation(s)
- Sertac N Kip
- Division of Nephrology and Hypertension, Mayo Clinic College of Medicine, Rochester, Minn 55905, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Tao Y, Kim J, Stanley M, He Z, Faubel S, Schrier RW, Edelstein CL. Pathways of caspase-mediated apoptosis in autosomal-dominant polycystic kidney disease (ADPKD). Kidney Int 2005; 67:909-19. [PMID: 15698430 DOI: 10.1111/j.1523-1755.2005.00155.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND We have recently demonstrated an increase in apoptosis in Han:SPRD rat kidneys with autosomal-dominant polycystic kidney disease (ADPKD). Caspase-3 and caspase-7 are major mediators of apoptosis. There are two pathways of caspase-3 and caspase-7-mediated apoptosis: (1) the "extrinsic" pathway involving the death receptor Fas, Fas ligand (FasL), and caspase-8 and (2) the "mitochondrial" or "intrinsic" pathway involving Bcl-2 proteins, caspase-2, cytochrome c release, and caspase-9. The aim of the present study was to investigate the pathways of apoptosis in 3-week-old Han:SPRD rats with ADPKD. METHODS Fluorescent substrates were used to measure caspase activity. mRNA and protein was determined by ribonuclease protection assays and immunoblotting, respectively. The effect of caspase inhibitors on caspase activity in polycystic kidneys was determined. RESULTS Caspase-3 and caspase-7 activity was more than 100% increased in homozygous (Cy/Cy) compared to heterozygous (Cy/+) and normal littermate control (+/+) kidneys. Ribonuclease protection assays demonstrated no difference in caspase-3 mRNA. On immunoblotting, there was an increase in the proform of caspase-3 and caspase-7 in Cy/Cy compared to +/+ and Cy/+ kidneys. Caspase-8 and caspase-9 activity was more than 100% increased in Cy/Cy compared to Cy/+ and +/+ kidneys. On immunoblotting, there was an increase of the proform of both caspase-8 and caspase-9 in Cy/Cy kidneys. There was also an increase in cytochrome c release into the cytosol and an increase in caspase-2 protein and activity in Cy/Cy kidneys. On ribonuclease protection assay there was no difference in FasL mRNA between +/+, Cy/+, and Cy/Cy kidneys. Short-term treatment of Cy/Cy rats with the caspase inhibitor IDN-8050 resulted in inhibition of caspase-3 and caspase-7 activity in the kidney. CONCLUSION In Cy/Cy kidneys with ADPKD, there was an increase of the proform of caspase-9, an increase in cytochrome c release into the cytosol, and an increase in caspase-2 protein and activity demonstrating involvement of the intrinsic pathway. There was an increase in the proform of caspase-8 demonstrating involvement of the extrinsic pathway. No differences in FasL mRNA were seen suggesting that the extrinsic pathway is independent of the death receptor ligand, FasL.
Collapse
Affiliation(s)
- Yunxia Tao
- Division of Renal Diseases and Hypertension, University of Colorado Health Sciences Center, Denver, Colorado, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Sharma M, Brantley JG, Alcalay NI, Zhou J, Heystek E, Maser RL, Vanden Heuvel GB. Differential expression of Cux-1 and p21 in polycystic kidneys from Pkd1 null and cpk mice. Kidney Int 2005; 67:432-42. [PMID: 15673290 DOI: 10.1111/j.1523-1755.2005.67099.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Cux-1 is a murine homeodomain protein that functions as a cell cycle-dependent transcriptional repressor in proliferating cells. Targets of Cux-1 repression include the cyclin kinase inhibitors p21 and p27. In the kidney, Cux-1 is spatially and temporally regulated, and ectopic expression of Cux-1 in transgenic mice results in renal hyperplasia. Previously, we observed that Cux-1 is deregulated in cystic kidneys from cpk mice. Recent studies have suggested a role for the cyclin kinase inhibitor p21 in the development of polycystic kidney disease (PKD) in mice lacking PKD1. METHODS Since p21 is a target of transcriptional repression by Cux-1, we compared the expression of Cux-1 and p21 in kidneys from Pkd1 null and cpk mice by immunohistochemistry and Western blotting. We also evaluated apoptosis and the expression of the cyclin kinase inhibitor p27 in Pkd1 null and cpk mice by terminal deoxynucleotidal transferase (TdT)-mediated deoxyuridine triphosphate (dUTP) nick-end labeling (TUNEL) staining, immunohistochemistry, and Western blotting. RESULTS In both early and late embryonic kidneys from Pkd1 null mice, Cux-1 was highly and ectopically expressed in normal-appearing tubule epithelium, interstitial cells, and in the epithelial cells lining the cysts, where it colocalized with proliferating cell nuclear antigen (PCNA). Increased Cux-1 expression in Pkd1 null kidneys was also associated with a decrease in p27 expression at late stages of cystogenesis. In cpk kidneys, Cux-1 was not up-regulated until late stages of cyst development. Moreover, in contrast to Pkd1 null kidneys, p21 and p27 were highly expressed in cpk kidneys. In late stages of cystogenesis, Cux-1 and p21 colocalized in cyst lining cells, which also showed a high incidence of apoptosis. CONCLUSION These results suggest that cyst development in Pkd1 null mice and cpk mice proceeds through different mechanisms. In Pkd1 null mice, ectopic expression of Cux-1 is associated with increased cell proliferation. In contrast, in cpk mice, ectopic expression of Cux-1 is associated with apoptosis.
Collapse
Affiliation(s)
- Madhulika Sharma
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Hillman KA, Woolf AS, Johnson TM, Wade A, Unwin RJ, Winyard PJD. The P2X7 ATP receptor modulates renal cyst development in vitro. Biochem Biophys Res Commun 2004; 322:434-9. [PMID: 15325248 DOI: 10.1016/j.bbrc.2004.07.148] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2004] [Indexed: 11/28/2022]
Abstract
P2X(7), a purinergic receptor, is expressed in renal collecting ducts as they undergo fulminant cystogenesis in the cpk/cpk mouse model of autosomal recessive polycystic kidney disease (ARPKD). Dissociated cpk/cpk kidneys generate cysts from cell aggregates within 24h of suspension culture and we demonstrate that BzATP, a P2X(7) agonist, reduces cystogenesis. This effect is P2X(7)-specific, because: (i) equimolar concentrations of other purinergic agonists, ATP and UTP, had lesser effects and (ii) the P2X(7) inhibitor, oxidized ATP, abrogated the BzATP-mediated reduction in cystogenesis. BzATP did not significantly affect total cell number, proliferation, LDH release or caspase 3 activity, and zVAD-fmk, a caspase blocker, failed to modulate BzATP effects. In addition, this P2X(7) agonist did not significantly alter cyst size, probably excluding altered vectorial transport. In vivo, ATP was detected in cyst fluid from cpk/cpk kidneys; moreover, P2X(7) protein was also upregulated in human fetal ARPKD epithelia versus normal fetal collecting ducts. Thus, ATP may inhibit pathological renal cyst growth through P2X(7) signaling.
Collapse
Affiliation(s)
- Kate A Hillman
- Nephrology and Physiology, Royal Free Campus, Institute of Child Health, University College London, London, UK.
| | | | | | | | | | | |
Collapse
|
46
|
Abstract
Significant advances have been made recently in elucidating the cellular consequences of urinary tract obstruction during renal development. Urinary tract obstruction impairs growth and maturation of the kidney, and can also cause renal maldevelopment. This includes a reduction in the number of nephrons, tubular atrophy, and progressive interstitial fibrosis. Apoptosis (programmed cell death) accounts for much of the loss of tubular epithelial cells. Factors contributing to apoptosis include stretching of cells in dilated tubules, altered renal production of growth factors, and infiltration of the renal interstitium by macrophages. Two major controversies remain regarding the surgical management of congenital obstructive nephropathy: first, which fetuses with bladder outlet obstruction should undergo prenatal intervention, and second, which infants should undergo early pyeloplasty for ureteropelvic junction obstruction? Even after successful surgery for congential obstructive nephropathy, all patients should be followed for hypertension, proteinuria, or renal deterioration.
Collapse
Affiliation(s)
- Robert L Chevalier
- Department of Pediatrics, Children's Medical Center, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
47
|
Sorenson CM. Bcl-2 family members and disease. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2004; 1644:169-77. [PMID: 14996501 DOI: 10.1016/j.bbamcr.2003.08.010] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2003] [Accepted: 08/05/2003] [Indexed: 11/20/2022]
Abstract
Apoptosis plays an important role during development and in the maintenance of multicellular organisms. Bcl-2 family members affect cell death in either a positive or negative fashion. Although some redundancy exists between family members, expression of certain family members is important during development in an organ-specific manner. The founding family member bcl-2 tends to be highly expressed in the embryo and declines postnatally following differentiation and maturation. Altered expression of bcl-2, as well as other family members, has been observed in disease states potentially affecting treatment modalities. Here we examine the distribution and role death repressors bcl-2, bcl-x(L) and bcl-w as well as death effectors bax and bak play regulating apoptosis in a tissue-specific manner. Understanding the normal role of these proteins during embryogenesis and in the mature organ will give us important insight into what goes awry in various disease states.
Collapse
Affiliation(s)
- Christine M Sorenson
- Department of Pediatrics, University of Wisconsin-Madison, H4/444 CSC, 600 Highland Ave., Madison, WI 53792-4108, USA.
| |
Collapse
|
48
|
Abstract
Despite the sound experimental basis and initial promise of early animal models, the results of antenatal intervention have been disappointing, with high rates of misdiagnosis of urethral valves, complications from vesicoamniotic shunting, perinatal mortality, and long-term renal impairment and bladder dysfunction in survivors. The recent development of a cystoscopic approach might obviate some of these problems, but to date the procedure been limited by technical difficulty in negotiating the urethrovesical angle. Overcoming these difficulties through equipment modifications might allow definitive testing of whether or not alleviating distal urinary obstruction in utero is beneficial.
Collapse
Affiliation(s)
- Sailesh Kumar
- Centre for Fetal Care, Queen Charlotte's and Chelsea Hospital, Du Cane Road, London W12 OHS, UK.
| | | |
Collapse
|
49
|
Hu MC, Piscione TD, Rosenblum ND. Elevated SMAD1/beta-catenin molecular complexes and renal medullary cystic dysplasia in ALK3 transgenic mice. Development 2003; 130:2753-66. [PMID: 12736218 DOI: 10.1242/dev.00478] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Renal dysplasia, the most frequent cause of childhood renal failure in humans, arises from perturbations in a complex series of morphogenetic events during embryonic renal development. The molecular pathogenesis of renal dysplasia is largely undefined. While investigating the role of a BMP-dependent pathway that inhibits branching morphogenesis in vitro, we generated a novel model of renal dysplasia in a transgenic (Tg) model of ALK3 receptor signaling. We report the renal phenotype, and our discovery of molecular interactions between effectors in the BMP and WNT signaling pathways in dysplastic kidney tissue. Expression of the constitutively active ALK3 receptor ALK3(QD), in two independent transgenic lines caused renal aplasia/severe dysgenesis in 1.5% and 8.4% of hemizygous and homozygous Tg mice, respectively, and renal medullary cystic dysplasia in 49% and 74% of hemizygous and homozygous Tg mice, respectively. The dysplastic phenotype, which included a decreased number of medullary collecting ducts, increased medullary mesenchyme, collecting duct cysts and decreased cortical thickness, was apparent by E18.5. We investigated the pathogenesis of dysplasia in these mice, and demonstrated a 30% decrease in branching morphogenesis at E13.5 before the appearance of histopathogical features of dysplasia, and the formation of beta-catenin/SMAD1/SMAD4 molecular complexes in dysplastic renal tissue. Increased transcriptional activity of a beta-catenin reporter gene in ALK3(QD);Tcf-gal mice demonstrated functional cooperativity between the ALK3 and beta-catenin-dependent signaling pathways in kidney tissue. Together with our results in the dysplastic mouse kidney, our findings that phospho-SMAD1 and beta-catenin are overexpressed in human fetal dysplastic renal tissue suggest that dysregulation of these signaling effectors is pathogenic in human renal dysplasia. Our work provides novel insights into the role that crucial developmental signaling pathways may play during the genesis of malformed renal tissue elements.
Collapse
Affiliation(s)
- Ming Chang Hu
- Program in Developmental Biology, Research Institute, The Hospital for Sick Children, 555 University Avenue, Toronto, Ontario M5G 1X8, Canada
| | | | | |
Collapse
|
50
|
McGregor L, Makela V, Darling SM, Vrontou S, Chalepakis G, Roberts C, Smart N, Rutland P, Prescott N, Hopkins J, Bentley E, Shaw A, Roberts E, Mueller R, Jadeja S, Philip N, Nelson J, Francannet C, Perez-Aytes A, Megarbane A, Kerr B, Wainwright B, Woolf AS, Winter RM, Scambler PJ. Fraser syndrome and mouse blebbed phenotype caused by mutations in FRAS1/Fras1 encoding a putative extracellular matrix protein. Nat Genet 2003; 34:203-8. [PMID: 12766769 DOI: 10.1038/ng1142] [Citation(s) in RCA: 177] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2002] [Accepted: 03/24/2003] [Indexed: 11/09/2022]
Abstract
Fraser syndrome (OMIM 219000) is a multisystem malformation usually comprising cryptophthalmos, syndactyly and renal defects. Here we report autozygosity mapping and show that the locus FS1 at chromosome 4q21 is associated with Fraser syndrome, although the condition is genetically heterogeneous. Mutation analysis identified five frameshift mutations in FRAS1, which encodes one member of a family of novel proteins related to an extracellular matrix (ECM) blastocoelar protein found in sea urchin. The FRAS1 protein contains a series of N-terminal cysteine-rich repeat motifs previously implicated in BMP metabolism, suggesting that it has a role in both structure and signal propagation in the ECM. It has been speculated that Fraser syndrome is a human equivalent of the blebbed phenotype in the mouse, which has been associated with mutations in at least five loci including bl. As mapping data were consistent with homology of FRAS1 and bl, we screened DNA from bl/bl mice and identified a premature termination of mouse Fras1. Thus, the bl mouse is a model for Fraser syndrome in humans, a disorder caused by disrupted epithelial integrity in utero.
Collapse
Affiliation(s)
- Lesley McGregor
- Molecular Medicine, Institute of Child Health, London WC1N 1EH, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|