1
|
A human multi-lineage hepatic organoid model for liver fibrosis. Nat Commun 2021; 12:6138. [PMID: 34686668 PMCID: PMC8536785 DOI: 10.1038/s41467-021-26410-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 09/28/2021] [Indexed: 12/13/2022] Open
Abstract
To investigate the pathogenesis of a congenital form of hepatic fibrosis, human hepatic organoids were engineered to express the most common causative mutation for Autosomal Recessive Polycystic Kidney Disease (ARPKD). Here we show that these hepatic organoids develop the key features of ARPKD liver pathology (abnormal bile ducts and fibrosis) in only 21 days. The ARPKD mutation increases collagen abundance and thick collagen fiber production in hepatic organoids, which mirrors ARPKD liver tissue pathology. Transcriptomic and other analyses indicate that the ARPKD mutation generates cholangiocytes with increased TGFβ pathway activation, which are actively involved stimulating myofibroblasts to form collagen fibers. There is also an expansion of collagen-producing myofibroblasts with markedly increased PDGFRB protein expression and an activated STAT3 signaling pathway. Moreover, the transcriptome of ARPKD organoid myofibroblasts resemble those present in commonly occurring forms of liver fibrosis. PDGFRB pathway involvement was confirmed by the anti-fibrotic effect observed when ARPKD organoids were treated with PDGFRB inhibitors. Besides providing insight into the pathogenesis of congenital (and possibly acquired) forms of liver fibrosis, ARPKD organoids could also be used to test the anti-fibrotic efficacy of potential anti-fibrotic therapies.
Collapse
|
2
|
Therapeutic Potential for CFTR Correctors in Autosomal Recessive Polycystic Kidney Disease. Cell Mol Gastroenterol Hepatol 2021; 12:1517-1529. [PMID: 34329764 PMCID: PMC8529398 DOI: 10.1016/j.jcmgh.2021.07.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND & AIMS Autosomal recessive polycystic kidney disease (ARPKD) is caused by mutations in PKHD1, encoding fibrocystin/polyductin (FPC). Severe disease occurs in perinates. Those who survive the neonatal period face a myriad of comorbidities, including systemic and portal hypertension, liver fibrosis, and hepatosplenomegaly. The goal here was to uncover therapeutic strategies for ARPKD. METHODS We used wild-type and an FPC-mutant cholangiocyte cell line in 3-dimenional cysts and in confluent monolayers to evaluate protein expression using western blotting and protein trafficking using confocal microscopy. RESULTS We found that the protein level of the cystic fibrosis transmembrane conductance regulator (CFTR) was downregulated. The levels of heat shock proteins (HSPs) were altered in the FPC-mutant cholangiocytes, with HSP27 being downregulated and HSP90 and HSP70 upregulated. FPC-mutant cholangiocytes formed cysts, but normal cells did not. Cyst growth could be reduced by increasing HSP27 protein levels, by HSP90 and HSP70 inhibitor treatments, by silencing HSP90 through messenger RNA inhibition, or by the novel approach of treating the cysts with the CFTR corrector VX-809. In wild-type cholangiocytes, CFTR is present in both apical and basolateral membranes. FPC malfunction resulted in altered colocalization of CFTR with both apical and basolateral membranes. Whereas, treatment with VX-809, increasing HSP27 or inhibiting HSP70 or HSP90 restored CFTR localization toward normal values. CONCLUSIONS FPC malfunction induces the formation of cysts, which are fueled by alterations in HSPs and in CFTR protein levels and miss-localization. We suggest that CFTR correctors, already in clinical use to treat cystic fibrosis, could also be used as a treatment for ARPKD.
Collapse
|
3
|
Torres Crigna A, Daniele C, Gamez C, Medina Balbuena S, Pastene DO, Nardozi D, Brenna C, Yard B, Gretz N, Bieback K. Stem/Stromal Cells for Treatment of Kidney Injuries With Focus on Preclinical Models. Front Med (Lausanne) 2018; 5:179. [PMID: 29963554 PMCID: PMC6013716 DOI: 10.3389/fmed.2018.00179] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 05/24/2018] [Indexed: 12/18/2022] Open
Abstract
Within the last years, the use of stem cells (embryonic, induced pluripotent stem cells, or hematopoietic stem cells), Progenitor cells (e.g., endothelial progenitor cells), and most intensely mesenchymal stromal cells (MSC) has emerged as a promising cell-based therapy for several diseases including nephropathy. For patients with end-stage renal disease (ESRD), dialysis or finally organ transplantation are the only therapeutic modalities available. Since ESRD is associated with a high healthcare expenditure, MSC therapy represents an innovative approach. In a variety of preclinical and clinical studies, MSC have shown to exert renoprotective properties, mediated mainly by paracrine effects, immunomodulation, regulation of inflammation, secretion of several trophic factors, and possibly differentiation to renal precursors. However, studies are highly diverse; thus, knowledge is still limited regarding the exact mode of action, source of MSC in comparison to other stem cell types, administration route and dose, tracking of cells and documentation of therapeutic efficacy by new imaging techniques and tissue visualization. The aim of this review is to provide a summary of published studies of stem cell therapy in acute and chronic kidney injury, diabetic nephropathy, polycystic kidney disease, and kidney transplantation. Preclinical studies with allogeneic or xenogeneic cell therapy were first addressed, followed by a summary of clinical trials carried out with autologous or allogeneic hMSC. Studies were analyzed with respect to source of cell type, mechanism of action etc.
Collapse
Affiliation(s)
- Adriana Torres Crigna
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, University of Heidelberg, German Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany
| | - Cristina Daniele
- Medical Faculty Mannheim, Medical Research Centre, University of Heidelberg, Mannheim, Germany
| | - Carolina Gamez
- Department for Experimental Orthopaedics and Trauma Surgery, Medical Faculty Mannheim, Orthopaedic and Trauma Surgery Centre (OUZ), Heidelberg University, Mannheim, Germany
| | - Sara Medina Balbuena
- Department of Medicine (Nephrology/Endrocrinology/Rheumathology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Diego O. Pastene
- Department of Medicine (Nephrology/Endrocrinology/Rheumathology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Daniela Nardozi
- Medical Faculty Mannheim, Medical Research Centre, University of Heidelberg, Mannheim, Germany
| | - Cinzia Brenna
- Medical Faculty Mannheim, Medical Research Centre, University of Heidelberg, Mannheim, Germany
| | - Benito Yard
- Department of Medicine (Nephrology/Endrocrinology/Rheumathology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Norbert Gretz
- Medical Faculty Mannheim, Medical Research Centre, University of Heidelberg, Mannheim, Germany
| | - Karen Bieback
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, University of Heidelberg, German Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany
| |
Collapse
|
4
|
Hamo S, Bacchetta J, Bertholet-Thomas A, Ranchin B, Cochat P, Michel-Calemard L. [PKHD1 mutations in autosomal recessive polycystic kidney disease (ARPKD): Genotype-phenotype correlations from a series of 308 cases to improve prenatal counselling]. Nephrol Ther 2018; 14:474-477. [PMID: 29703621 DOI: 10.1016/j.nephro.2018.03.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 03/17/2018] [Accepted: 03/18/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVES ARPKD is a recessive rare disease due to PKHD1 mutation. The main objective of the study was to characterize the phenotypic variability according to the different types of PKHD1 mutations. METHODS This study was performed in 308 ARPKD patients with a genetic diagnosis from our genetic center. Related physicians provided minimal clinical and biological data. RESULTS Patients were divided into three genotypic groups: the first group (G1; n=65) consisted of patients with two truncating mutations, the second group (G2; n=117) of patients with one truncating and one non-truncating mutation, and the third group (G3; n=126) of patients with two non-truncating mutations. In the entire cohort, the outcomes consisted of 31% of pregnancy termination, 18% of neonatal deaths and 51% of patient survival after the neonatal period. The proportion of severe ARPKD (pregnancy termination or neonatal death) was significantly greater in G1: 94% versus 47% in G2 and 27% in G3 (P<0.001). CONCLUSION The presence of two truncating mutations in PKHD1 is associated with the most severe perinatal phenotype. However, the phenotypic variability observed in the other genotypic groups requires caution for prenatal counseling.
Collapse
Affiliation(s)
- Suzy Hamo
- Centre de référence des maladies rénales rares néphrogones, filière ORKID, hôpital Femme-Mère-Enfant, 59, boulevard Pinel, 69677 Bron cedex, France.
| | - Justine Bacchetta
- Centre de référence des maladies rénales rares néphrogones, filière ORKID, hôpital Femme-Mère-Enfant, 59, boulevard Pinel, 69677 Bron cedex, France; Faculté de médecine Lyon Est, université Claude-Bernard Lyon 1, 69008 Lyon, France
| | - Aurélia Bertholet-Thomas
- Centre de référence des maladies rénales rares néphrogones, filière ORKID, hôpital Femme-Mère-Enfant, 59, boulevard Pinel, 69677 Bron cedex, France
| | - Bruno Ranchin
- Centre de référence des maladies rénales rares néphrogones, filière ORKID, hôpital Femme-Mère-Enfant, 59, boulevard Pinel, 69677 Bron cedex, France
| | - Pierre Cochat
- Centre de référence des maladies rénales rares néphrogones, filière ORKID, hôpital Femme-Mère-Enfant, 59, boulevard Pinel, 69677 Bron cedex, France; Faculté de médecine Lyon Est, université Claude-Bernard Lyon 1, 69008 Lyon, France
| | - Laurence Michel-Calemard
- UM pathologies endocriniennes, rénales, musculaires et mucoviscidose, CBPE, groupement hospitalier Est, 59, boulevard Pinel, 69677 Bron cedex, France
| |
Collapse
|
5
|
Dillman JR, Trout AT, Smith EA, Towbin AJ. Hereditary Renal Cystic Disorders: Imaging of the Kidneys and Beyond. Radiographics 2017; 37:924-946. [PMID: 28493804 DOI: 10.1148/rg.2017160148] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The purpose of this article is to review the hereditary renal cystic diseases that can manifest in children and adults, with specific attention to pathogenesis and imaging features. Various common and uncommon hereditary renal cystic diseases are reviewed in terms of their underlying etiology, including the involved genetic mutations and the affected proteins and cellular structures. Focus is placed on the morphologic findings in each condition and the features that distinguish one disorder from another. The two most common categories of hereditary renal cystic disease are (a) the ciliopathic disorders, which are related to mutations affecting the primary cilia (called "ciliopathies"), and (b) the phakomatoses. Autosomal dominant polycystic kidney disease, autosomal recessive polycystic kidney disease, and the "medullary cystic disease complex" are all ciliopathies but have different phenotypes. Tuberous sclerosis complex and the associated "contiguous gene syndrome," as well as von Hippel-Lindau syndrome, are phakomatoses that can manifest with cystic renal lesions but have uniquely different extrarenal manifestations. Finally, DICER1 mutations can manifest with renal cystic lesions (typically, cystic nephromas) in patients predisposed to other malignancies in the chest, ovaries, and thyroid. Although some overlap exists in the appearance of the renal cysts associated with each of these diseases, there are clear morphologic differences (eg, cyst size, location, and complexity) that are emphasized in this review. To improve patient outcomes, it is important for the radiologist to recognize the various hereditary renal cystic diseases so that a correct diagnosis is assigned and so that the patient is adequately evaluated and followed up. ©RSNA, 2017.
Collapse
Affiliation(s)
- Jonathan R Dillman
- From the Department of Radiology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229-3039 (J.R.D., A.T.T., A.J.T.); and the Section of Pediatric Radiology, Department of Radiology, University of Michigan Health System, Ann Arbor, Mich (E.A.S.)
| | - Andrew T Trout
- From the Department of Radiology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229-3039 (J.R.D., A.T.T., A.J.T.); and the Section of Pediatric Radiology, Department of Radiology, University of Michigan Health System, Ann Arbor, Mich (E.A.S.)
| | - Ethan A Smith
- From the Department of Radiology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229-3039 (J.R.D., A.T.T., A.J.T.); and the Section of Pediatric Radiology, Department of Radiology, University of Michigan Health System, Ann Arbor, Mich (E.A.S.)
| | - Alexander J Towbin
- From the Department of Radiology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229-3039 (J.R.D., A.T.T., A.J.T.); and the Section of Pediatric Radiology, Department of Radiology, University of Michigan Health System, Ann Arbor, Mich (E.A.S.)
| |
Collapse
|
6
|
|
7
|
Evidence for a "Pathogenic Triumvirate" in Congenital Hepatic Fibrosis in Autosomal Recessive Polycystic Kidney Disease. BIOMED RESEARCH INTERNATIONAL 2016; 2016:4918798. [PMID: 27891514 PMCID: PMC5116503 DOI: 10.1155/2016/4918798] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 09/20/2016] [Accepted: 10/13/2016] [Indexed: 12/29/2022]
Abstract
Autosomal recessive polycystic kidney disease (ARPKD) is a severe monogenic disorder that occurs due to mutations in the PKHD1 gene. Congenital hepatic fibrosis (CHF) associated with ARPKD is characterized by the presence of hepatic cysts derived from dilated bile ducts and a robust, pericystic fibrosis. Cyst growth, due to cyst wall epithelial cell hyperproliferation and fluid secretion, is thought to be the driving force behind disease progression. Liver fibrosis is a wound healing response in which collagen accumulates in the liver due to an imbalance between extracellular matrix synthesis and degradation. Whereas both hyperproliferation and pericystic fibrosis are hallmarks of CHF/ARPKD, whether or not these two processes influence one another remains unclear. Additionally, recent studies demonstrate that inflammation is a common feature of CHF/ARPKD. Therefore, we propose a "pathogenic triumvirate" consisting of hyperproliferation of cyst wall growth, pericystic fibrosis, and inflammation which drives CHF/ARPKD progression. This review will summarize what is known regarding the mechanisms of cyst growth, fibrosis, and inflammation in CHF/ARPKD. Further, we will discuss the potential advantage of identifying a core pathogenic feature in CHF/ARPKD to aid in the development of novel therapeutic approaches. If a core pathogenic feature does not exist, then developing multimodality therapeutic approaches to target each member of the "pathogenic triumvirate" individually may be a better strategy to manage this debilitating disease.
Collapse
|
8
|
Edrees BM, Athar M, Al-Allaf FA, Taher MM, Khan W, Bouazzaoui A, Al-Harbi N, Safar R, Al-Edressi H, Alansary K, Anazi A, Altayeb N, Ahmed MA, Abduljaleel Z. Next-generation sequencing for molecular diagnosis of autosomal recessive polycystic kidney disease. Gene 2016; 591:214-226. [DOI: 10.1016/j.gene.2016.07.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 06/26/2016] [Accepted: 07/07/2016] [Indexed: 12/18/2022]
|
9
|
Beaudry JB, Cordi S, Demarez C, Lepreux S, Pierreux CE, Lemaigre FP. Proliferation-Independent Initiation of Biliary Cysts in Polycystic Liver Diseases. PLoS One 2015; 10:e0132295. [PMID: 26125584 PMCID: PMC4488361 DOI: 10.1371/journal.pone.0132295] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 06/11/2015] [Indexed: 12/14/2022] Open
Abstract
Biliary cysts in adult patients affected by polycystic liver disease are lined by cholangiocytes that proliferate, suggesting that initiation of cyst formation depends on proliferation. Here, we challenge this view by analyzing cyst-lining cell proliferation and differentiation in Cpk mouse embryos and in livers from human fetuses affected by Autosomal Recessive Polycystic Kidney Disease (ARPKD), at early stages of cyst formation. Proliferation of fetal cholangiocyte precursors, measured by immunostaining in human and mouse livers, was low and did not differ between normal and ARPKD or Cpk livers, excluding excessive proliferation as an initiating cause of liver cysts. Instead, our analyses provide evidence that the polycystic livers exhibit increased and accelerated differentiation of hepatoblasts into cholangiocyte precursors, eventually coalescing into large biliary cysts. Lineage tracing experiments, performed in mouse embryos, indicated that the cholangiocyte precursors in Cpk mice generate cholangiocytes and periportal hepatocytes, like in wild-type animals. Therefore, contrary to current belief, cyst formation in polycystic liver disease does not necessarily depend on overproliferation. Combining our prenatal data with available data from adult livers, we propose that polycystic liver can be initiated by proliferation-independent mechanisms at a fetal stage, followed by postnatal proliferation-dependent cyst expansion.
Collapse
Affiliation(s)
| | - Sabine Cordi
- Université catholique de Louvain, de Duve Institute, Brussels, Belgium
| | - Céline Demarez
- Université catholique de Louvain, de Duve Institute, Brussels, Belgium
| | | | | | | |
Collapse
|
10
|
Kurschat CE, Müller RU, Franke M, Maintz D, Schermer B, Benzing T. An approach to cystic kidney diseases: the clinician's view. Nat Rev Nephrol 2014; 10:687-99. [DOI: 10.1038/nrneph.2014.173] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
11
|
Novel Mutation in the PKHD1 Gene Diagnosed Prenatally in a Fetus with Autosomal Recessive Polycystic Kidney Disease. Case Rep Genet 2014; 2014:517952. [PMID: 25114813 PMCID: PMC4120792 DOI: 10.1155/2014/517952] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 06/11/2014] [Accepted: 06/25/2014] [Indexed: 11/24/2022] Open
Abstract
We report a 29-year-old gravida 2, para 0100, who presented at 19 weeks and 4 days of gestation for ultrasound to assess fetal anatomy. Routine midtrimester fetal anatomy ultrasound revealed enlarged, hyperechoic fetal kidneys and normal amniotic fluid index. Follow-up ultrasound at 23 weeks and 5 days revealed persistently enlarged, hyperechoic fetal kidneys. Progressive oligohydramnios was not evident until 29 weeks of gestation, with anhydramnios noted by 35 weeks of gestation. Amniocentesis was performed for karyotype and to search for mutations in the PKHD1 for the presumptive diagnosis of autosomal recessive polycystic kidney disease (ARPKD). In our patient, a maternally inherited, previously reported pathogenic missense mutation in the PKHD1 gene, c.10444C>T, was identified. A second, previously unreported de novo mutation, c.5909-2delA, was also identified. This mutation affects the canonical splice site and is most likely pathogenic. Our case highlights PKHD1 allelic heterogeneity and the importance of genetic testing in the prenatal setting where many other genetic etiologies can phenocopy ARPKD.
Collapse
|
12
|
Sweeney WE, Avner ED. Pathophysiology of childhood polycystic kidney diseases: new insights into disease-specific therapy. Pediatr Res 2014; 75:148-57. [PMID: 24336431 PMCID: PMC3953890 DOI: 10.1038/pr.2013.191] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 07/11/2013] [Indexed: 12/22/2022]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) and autosomal recessive polycystic kidney disease (ARPKD) are significant causes of morbidity and mortality in children and young adults. ADPKD, with an incidence of 1:400 to 1:1,000, affects more than 13 million individuals worldwide and is a major cause of end-stage renal disease in adults. However, symptomatic disease is increasingly recognized in children. ARPKD is a dual-organ hepatorenal disease with an incidence of 1:20,000 to 1:40,000 and a heterozygote carrier rate of 1 in 70. Currently, no clinically significant disease-specific therapy exists for ADPKD or ARPKD. The genetic basis of both ADPKD and ARPKD have been identified, and delineation of the basic molecular and cellular pathophysiology has led to the discovery that abnormal ADPKD and ARPKD gene products interact to create "polycystin complexes" located at multiple sites within affected cells. The extracellular matrix and vessels produce a variety of soluble factors that affect the biology of adjacent cells in many dynamic ways. This review will focus on the molecular and cellular bases of the abnormal cystic phenotype and discuss the clinical translation of such basic data into new therapies that promise to alter the natural history of disease for children with genetic PKDs.
Collapse
Affiliation(s)
- William E. Sweeney
- Department of Pediatrics and Children’s Research Institute, Medical College of Wisconsin and Children’s Hospital Health System of Wisconsin, Milwaukee, WI
| | - Ellis D. Avner
- Department of Pediatrics and Children’s Research Institute, Medical College of Wisconsin and Children’s Hospital Health System of Wisconsin, Milwaukee, WI,Department of Physiology, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
13
|
Freedman BS, Lam AQ, Sundsbak JL, Iatrino R, Su X, Koon SJ, Wu M, Daheron L, Harris PC, Zhou J, Bonventre JV. Reduced ciliary polycystin-2 in induced pluripotent stem cells from polycystic kidney disease patients with PKD1 mutations. J Am Soc Nephrol 2013; 24:1571-86. [PMID: 24009235 PMCID: PMC3785271 DOI: 10.1681/asn.2012111089] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Accepted: 04/22/2013] [Indexed: 11/03/2022] Open
Abstract
Heterozygous mutations in PKD1 or PKD2, which encode polycystin-1 (PC1) and polycystin-2 (PC2), respectively, cause autosomal dominant PKD (ADPKD), whereas mutations in PKHD1, which encodes fibrocystin/polyductin (FPC), cause autosomal recessive PKD (ARPKD). However, the relationship between these proteins and the pathogenesis of PKD remains unclear. To model PKD in human cells, we established induced pluripotent stem (iPS) cell lines from fibroblasts of three ADPKD and two ARPKD patients. Genetic sequencing revealed unique heterozygous mutations in PKD1 of the parental ADPKD fibroblasts but no pathogenic mutations in PKD2. Undifferentiated PKD iPS cells, control iPS cells, and embryonic stem cells elaborated primary cilia and expressed PC1, PC2, and FPC at similar levels, and PKD and control iPS cells exhibited comparable rates of proliferation, apoptosis, and ciliogenesis. However, ADPKD iPS cells as well as somatic epithelial cells and hepatoblasts/biliary precursors differentiated from these cells expressed lower levels of PC2 at the cilium. Additional sequencing confirmed the retention of PKD1 heterozygous mutations in iPS cell lines from two patients but identified possible loss of heterozygosity in iPS cell lines from one patient. Furthermore, ectopic expression of wild-type PC1 in ADPKD iPS-derived hepatoblasts rescued ciliary PC2 protein expression levels, and overexpression of PC1 but not a carboxy-terminal truncation mutant increased ciliary PC2 expression levels in mouse kidney cells. Taken together, these results suggest that PC1 regulates ciliary PC2 protein expression levels and support the use of PKD iPS cells for investigating disease pathophysiology.
Collapse
Affiliation(s)
- Benjamin S. Freedman
- Renal Division and Harvard Center for Polycystic Kidney Disease Research, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Albert Q. Lam
- Renal Division and Harvard Center for Polycystic Kidney Disease Research, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- Harvard Stem Cell Institute, Harvard University, Cambridge and Boston Massachusetts
| | - Jamie L. Sundsbak
- Mayo Translational Polycystic Kidney Disease Center, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota; and
| | - Rossella Iatrino
- Renal Division and Harvard Center for Polycystic Kidney Disease Research, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- Nephrology, Dialysis, and Transplant Division, Policlinico Universitario di Modena, Modena, Italy
| | - Xuefeng Su
- Renal Division and Harvard Center for Polycystic Kidney Disease Research, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sarah J. Koon
- Mayo Translational Polycystic Kidney Disease Center, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota; and
| | - Maoqing Wu
- Renal Division and Harvard Center for Polycystic Kidney Disease Research, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Laurence Daheron
- Harvard Stem Cell Institute, Harvard University, Cambridge and Boston Massachusetts
| | - Peter C. Harris
- Mayo Translational Polycystic Kidney Disease Center, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota; and
| | - Jing Zhou
- Renal Division and Harvard Center for Polycystic Kidney Disease Research, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- Harvard Stem Cell Institute, Harvard University, Cambridge and Boston Massachusetts
| | - Joseph V. Bonventre
- Renal Division and Harvard Center for Polycystic Kidney Disease Research, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- Harvard Stem Cell Institute, Harvard University, Cambridge and Boston Massachusetts
| |
Collapse
|
14
|
Yengkopiong JP, Lako JDW. Genetic background of nonmutant Piebald-Virol-Glaxo rats does not influence nephronophthisis phenotypes. Int J Nephrol Renovasc Dis 2013; 6:39-46. [PMID: 23549608 PMCID: PMC3579405 DOI: 10.2147/ijnrd.s39295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Nephronophthisis (NPHP), which affects multiple organs, is a hereditary cystic kidney disease (CKD), characterized by interstitial fibrosis and numerous fluid-filled cysts in the kidneys. It is caused by mutations in NPHP genes, which encode for ciliary proteins known as nephrocystins. The disorder affects many people across the world and leads to end-stage renal disease. The aim of this study was to determine if the genetic background of the nonmutant female Piebald-Virol-Glaxo (PVG/Seac(-/-)) rat influences phenotypic inheritance of NPHP from mutant male Lewis polycystic kidney rats. METHODS Mating experiments were performed between mutant Lewis polycystic kidney male rats with CKD and nonmutant PVG and Wistar Kyoto female rats without cystic kidney disease to raise second filial and backcross 1 progeny, respectively. Rats that developed cystic kidneys were identified. Systolic blood pressure was determined in each rat at 12 weeks of age using the tail and cuff method. After euthanasia, blood samples were collected and chemistry was determined. Histological examination of the kidneys, pancreas, and liver of rats with and without cystic kidney disease was performed. RESULTS It was established that the genetic background of nonmutant female PVG rats did not influence the phenotypic inheritance of the CKD from mutant male Lewis polycystic kidney rats. The disease arose as a result of a recessive mutation in a single gene (second filial generation, CKD = 13, non-CKD = 39, χ (2) = 0.00, P ≥ 0.97; backcross 1 generation, CKD = 67, non-CKD = 72, χ (2) = 0.18, P > 0.05) and inherited as NPHP. The rats with CKD developed larger fluid-filled cystic kidneys, higher systolic blood pressure, and anemia, but there were no extrarenal cysts and disease did not lead to early pup mortality. CONCLUSION The genetic background of the nonmutant PVG rats does not influence the genetic and phenotypic inheritance of CKD from mutant Lewis polycystic kidney rats. A single recessive mutation incapacitated the gene, which relaxed its functional constraints, and led to formation of multiple cysts in the kidneys of the homozygous mutant rats.
Collapse
Affiliation(s)
- Jada Pasquale Yengkopiong
- John Garang Memorial University of Science and Technology, Faculty of Science and Technology, Bor, Jonglei State, Republic of South Sudan
| | | |
Collapse
|
15
|
Sun Y, Zhou H, Yang BX. Drug discovery for polycystic kidney disease. Acta Pharmacol Sin 2011; 32:805-16. [PMID: 21642949 PMCID: PMC4009953 DOI: 10.1038/aps.2011.29] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2011] [Accepted: 03/17/2011] [Indexed: 12/19/2022] Open
Abstract
In polycystic kidney disease (PKD), a most common human genetic diseases, fluid-filled cysts displace normal renal tubules and cause end-stage renal failure. PKD is a serious and costly disorder. There is no available therapy that prevents or slows down the cystogenesis and cyst expansion in PKD. Numerous efforts have been made to find drug targets and the candidate drugs to treat PKD. Recent studies have defined the mechanisms underlying PKD and new therapies directed toward them. In this review article, we summarize the pathogenesis of PKD, possible drug targets, available PKD models for screening and evaluating new drugs as well as candidate drugs that are being developed.
Collapse
Affiliation(s)
- Ying Sun
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing 100191, China
| | - Hong Zhou
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing 100191, China
| | - Bao-xue Yang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing 100191, China
| |
Collapse
|
16
|
Hu B, He X, Li A, Qiu Q, Li C, Liang D, Zhao P, Ma J, Coffey RJ, Zhan Q, Wu G. Cystogenesis in ARPKD results from increased apoptosis in collecting duct epithelial cells of Pkhd1 mutant kidneys. Exp Cell Res 2010; 317:173-87. [PMID: 20875407 DOI: 10.1016/j.yexcr.2010.09.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Revised: 09/09/2010] [Accepted: 09/19/2010] [Indexed: 12/14/2022]
Abstract
Mutations in the PKHD1 gene result in autosomal recessive polycystic kidney disease (ARPKD) in humans. To determine the molecular mechanism of the cystogenesis in ARPKD, we recently generated a mouse model for ARPKD that carries a targeted mutation in the mouse orthologue of human PKHD1. The homozygous mutant mice display hepatorenal cysts whose phenotypes are similar to those of human ARPKD patients. By littermates of this mouse, we developed two immortalized renal collecting duct cell lines with Pkhd1 and two without. Under nonpermissive culture conditions, the Pkhd1(-/-) renal cells displayed aberrant cell-cell contacts and tubulomorphogenesis. The Pkhd1(-/-) cells also showed significantly reduced cell proliferation and elevated apoptosis. To validate this finding in vivo, we examined proliferation and apoptosis in the kidneys of Pkhd1(-/-) mice and their wildtype littermates. Using proliferation (PCNA and Histone-3) and apoptosis (TUNEL and caspase-3) markers, similar results were obtained in the Pkhd1(-/-) kidney tissues as in the cells. To identify the molecular basis of these findings, we analyzed the effect of Pkhd1 loss on multiple putative signaling regulators. We demonstrated that the loss of Pkhd1 disrupts multiple major phosphorylations of focal adhesion kinase (FAK), and these disruptions either inhibit the Ras/C-Raf pathways to suppress MEK/ERK activity and ultimately reduce cell proliferation, or suppress PDK1/AKT to upregulate Bax/caspase-9/caspase-3 and promote apoptosis. Our findings indicate that apoptosis may be a major player in the cyst formation in ARPKD, which may lead to new therapeutic strategies for human ARPKD.
Collapse
MESH Headings
- Animals
- Apoptosis/genetics
- Caspase 3/genetics
- Caspase 3/metabolism
- Caspase 9/genetics
- Caspase 9/metabolism
- Cell Line, Transformed
- Cell Proliferation
- Crosses, Genetic
- Cysts/genetics
- Disease Models, Animal
- Epithelial Cells/metabolism
- Genes, cdc
- Genotype
- Humans
- In Vitro Techniques
- Kidney/metabolism
- Kidney Tubules, Collecting/metabolism
- Mice
- Mice, Congenic
- Mice, Inbred C57BL
- Mice, Knockout
- Mutation
- Phenotype
- Polycystic Kidney, Autosomal Recessive/genetics
- Polycystic Kidney, Autosomal Recessive/metabolism
- Polycystic Kidney, Autosomal Recessive/pathology
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- Receptors, Cell Surface/genetics
- Signal Transduction/genetics
Collapse
Affiliation(s)
- Bo Hu
- Cancer Research Institute, University of South China, Hengyang, Hunan, 421001, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Deltas C, Papagregoriou G. Cystic diseases of the kidney: molecular biology and genetics. Arch Pathol Lab Med 2010; 134:569-82. [PMID: 20367309 DOI: 10.5858/134.4.569] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT Cystic diseases of the kidney are a very heterogeneous group of renal inherited conditions, with more than 33 genes involved and encompassing X-linked, autosomal dominant, and autosomal recessive inheritance. Although mostly monogenic with mendelian inheritance, there are clearly examples of oligogenic inheritance, such as 3 mutations in 2 genes, while the existence of genetic modifiers is perhaps the norm, based on the extent of variable expressivity and the broad spectrum of symptoms. OBJECTIVES To present in the form of a mini review the major known cystic diseases of the kidney for which genes have been mapped or cloned and characterized, with some information on their cellular and molecular biology and genetics, and to pay special attention to commenting on the issues of molecular diagnostics, in view of the genetic and allelic heterogeneity. Data Sources.-We used major reviews that make excellent detailed presentation of the various diseases, as well as original publications. CONCLUSIONS There is already extensive genetic heterogeneity in the group of cystic diseases of the kidney; however, there are still many more genes awaiting to be discovered that are implicated or mutated in these diseases. In addition, the synergism and interaction among this repertoire of gene products is largely unknown, while a common unifying aspect is the expression of nearly all of them at the primary cilium or the basal body. A major interplay of functions is anticipated, while mutations in all converge in the unifying phenotype of cyst formation.
Collapse
|
18
|
Di Bisceglie AM, Befeler AS. Tumors and Cysts of the Liver. SLEISENGER AND FORDTRAN'S GASTROINTESTINAL AND LIVER DISEASE 2010:1569-1592.e6. [DOI: 10.1016/b978-1-4160-6189-2.00094-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
19
|
Wilson PD, Goilav B. Cystic disease of the kidney. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2008; 2:341-68. [PMID: 18039103 DOI: 10.1146/annurev.pathol.2.010506.091850] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
This review focuses on the mechanisms that underlie the development of human renal cystic diseases. A pathological, clinical, and pathophysiological overview is given. Initial analysis of the cell biology of inappropriate hyperproliferation accompanied by fluid secretion of cyst-lining epithelia has been followed by the elucidation of fundamental defects in epithelial polarity, cell-matrix and cell-cell interactions, and apoptosis, all of which are discussed. Identification of the genes and proteins responsible for several renal cystic diseases has led to a more complete understanding of defects in renal developmental programming, differentiation, and morphogenesis, all of which underlie cystic diseases of the kidney.
Collapse
Affiliation(s)
- Patricia D Wilson
- Division of Nephrology, Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029, USA.
| | | |
Collapse
|
20
|
Abstract
An explosion of work over the last decade has produced insight into the multiple hereditary causes of a nonimmunological form of diabetes diagnosed most frequently within the first 6 months of life. These studies are providing increased understanding of genes involved in the entire chain of steps that control glucose homeostasis. Neonatal diabetes is now understood to arise from mutations in genes that play critical roles in the development of the pancreas, of beta-cell apoptosis and insulin processing, as well as the regulation of insulin release. For the basic researcher, this work is providing novel tools to explore fundamental molecular and cellular processes. For the clinician, these studies underscore the need to identify the genetic cause underlying each case. It is increasingly clear that the prognosis, therapeutic approach, and genetic counseling a physician provides must be tailored to a specific gene in order to provide the best medical care.
Collapse
Affiliation(s)
- Lydia Aguilar-Bryan
- Pacific Northwest Diabetes Research Institute, 720 Broadway, Seattle, Washington 98122, USA.
| | | |
Collapse
|
21
|
Yang B, Sonawane ND, Zhao D, Somlo S, Verkman AS. Small-molecule CFTR inhibitors slow cyst growth in polycystic kidney disease. J Am Soc Nephrol 2008; 19:1300-10. [PMID: 18385427 DOI: 10.1681/asn.2007070828] [Citation(s) in RCA: 161] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Cyst expansion in polycystic kidney disease (PKD) involves progressive fluid accumulation, which is believed to require chloride transport by the cystic fibrosis transmembrane conductance regulator (CFTR) protein. Herein is reported that small-molecule CFTR inhibitors of the thiazolidinone and glycine hydrazide classes slow cyst expansion in in vitro and in vivo models of PKD. More than 30 CFTR inhibitor analogs were screened in an MDCK cell model, and near-complete suppression of cyst growth was found by tetrazolo-CFTR(inh)-172, a tetrazolo-derived thiazolidinone, and Ph-GlyH-101, a phenyl-derived glycine hydrazide, without an effect on cell proliferation. These compounds also inhibited cyst number and growth by >80% in an embryonic kidney cyst model involving 4-d organ culture of embryonic day 13.5 mouse kidneys in 8-Br-cAMP-containing medium. Subcutaneous delivery of tetrazolo-CFTR(inh)-172 and Ph-GlyH-101 to neonatal, kidney-specific PKD1 knockout mice produced stable, therapeutic inhibitor concentrations of >3 microM in urine and kidney tissue. Treatment of mice for up to 7 d remarkably slowed kidney enlargement and cyst expansion and preserved renal function. These results implicate CFTR in renal cyst growth and suggest that CFTR inhibitors may hold therapeutic potential to reduce cyst growth in PKD.
Collapse
Affiliation(s)
- Baoxue Yang
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | | | | | | | | |
Collapse
|
22
|
Abstract
Autosomal recessive polycystic kidney disease (ARPKD) is a serious genetic disease characterized by cystic changes in the collecting ducts of the kidney and bile ducts within the liver. The gene for ARPKD (PKHD1) is located on chromosome 6p12 and encodes a protein called fibrocystin/polyductin (FPC), 1 of many proteins that are normally present at the primary cilia of the renal tubules and intrahepatic bile ducts. The severity of the clinical disease depends on the type of genetic mutations. Although exact function of FPC is not fully known, it is generally felt that like many of the other ciliary proteins, it plays a vital role in maintaining the structural integrity of organs such as kidney and liver, by modulating important cellular functions, including proliferation, secretion, apoptosis, and terminal differentiation. FPC probably works in conjunction with cellular proteins involved in autosomal dominant polycystic kidney disease that is, polycystin-1 and polycystin-2, which are also located in the primary cilia. Genetic abnormalities in PKHD1 may result in structural and functional abnormalities of FPC, leading to cystic phenotype.
Collapse
|
23
|
Serra AL, Kistler AD, Poster D, Struker M, Wüthrich RP, Weishaupt D, Tschirch F. Clinical proof-of-concept trial to assess the therapeutic effect of sirolimus in patients with autosomal dominant polycystic kidney disease: SUISSE ADPKD study. BMC Nephrol 2007; 8:13. [PMID: 17868472 PMCID: PMC2048941 DOI: 10.1186/1471-2369-8-13] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2007] [Accepted: 09/15/2007] [Indexed: 12/15/2022] Open
Abstract
Background Currently there is no effective treatment available to retard cyst growth and to prevent the progression to end-stage renal failure in patients with autosomal dominant polycystic kidney disease (ADPKD). Evidence has recently been obtained from animal experiments that activation of the mammalian target of rapamycin (mTOR) signaling pathway plays a crucial role in cyst growth and renal volume expansion, and that the inhibition of mTOR with rapamycin (sirolimus) markedly slows cyst development and renal functional deterioration. Based on these promising results in animals we have designed and initiated the first randomized controlled trial (RCT) to examine the effectiveness, safety and tolerability of sirolimus to retard disease progression in ADPKD. Method/design This single center, randomised controlled, open label trial assesses the therapeutic effect, safety and tolerability of the mTOR inhibitor sirolimus (Rapamune®) in patients with autosomal dominant polycystic kidney disease and preserved renal function. The primary outcome will be the inhibition of kidney volume growth measured by magnetic resonance imaging (MRI) volumetry. Secondary outcome parameters will be preservation of renal function, safety and tolerability of sirolimus. Discussion The results from this proof-of-concept RCT will for the first time show whether treatment with sirolimus effectively retards cyst growth in patients with ADPKD. Trial registration NCT00346918
Collapse
Affiliation(s)
- Andreas L Serra
- Clinic for Nephrology, University Hospital, CH-8091 Zürich, Switzerland
| | - Andreas D Kistler
- Clinic for Nephrology, University Hospital, CH-8091 Zürich, Switzerland
| | - Diane Poster
- Clinic for Nephrology, University Hospital, CH-8091 Zürich, Switzerland
| | - Marian Struker
- Clinic for Nephrology, University Hospital, CH-8091 Zürich, Switzerland
| | - Rudolf P Wüthrich
- Clinic for Nephrology, University Hospital, CH-8091 Zürich, Switzerland
| | - Dominik Weishaupt
- Institute of Diagnostic Radiology, University Hospital, CH-8091 Zürich, Switzerland
| | - Frank Tschirch
- Institute of Diagnostic Radiology, University Hospital, CH-8091 Zürich, Switzerland
| |
Collapse
|
24
|
Michael L, Sweeney DE, Davies JA. The lectin Dolichos biflorus agglutinin is a sensitive indicator of branching morphogenetic activity in the developing mouse metanephric collecting duct system. J Anat 2007; 210:89-97. [PMID: 17229286 PMCID: PMC2100263 DOI: 10.1111/j.1469-7580.2006.00670.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
The urine collecting duct system of the metanephric kidney develops by growth and branching morphogenesis of an unbranched progenitor tubule, the ureteric bud. Bud branching is mainly dichotomous and new branches form from existing branch tips, which are also the main sites of cell proliferation in the system. This behaviour, and the fact that some genes (e.g. Wnt11, Sox9) are expressed only in tips, suggests that tip cells are in a specific state of differentiation. In this report, we show that the lectin Dolichos biflorus agglutinin (DBA), hitherto regarded and used as a general marker of developing renal collecting ducts, binds to most of the duct system but does not bind to the very tips of growing branches. The zone avoided by DBA corresponds to the zone that expresses Wnt11, and the zone that shows enhanced cell proliferation. If branching of the ureteric bud of cultured embryonic kidneys is inhibited in organ culture, by blocking the kidney's endogenous glial cell-derived neurothrophic factor (GDNF)-based branch-promoting signals, the DBA-binding zone extends to the very end of the tip but is lost from there when branching is re-activated. Similarly, if excess GDNF is provided to growing kidneys, the DBA-free zone expands. DBA-staining status therefore appears to be a sensitive indicator of the morphogenetic activity of the collecting duct system.
Collapse
Affiliation(s)
- Lydia Michael
- Centre for Integrative Physiology, Edinburgh University College of Medicine, Edinburgh, UK
| | | | | |
Collapse
|
25
|
Phillips JK, Hopwood D, Loxley RA, Ghatora K, Coombes JD, Tan YS, Harrison JL, McKitrick DJ, Holobotvskyy V, Arnolda LF, Rangan GK. Temporal relationship between renal cyst development, hypertension and cardiac hypertrophy in a new rat model of autosomal recessive polycystic kidney disease. Kidney Blood Press Res 2007; 30:129-44. [PMID: 17446713 DOI: 10.1159/000101828] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2006] [Accepted: 02/06/2007] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND/METHODS We have examined the hypothesis that cyst formation is key in the pathogenesis of cardiovascular disease in a Lewis polycystic kidney (LPK) model of autosomal-recessive polycystic kidney disease (ARPKD), by determining the relationship between cyst development and indices of renal function and cardiovascular disease. RESULTS In the LPK (n = 35), cysts appear at week 3 (1.1 +/- 0.1 mm) increasing to week 24 (2.8 +/- 2 mm). Immunostaining for nephron-specific segments indicate cysts develop predominantly from the collecting duct. Cyst formation preceded hypertension (160 +/- 22 vs. Lewis control 105 +/- 20 mm Hg systolic blood pressure (BP), n = 12) at week 6, elevated creatinine (109 +/- 63 vs. 59 +/- 6 micromol/l, n = 16) and cardiac mass (0.7 vs. 0.4% bodyweight, n = 15) at week 12, and left ventricular hypertrophy (2,898 +/- 207 vs. 1,808 +/- 192 mum, n = 14) at week 24 (all p < or = 0.05). Plasma-renin activity and angiotensin II were reduced in 10- to 12-week LPK (2.2 +/- 2.9 vs. Lewis 11.9 +/- 4.9 ng/ml/h, and 25.0 +/- 19.1 vs. 94.9 +/- 64.4 pg/ml, respectively, n = 26, p < or = 0.05). Ganglionic blockade (hexamethonium 3.3 mg/kg) significantly reduced mean BP in the LPK (52 vs. Lewis 4%, n = 9, p < or = 0.05). CONCLUSION Cyst formation is a key event in the genesis of hypertension while the sympathetic nervous system is important in the maintenance of hypertension in this model of ARPKD.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal
- Biomarkers/metabolism
- Creatinine/blood
- Disease Models, Animal
- Female
- Hypertension, Renal/etiology
- Hypertension, Renal/genetics
- Hypertension, Renal/pathology
- Hypertrophy, Left Ventricular/etiology
- Hypertrophy, Left Ventricular/genetics
- Hypertrophy, Left Ventricular/pathology
- Immunohistochemistry
- Kidney Cortex/innervation
- Kidney Cortex/metabolism
- Kidney Cortex/pathology
- Kidney Medulla/innervation
- Kidney Medulla/metabolism
- Kidney Medulla/pathology
- Kidney Tubules, Distal/innervation
- Kidney Tubules, Distal/metabolism
- Kidney Tubules, Distal/pathology
- Kidney Tubules, Proximal/innervation
- Kidney Tubules, Proximal/metabolism
- Kidney Tubules, Proximal/pathology
- Liver/metabolism
- Liver/pathology
- Male
- Myocardium/metabolism
- Myocardium/pathology
- Polycystic Kidney, Autosomal Recessive/complications
- Polycystic Kidney, Autosomal Recessive/genetics
- Polycystic Kidney, Autosomal Recessive/pathology
- Predictive Value of Tests
- Rats
- Rats, Inbred Lew
- Rats, Mutant Strains
- Renin-Angiotensin System/physiology
- Sympathetic Nervous System/drug effects
- Sympathetic Nervous System/physiology
- Sympatholytics/pharmacology
- Time Factors
- Urea/blood
Collapse
Affiliation(s)
- Jacqueline K Phillips
- Division of Health Sciences, School of Veterinary and Biomedical Science, Murdoch University, Perth, Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Masyuk TV, Masyuk AI, Torres VE, Harris PC, Larusso NF. Octreotide inhibits hepatic cystogenesis in a rodent model of polycystic liver disease by reducing cholangiocyte adenosine 3',5'-cyclic monophosphate. Gastroenterology 2007; 132:1104-16. [PMID: 17383431 DOI: 10.1053/j.gastro.2006.12.039] [Citation(s) in RCA: 192] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2006] [Accepted: 11/30/2007] [Indexed: 12/25/2022]
Abstract
BACKGROUND AND AIMS In polycystic liver diseases (PCLDs), increased cholangiocyte proliferation and fluid secretion are key features and cholangiocyte adenosine 3',5'-cyclic monophosphate (cAMP) is an important regulator of these processes. Thus, we assessed cAMP levels and evaluated octreotide (an analogue of somatostatin known to inhibit cAMP) in hepatic cyst growth using an in vitro model of cystogenesis and an in vivo animal model of autosomal recessive polycystic kidney disease (ARPKD), one of the PCLDs. METHODS Expression of somatostatin receptors (SSTRs) was assessed by reverse-transcription polymerase chain reaction and confocal microscopy in cholangiocytes from normal and polycystic kidney (PCK) rats, the ARPKD model of autosomal recessive polycystic kidney disease. Effects of octreotide on cAMP levels and cyst expansion were studied in vitro using PCK bile ducts grown in 3-dimensional culture. The effects of octreotide on hepatic and renal cystogenesis were investigated in PCK rats in vivo. RESULTS In cholangiocytes and serum of PCK rats, cAMP concentrations were approximately 2 times higher than in normal rats. SSTR subtypes that bind octreotide (ie, SSTR2, SSTR3, and SSTR5) were expressed in both normal and PCK cholangiocytes. In vitro, octreotide inhibited cAMP levels by 35% and reduced cyst growth by 44%. In vivo, octreotide lowered cAMP content in cholangiocytes and serum by 32%-39% and inhibited hepatic disease progression, leading to 22%-60% reductions in liver weight, cyst volume, hepatic fibrosis, and mitotic indices. Similar effects were observed in kidneys of PCK rats. CONCLUSIONS This preclinical study provides a strong rationale for assessing the potential value of octreotide in the treatment of PCLDs.
Collapse
Affiliation(s)
- Tatyana V Masyuk
- Center for Basic Research in Digestive Diseases, Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota 55905, USA
| | | | | | | | | |
Collapse
|
27
|
Abstract
When evaluating renal cystic mass lesions, ultrasound plays an important part in distinguishing cysts from solid lesions and in demonstrating the complex internal architecture of cystic lesions. Ultrasound is usually the initial imaging modality for patients who have medical renal disease. When renal cystic disease is identified, it is extremely important to accurately communicate the findings and attempt to distinguish benign from surgical lesions.
Collapse
Affiliation(s)
- Therese M Weber
- Department of Radiology, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157-1088, USA.
| |
Collapse
|
28
|
Harris PC, Torres VE. Understanding pathogenic mechanisms in polycystic kidney disease provides clues for therapy. Curr Opin Nephrol Hypertens 2006; 15:456-63. [PMID: 16775462 DOI: 10.1097/01.mnh.0000232888.65895.e7] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Polycystic kidney diseases are a group of inherited disorders that result in tubular dilatation and/or the development of fluid-filled cysts in the kidney. Identification and analysis of the primary defective protein in many of these diseases are providing insights into a common pathogenesis to polycystic kidney disease. This review explores this pathogenesis and determines the role that this knowledge is playing in the development of potential therapies. RECENT FINDINGS Study of simple and syndromic forms of polycystic kidney disease has revealed that the defective proteins are localized to the primary cilia/basal body and that the pleiotropic phenotypes are often associated with defective ciliogenesis. Data indicating that the polycystins are involved in ciliary mechanosensation, and cellular changes in intracellular Ca and cAMP, have provided clues for possible therapeutic approaches that have proved highly effective in pre-clinical trials. SUMMARY Polycystic kidney diseases are associated with defects to proteins involved in developing functional, sensory cilia in the kidney. While the primary defects in these disorders cannot be corrected at present, downsteam cellular changes can be targeted. Potential therapeutic agents are now being tested in patients, moving polycystic kidney disease research into a new and exciting phase.
Collapse
Affiliation(s)
- Peter C Harris
- Mayo Clinic College of Medicine, Rochester, Minnesota 55905, USA.
| | | |
Collapse
|
29
|
Sato Y, Harada K, Furubo S, Kizawa K, Sanzen T, Yasoshima M, Ozaki S, Isse K, Sasaki M, Nakanuma Y. Inhibition of intrahepatic bile duct dilation of the polycystic kidney rat with a novel tyrosine kinase inhibitor gefitinib. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 169:1238-50. [PMID: 17003482 PMCID: PMC1698840 DOI: 10.2353/ajpath.2006.051136] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The polycystic kidney (PCK) rat represents a liver and kidney cyst pathology corresponding to Caroli's disease with congenital hepatic fibrosis and autosomal recessive polycystic kidney disease. We previously reported that an epidermal growth factor receptor tyrosine kinase inhibitor, gefitinib (Iressa), significantly inhibited the abnormal growth of biliary epithelial cells of PCK rats in vitro. This study investigated the effects of gefitinib on cyst pathogenesis of the PCK rat both in vitro and in vivo. A three-dimensional culture model of biliary epithelial cells in the collagen gel matrix was used for in vitro analysis. For in vivo experiments, PCK and control rats were treated with gefitinib between 3 and 10 weeks of age. In vitro, gefitinib had strong inhibitory effects on biliary cyst formation of PCK rats. In vivo, treatment with gefitinib significantly inhibited the cystic dilatation of the intrahepatic bile ducts of PCK rats, which was accompanied by improvement of liver fibrosis. By contrast, no beneficial effects were observed on renal cyst development because of the treatment. These results suggest that signaling pathways mediated by epidermal growth factor receptor are involved in biliary dysgenesis of the PCK rat, with the mechanisms of cyst progression being different between the liver and kidney.
Collapse
Affiliation(s)
- Yasunori Sato
- Department of Human Pathology, Kanazawa University, Graduate School of Medicine, Kanazawa 920-8640, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
This article cannot comprehensively cover the enormous strides made in defining the molecular and cellular basis of renal cystic diseases over the last decade. Therefore, it provides a brief overview and categorization of inherited, developmental, and acquired renal cystic diseases, providing a relevant, up-to-date bibliography as well as a useful list of informative Internet Web sites. Its major focus is the translational biology of polycystic kidney disease. It demonstrates how emerging molecular and cellular knowledge of the pathophysiology of particular diseases such as autosomal dominant polycystic kidney disease (ADPKD) and autosomal recessive polycystic kidney disease (ADPKD) can translate into innovative therapeutic insights.
Collapse
Affiliation(s)
- Ellis D Avner
- Children's Research Institute, Children's Hospital & Health System of Wisconsin, and Medical College of Wisconsin, Department of Pediatrics, Division of Pediatrics, 8701 Watertown Plank Road, Milwaukee 53225, USA.
| | | |
Collapse
|
31
|
Sweeney WE, Avner ED. Molecular and cellular pathophysiology of autosomal recessive polycystic kidney disease (ARPKD). Cell Tissue Res 2006; 326:671-85. [PMID: 16767405 DOI: 10.1007/s00441-006-0226-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2006] [Accepted: 04/20/2006] [Indexed: 12/19/2022]
Abstract
Autosomal recessive polycystic kidney disease (ARPKD) belongs to a group of congenital hepatorenal fibrocystic syndromes characterized by dual renal and hepatic involvement of variable severity. Despite the wide clinical spectrum of ARPKD (MIM 263200), genetic linkage studies indicate that mutations at a single locus, PKHD1 (polycystic kidney and hepatic disease 1), located on human chromosome region 6p21.1-p12, are responsible for all phenotypes of ARPKD. Identification of cystic disease genes and their encoded proteins has provided investigators with critical tools to begin to unravel the molecular and cellular mechanisms of PKD. PKD cystic epithelia share common phenotypic abnormalities despite the different genetic mutations that underlie the disease. Recent studies have shown that many cyst-causing proteins are expressed in multimeric complexes at distinct subcellular locations within epithelia. This co-expression of cystoproteins suggests that cyst formation, regardless of the underlying disease gene, results from perturbations in convergent and/or integrated signal transduction pathways. To date, no specific therapies are in clinical use for ameliorating cyst growth in ARPKD. However, studies noted in this review suggest that therapeutic targeting of the cAMP and epidermal growth factor receptor (EGFR)-axis abnormalities in cystic epithelia may translate into effective therapies for ARPKD and, by analogy, autosomal dominant polycystic kidney disease (ADPKD). A particularly promising approach appears to be the targeting of downstream intermediates of both the cAMP and EGFR axis. This review focuses on ARPKD and presents a concise summary of the current understanding of the molecular genetics and cellular pathophysiology of this disease. It also highlights phenotypic and mechanistic similarities between ARPKD and ADPKD.
Collapse
Affiliation(s)
- William E Sweeney
- Children's Research Institute, Children's Hospital Health System of Wisconsin, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | |
Collapse
|
32
|
Wang Q, Pan J, Snell WJ. Intraflagellar Transport Particles Participate Directly in Cilium-Generated Signaling in Chlamydomonas. Cell 2006; 125:549-62. [PMID: 16678098 DOI: 10.1016/j.cell.2006.02.044] [Citation(s) in RCA: 127] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2005] [Revised: 11/18/2005] [Accepted: 02/07/2006] [Indexed: 12/27/2022]
Abstract
Primary cilia are widely used for signal transduction during development and in homeostasis and are assembled and maintained by intraflagellar transport (IFT). Here, we have dissected the role of IFT in signaling within the flagella (structural and functional counterparts of cilia) of the biflagellated green alga Chlamydomonas. Using a conditional IFT mutant enables us to deplete the IFT machinery from intact, existing flagella. We identify a cGMP-dependent protein kinase (CrPKG) within flagella as the substrate of a protein tyrosine kinase activated by flagellar adhesion during fertilization. We demonstrate that flagellar adhesion stimulates association of CrPKG with a new flagellar compartment. Moreover, formation of the compartment requires IFT, and IFT particles themselves are part of the compartment. Our results lead to a model in which the IFT machinery is required not only for assembling cilia and flagella but also for organizing a signaling pathway within the organelles during cilium-generated signaling.
Collapse
Affiliation(s)
- Qian Wang
- Department of Cell Biology, University of Texas Southwestern Medical School, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | | | | |
Collapse
|
33
|
Prelog M, Bergmann C, Ausserlechner MJ, Fischer H, Margreiter R, Gassner I, Brunner A, Jungraithmayr TC, Zerres K, Sergi C, Sergi EC, Zimmerhackl LB. Successful transplantation in a child with rapid progression of autosomal recessive polycystic kidney disease associated with a novel mutation. Pediatr Transplant 2006; 10:362-6. [PMID: 16677362 DOI: 10.1111/j.1399-3046.2005.00464.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Autosomal recessive polycystic kidney disease (ARPKD) is the most common pediatric renal cystic disease with liver involvement. The vast majority of patients with ARPKD carry mutations in the recently characterized PKHD1 gene on chromosome 6p12. A Turkish female demonstrated rapid growth of both kidneys after delivery. Accelerated growth of both kidneys and increasing respiratory distress necessitated right-sided nephrectomy at the age of three months. Because of persistent dyspnea and ongoing growth of the remaining kidney, the second kidney also had to be removed one month later. Biopsies taken from the kidney and the liver confirmed the diagnosis of ARPKD histologically. Renal ultrasound of the patient's consanguineous parents and her older brother showed normal results. PKHD1 mutation analysis yielded a novel homozygous missense mutation (c.1116C >G, F372L) in exon 14, coding for an Ig-like domain (TIG), possibly involved in the increased growth of the kidneys. Peritoneal dialysis was performed for 12 months. The patient had successful transplantation at the age of 15 months and is doing well with actual immunosuppression with cyclosporine, mycophenolate mofetil, and prednisolone. In conclusion, the present case clearly demonstrates the favorable outcome of a child with severe ARPKD after bilateral nephrectomy, pre-emptive dialysis, and successful transplantation.
Collapse
Affiliation(s)
- Martina Prelog
- Department of Pediatrics, Medical University Innsbruck, Innsbruck, Austria
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
|
35
|
Abstract
This review aims to assist in the categorization of inherited, developmental, and acquired cystic disease of the kidney as well as to provide a pertinent, up-to-date bibliography. The conditions included are autosomal-dominant polycystic kidney disease, autosomal-recessive polycystic kidney disease, unilateral renal cystic disease (localized cystic disease), renal simple cysts, multicystic dysplastic kidney, pluricystic kidney of the multiple malformation syndromes, juvenile nephronophthisis and medullary cystic disease, medullary sponge kidney, primary glomerulocystic kidney disease, and glomerulocystic kidney associated with several systemic disorders mainly of genetic or chromosomal etiology, cystic kidney in tuberous sclerosis, and in von Hippel-Lindau syndrome, cystic nephroma, cystic variant of congenital mesoblastic nephroma, mixed epithelial stromal tumor of the kidney, renal lymphangioma, pyelocalyceal cyst, peripylic cyst and perinephric pseudocyst, acquired renal cystic disease of long-term dialysis, and cystic renal cell carcinoma and sarcoma. Whereas the gross and histologic appearance of some of these conditions may be diagnostic, clinical and sometimes molecular studies may be necessary to define other types.
Collapse
Affiliation(s)
- Michele Bisceglia
- Division of Anatomic Pathology, IRCCS Casa Sollievo della Sofferenza Hospital, I-71013 San Giovanni Rotondo (FG), Italy.
| | | | | | | | | |
Collapse
|
36
|
Nagano J, Kitamura K, Hujer KM, Ward CJ, Bram RJ, Hopfer U, Tomita K, Huang C, Miller RT. Fibrocystin interacts with CAML, a protein involved in Ca2+ signaling. Biochem Biophys Res Commun 2005; 338:880-9. [PMID: 16243292 DOI: 10.1016/j.bbrc.2005.10.022] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2005] [Accepted: 10/05/2005] [Indexed: 11/30/2022]
Abstract
The predicted structure of the autosomal recessive polycystic kidney disease protein, fibrocystin, suggests that it may function as a receptor, but its function remains unknown. To understand its function, we searched for proteins that interact with the intracellular C-terminus of fibrocystin using the yeast two-hybrid system. From the screening, we found calcium modulating cyclophilin ligand (CAML), a protein involved in Ca(2+) signaling. Immunofluorescent analysis showed that both proteins are co-localized in the apical membrane, primary cilia, and the basal body of cells derived from the distal nephron Epitope-tagged expression constructs of both proteins were co-immunoprecipitated from COS7 cells. The intracellular C-terminus of fibrocystin interacts with CAML, a protein with an intracellular distribution that is similar to that of PKD2. Fibrocystin may participate in regulation of intracellular Ca(2+) in the distal nephron in a manner similar to PKD1 and PKD2 that are involved in autosomal dominant polycystic kidney disease.
Collapse
Affiliation(s)
- Junko Nagano
- Division of Nephrology, Department of Medicine, Case Western Reserve University, Lois Stokes Veterans Affairs Medical Center, Cleveland, OH, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Hepatic complications occur in a significant proportion of children with autosomal recessive polycystic kidney disease (ARPKD). PKHD1/fibrocystin, the defective gene in ARPKD, is expressed in the cilia of bile duct epithelium and leads to abnormalities in the rubric of the ductal plate malformation. Portal hypertension and biliary disease are the major liver problems seen in ARPKD. Complete blood counting, physical examination, ultrasonography and magnetic resonance (MR) cholangiography are indicated as screening procedures for hepatic disease in ARPKD. Medical and surgical interventions are potentially indicated for children with portal hypertension and/or biliary disease. A high index of suspicion for the diagnosis of cholangitis needs to be maintained in children with biliary disease. The implications of hepatic disease need to be considered in the decision-making regarding renal transplantation in ARPKD.
Collapse
|
38
|
Trizzino A, Farruggia P, Russo D, D'Angelo P, Tropia S, Benigno V, Tarantino G, Di Marco V, Aricò M. Congenital hepatic fibrosis: a very uncommon cause of pancytopenia in children. J Pediatr Hematol Oncol 2005; 27:567-8. [PMID: 16217264 DOI: 10.1097/01.mph.0000184577.46458.7e] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The disease presentation of autosomal recessive polycystic kidney disease (OMIM #263200, ARPKD) is highly variable and includes polycystic kidneys, pulmonary hypoplasia, and congenital hepatic fibrosis. The authors report an unusual case of ARPKD presenting with hepatosplenomegaly and cytopenia mimicking acute leukemia.
Collapse
Affiliation(s)
- Antonino Trizzino
- Onco Ematologia Pediatrica, Ospedale dei Bambini G. Di Cristina, Palermo, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Zimmermann A. The emerging family of hepatoblastoma tumours: from ontogenesis to oncogenesis. Eur J Cancer 2005; 41:1503-14. [PMID: 15982867 DOI: 10.1016/j.ejca.2005.02.035] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2004] [Revised: 12/13/2004] [Accepted: 02/25/2005] [Indexed: 01/03/2023]
Abstract
The identification of distinct types and subtypes of hepatoblastoma has led to a successful classification of these lesions. In recent years, and particularly within large tumour trials, the spectrum of paediatric epithelial liver tumours has increased. This, together with the need for defining clinically relevant risk groups, will require a new approach to defining and classifying these cancers. Furthermore, an impressive amount of molecular biological information on liver ontogenesis and growth regulation of hepatic tumours has recently accumulated, which will allow the development of a comprehensive classification system with particular emphasis on prognostics. In this review, novel findings relating to these issues are discussed.
Collapse
Affiliation(s)
- Arthur Zimmermann
- SIOPEL Liver Tumour Study Group, Institute of Pathology of the University, Berne, Switzerland.
| |
Collapse
|
40
|
Consugar MB, Anderson SA, Rossetti S, Pankratz VS, Ward CJ, Torra R, Coto E, El-Youssef M, Kantarci S, Utsch B, Hildebrandt F, Sweeney WE, Avner ED, Torres VE, Cunningham JM, Harris PC. Haplotype analysis improves molecular diagnostics of autosomal recessive polycystic kidney disease. Am J Kidney Dis 2005; 45:77-87. [PMID: 15696446 DOI: 10.1053/j.ajkd.2004.09.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Autosomal recessive polycystic kidney disease (ARPKD) is characterized by wide phenotypic variability, ranging from in utero detection with enlarged, echogenic kidneys to an adult presentation with congenital hepatic fibrosis. The ARPKD gene, PKHD1 , covers about 470 kb of DNA (67 exons), and mutation studies have found marked allelic heterogeneity with a high level of novel missense changes and neutral polymorphisms. To improve the prospects for molecular diagnostics and to study the origin of some relatively common mutations, the authors have developed a strategy for improved ARPKD haplotyping. METHODS A protocol of multiplex PCR and fluorescence genotyping in a single capillary has been developed to assay 7 highly informative simple sequence repeat (SSR) markers that are intragenic or closely flanking PKHD1. RESULTS Examples in which haplotype analysis, used in combination with mutation screening, improved the utility of molecular diagnostics, especially in families in which just a single PKHD1 mutation has been identified, are illustrated. The new markers also allow screening for larger DNA deletions, detecting unknown consanguinity and exploring the disease mechanism. Analysis of 8 recurring mutations has shown likely common haplotypes for each, and the divergence from the ancestral haplotype, by recombination, can be used to trace the history of the mutation. The common mutation, T36M, was found to have a single European origin, about 1,225 years ago. CONCLUSION Improved haplotype analysis of ARPKD complements mutation-based diagnostics and helps trace the history of common PKHD1 mutations.
Collapse
Affiliation(s)
- Mark B Consugar
- Division of Nephrology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Loghman-Adham M, Soto CE, Inagami T, Sotelo-Avila C. Expression of components of the renin-angiotensin system in autosomal recessive polycystic kidney disease. J Histochem Cytochem 2005; 53:979-88. [PMID: 15879580 DOI: 10.1369/jhc.4a6494.2005] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Hypertension is a common complication in children with autosomal recessive polycystic kidney disease (ARPKD) who have survived the neonatal period. No information is available regarding the mechanism of hypertension in this condition. The renin-angiotensin system (RAS) is thought to play a role in hypertension associated with the more common autosomal dominant polycystic kidney disease (ADPKD). Occasional reports have documented increased activity of the intrarenal RAS in ADPKD, with ectopic renin expression within cysts and dilated tubules. Because of similarities between ARPKD and ADPKD, we hypothesized that increased intrarenal RAS activity might also be found in ARPKD. We performed immunohistochemical studies on kidney tissues from two infants with ARPKD and two control kidneys. The cystic dilated tubules showed staining with the peanut lectin arachis hypogaea, a marker of distal tubules and collecting ducts, but not with lotus tetragonolobus, a marker of proximal tubules. Strong renin staining was seen in many cysts and tubules of ARPKD kidneys, but only in the afferent arterioles of the normal control kidneys. Angiotensinogen staining was also observed in some cysts and in proximal tubules. Staining for angiotensin-converting enzyme, angiotensin II type 1 receptor, and angiotensin II peptide was present in many cystic dilated tubules. These immunohistochemical studies document for the first time ectopic expression of components of the RAS in cystic-dilated tubules of ARPKD and suggest that overactivity of RAS could result in increased intrarenal angiotensin II production, which may contribute to the development of hypertension in ARPKD.
Collapse
Affiliation(s)
- Mahmoud Loghman-Adham
- Department of Pediatrics and Pediatric Research Institute, Saint Louis University, St. Louis, Missouri, USA.
| | | | | | | |
Collapse
|
42
|
Roume J, Ville Y. Prenatal diagnosis of genetic renal diseases: breaking the code. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2004; 24:10-18. [PMID: 15229910 DOI: 10.1002/uog.1109] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Affiliation(s)
- J Roume
- Department of Medical Genetics, Université UVSQ-Paris Ouest, CHI Poissy, France
| | | |
Collapse
|