Submit Paper

Article Processing Fee

Pay Online

           

Crossref logo

  DOI Prefix   10.20431


 

ARC Journal of Nephrology
Volume-3 Issue-1, 2018, Page No: 13-18

Wnt Signaling Pathway in Polycystic Kidney Disease

Ao Li

Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA.

Citation : Ao Li, "Wnt Signaling Pathway in Polycystic Kidney Disease" ARC Journal of Nephrology. 2018; 3(1) : 13-18.

Copyright : © 2018 . This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.


Abstract:

Wnt signaling involves a variety of signaling cascades that can be activated by secreted Wnt ligands. Two major Wnt pathways, the canonical or β‐catenin pathway and the planar cell polarity (PCP) pathway, have recently been demonstrated that plays important roles in multiple cellular processes within the kidney. Both of these pathways are essential for kidney development as well as homeostasis and injury repair. The disruption of Wnt pathway will result in cystic kidney disease, a group of genetic diseases that includes the most common hereditary life‐threatening syndrome polycystic kidney disease (PKD). Recent studies implicate canonical and noncanonical Wnt pathways in cystogenesis and points to a remarkable role for developmental processes in the adult kidney.


Keywords: Wnt Signaling, Polycystic kidney disease,Nephrology


1. Polycystic Kidney Disease


Polycystic kidney disease (PKD) is an inherited disorder in which the renal tubules become structurally abnormal, resulting in the development and growth of multiple cysts within the kidney [1]. These cysts may begin to develop before birth or in infancy, in childhoOd, or in adulthood [2]. Cysts are non‐ functioning tubules filled with fluid pumped into them, which range in size from microscopic to enormous, crushing adjacent normal tubules and eventually rendering them non‐functional also. The two main types of polycystic kidney disease are: autosomal dominant polycystic kidney disease (ADPKD) and autosomal recessive polycystic kidney disease (ARPKD).

Autosomal dominant polycystic kidney disease (ADPKD) is the most common of a group of inherited kidney disorders characterized by progressive cyst development and various extrarenal manifestations [3,4]. Cystogenesis in human kidneys progressively occupy the normal parenchyma of the kidney and lead to renal failure, which usually occurs in mid‐to‐late adulthood. This disease is the fourth most common single cause of end‐stage renal failure worldwide [5,6].

ADPKD is caused by mutations in the PKD1 or PKD2 genes, which encode the proteins polycystin‐1 (PC1) and polycystin‐2 (PC2), respectively. Approximately 85% of ADPKD patients have mutations in PKD1, and the remaining 15% have mutations in PKD2 [7,8].

The most common extra renal manifestation of ADPKD is the formation of bile‐duct‐derived cysts in the liver [4,9]. Liver cysts occur in 83% of all ADPKD patients, and 94% of the patients with liver cysts are over 35 years old [10,11]. Other ADPKD phenotypes include pancreatic cysts [12,13], aneurysms [14-17], and aortic root/thoracic aorta abnormalities [18-20].Autosomal recessive polycystic kidney disease (ARPKD) is one of the most common hereditary renal cystic diseases in infants and children, with an estimated incidence of ~1 in 20,000 live births and a prevalence of heterozygous carriers of ~1 in 70 [21,22]. The disease is caused by mutations in PKHD1, which encodes a 16‐kb transcript, contains at least 86 exons, and spans 470 kb on chromosome 6p12 [23]. The longest ORF is predicted to be 66 exons and yields a 4074‐amino acid membrane‐associated receptor‐like protein, fibrocystin/polyductin (FPC) [24-28]. Its major clinical manifestations include fusi form ectasia of the renal collecting and hepatic biliary ducts and fibrosis of the liver and kidneys [29-31], although the renal lesions predominate at the time of diagnosis [31,32]. Approximately 50% of ARPKD patients present as neonates [33] when they are born with dramatically enlarged, symmetric kidneys and ectasia of the collecting duct [34,35]. The mortality rate is 30‐50% for neonates due to respiratory and/or renal dysfunction [36].


2. Wnt Signaling Pathway


Wnt protein family is a class of highly conserved evolutionarily secreting glycoproteins involved in cell proliferation, survival, differentiation, polarity, cell fate determination in embryonic development and homeostasis of adult tissues [37]. Wnt signaling pathway includes the canonical Wnt/β‐catenin signaling pathway and non‐canonical pathway [38]. In the canonical Wnt pathway, two cell membrane proteins, Frizzled (FZD) and LDL‐receptor related protein 5/6 (LRP5/6), function together as receptors for the Wnt ligands. In the absence of a Wnt signal, β‐catenin is continuously phosphorylated by a multiprotein destruction complex, which includes Axin, adenomatous polyposis coli (APC), casein kinase 1α (CK1α) and glycogen synthase kinase 3β (GSK3β). Phosphorylated β‐catenin is targeted for proteasomal degradation. As soon as Wnts binds to their receptors Frizzled and LRP5/6, the destruction complex function becomes disrupted, which in turn promotes β‐catenin accumulation and translocation to the nucleus. In the nucleus, β‐catenin forms a complex with TCF/LEF transcription factors that regulate target gene transcription such as Cyclin D1, c‐Jun, c‐Myc [39].

Non‐canonical Wnt signaling pathway mainly refers to Wnt/Ca2+ signaling pathway and Wnt/PCP signaling pathway. Non‐canonical Wnt signaling controls cell polarity through activation of RhoGTPase or via β‐ catenin‐independent mechanisms to increase intracellular Ca2+ concentration. Wnt/PCP pathway regulates small GTPaseRhoA and c‐Jun N‐terminal kinase (JNK), activates RhoGTPase to cause cytoskeleton and microtubule convergent extension which is crucial for cell shape and adhesion function of epithelial cells. Wnt/Ca2+ pathway regulates the movement of cells in the early stages of development. Frizzled appears to activate phospholipase C and phosphodiesterase to increase cells intracellular calcium and reduce intracellular cyclic guanosine (cGMP) concentrations [40]. Canonical and non‐canonical Wnt signaling are involved in the regulation of kidney development development as well as homeostasis and injury repair [41].


3. Pkd And Wnt Signaling


There has been considerable progress in elucidating the molecular mechanisms and pathogenesis of ADPKD [5,7,42-44]. Previous studies showed that human cystic disease may involve Wnt signal transduction [41,45,46]. In the kidney, canonical Wnt signaling is indispensible for induction of the metanephric mesenchyme and ureteric bud branching. The noncanonical Wnt signaling regulates planar cell polarity, cell migration and mitotic spindle orientation. These processes are critical for proper tubular morphology. Hitherto, many reports have demonstrated that renal cystic disease may result from dysregulation of non‐canonical Wnt pathway by disrupting Wnt/Ca2+ signaling and/or PCP processes in renal epithelial cells [47-51].

For example, Kim et al., found that Wnts bind to the extracellular domain of polycystin‐1 and induce whole‐cell currents and Ca2+ influx dependent on polycystin‐2. Mutations of either polycystin‐1 or polycystin‐2 that disrupt complex formation, compromise cell surface expression of polycystin‐1, or decrease polycystin‐2 channel activity suppress activation by Wnts. Pkd2‐/‐ fibroblasts lack Wnt‐induced Ca2+ currents and are unable to polarize during directed cell migration [47]. Meanwhile, result from Luyten group indicated that cystic kidneys exhibited remarkable up regulation and activation of Frizzled 3 (Fz3), a regulator of PCP, and its downstream effector, CDC42. Fz3 was expressed on the cilia of cystic kidneys but barely detected on the cilia of normal kidneys. In vitro, polycystin‐1 and Fz3 antagonized each other to control CDC42 expression and the rate of cell migration in HEK293T cells. All their data suggested that polycystin‐1 controlled oriented cell division and that aberrant PCP signaling contributed to cystogenesis.

In spite of these findings, functional roles of canonical Wnt signaling in pathogenesis of ADPKD remain to be unequivocally defined. A transgenic mouse for β‐catenin, a key factor for canonical Wnt signaling, exhibited severe PKD phenotypes, showing that β‐catenin up regulation alone was sufficient to induce cyst formation in the kidney [52]. These mice developed severe polycystic lesions soon after birth that affected the glomeruli, proximal, distal tubules and collecting ducts. This phenotype was similar to the human ADPKD phenotype. Cyst formation was associated with an increase in cell proliferation and apoptosis. The cell proliferation and apoptotic indexes was increased 4‐5‐fold and 3‐4‐fold, respectively, in cystic tubules of the transgenic mice compared to that of littermate controls. Their findings provided experimental genetic evidence that activation of the Wnt/β‐catenin signaling pathway causes polycystic kidney disease and supported the view that dysregulation of the Wnt/β‐catenin signaling was involved in its pathogenesis. Another research group also demonstrated that overexpression of c‐Myc, a canonical Wnt targeting product, also induced cystogenesis in the kidney of mouse models [53]. This phenotype appeared to result from the overexpression of c‐Myc in the renal tubular epithelium and consequent abnormal cell proliferation. These animals reproducibly developed PKD and died of renal failure. Furthermore, Qian et al., found that disruptions of Apc, which was a co‐factor for the β‐catenin degradation complex, promoted cyst formation in the kidney [54]. They generated mice carrying a conditional deletion of the Apc tumor suppressor gene specifically in the renal epithelium. Loss of Apc accounted for upregulation of β‐catenin protein in renal epithelium. Most of these mice died shortly after birth, and multiple kidney cysts were found upon histological examination. This result illustrated that Wnt/β‐catenin signaling played essential role in renal development and provided evidence that dysregulation of the pathway can initiate tumorigenesis in the kidney.

Moreover, results from our and other groups suggested that aberrantly activating canonical Wnt signaling could be detected in spectrum of Pkd1‐ and Pkd2‐deficient cells and tissues [49,55,56]. The polycystin‐ 1 C‐terminal inhibited the ability of both β‐catenin and Wnt ligands to activate T‐cell factor (TCF) ‐ dependent gene transcription. DNA microarray analysis revealed that the canonical Wnt signaling pathway was activated in ADPKD patient cysts, suggesting that increased canonical Wnt signaling involved in cyst formation. Our previous data also confirmed that loss of polycystin‐2 disrupted normal behavior of renal epithelial cells through dysregulation of β‐catenin‐dependent signaling. Mutation of Pkd2 resulted in cystogenesis and upregulated β‐catenin expression. These findings directly or indirectly supported that hyperactivated canonical Wnt signaling may cause cyst formation in the kidney of animal models.

However, other reports indicated that increased canonical Wnt signaling may not play a key role in cystogenesis of PKD [50,57-60]. The opposite effect has been interpreted by which cyst formation might disturb Wnt/PCP homeostasis via losing balance between the canonical and non‐canonical Wnt activity [45,48,49,61-63].

To directly reveal functional role of canonical Wnt signaling in cystogenesis of ADPKD, we employed a standardized mouse ortholog of human ADPKD [44] to investigate the importance of β‐catenin in ADPKD phenotypes. Our data demonstrated that the elevated β‐catenin signaling caused by polycystin‐2 deficiency contributed to disease phenotypes in our mouse ortholog of human ADPKD. Pharmacologically inhibiting the β‐catenin stability or the production of mature Wnt protein, or genetically reducing the expression of Ctnnb1 (which encodes β‐catenin), suppressed the formation of renal cysts, improved renal function, and extended survival in ADPKD mice [64].


4. Conclusion And Perspective


Polycystic kidney diseases have been linked to aberrant Wnt signaling. Disruptions of cystic disease genes account for dysregulation of Wnt signaling in model organisms and cultured cells. Inappropriate levels of Wnt signaling result in renal cyst formation in mice. These observations have prompted the idea that abnormal Wnt signaling may implicate a common causative event in cyst formation. Wnt pathway has become a potential avenue for urgently required novel therapeutics for treating human kidney diseases.


References


  1. Wilson P. Polycystic kidney disease. N Engl J Med. 2004; 350(151‐64.
  2. Chapin HC and Caplan MJ. The cell biology of polycystic kidney disease. J Cell Biol. 2010; 191(4):701‐10.
  3. Grantham JJ and Torres VE. The importance of total kidney volume in evaluating progression of polycystic kidney disease. Nat Rev Nephrol. 2016; 12(11):667‐77.
  4. Gallagher AR, Germino GG, and Somlo S. Molecular advances in autosomal dominant polycystic kidney disease. Adv Chronic Kidney Dis. 2010; 17(2):118‐30.
  5. Harris PC and Torres VE. Genetic mechanisms and signaling pathways in autosomal dominant polycystic kidney disease. J Clin Invest. 2014; 124(6):2315‐24.
  6. Ong AC, Devuyst O, Knebelmann B, Walz G, and Diseases E‐EWGfIK. Autosomal dominant polycystic kidney disease: the changing face of clinical management. Lancet. 2015; 385 (9981): 1993‐2002.
  7. Fedeles SV, Gallagher AR, and Somlo S. Polycystin‐1: a master regulator of intersecting cystic pathways. Trends Mol Med. 2014; 20(5):251‐60.
  8. Igarashi P and Somlo S. Polycystic kidney disease. J Am SocNephrol. 2007; 18(5):1371‐3.
  9. Spirli C, Okolicsanyi S, Fiorotto R, Fabris L, Cadamuro M, Lecchi S, Tian X, Somlo S, and Strazzabosco M. Mammalian target of rapamycin regulates vascular endothelial growth factor‐ dependent liver cyst growth in polycystin‐2‐defective mice. Hepatology. 2010; 51(5):1778‐88.
  10. Bae KT, Zhu F, Chapman AB, Torres VE, Grantham JJ, Guay‐Woodford LM, Baumgarten DA, King BF, Jr., Wetzel LH, Kenney PJ, et al. Magnetic resonance imaging evaluation of hepatic cysts in early autosomal‐dominant polycystic kidney disease: the Consortium for Radiologic Imaging Studies of Polycystic Kidney Disease cohort. Clin J Am SocNephrol. 2006; 1(1):64‐9.
  11. Bae KT, and Grantham JJ. Imaging for the prognosis of autosomal dominant polycystic kidney disease. Nat Rev Nephrol. 2010; 6(2): 96‐106.
  12. Torra R, Nicolau C, Badenas C, Navarro S, Perez L, Estivill X, and Darnell A. Ultrasono graphic study of pancreatic cysts in autosomal dominant polycystic kidney disease. Clin Nephrol. 1997; 47(1):19‐ 22.
  13. Yazdanpanah K, Manouchehri N, Hosseinzadeh E, Emami MH, Karami M, and Sarrami AH. Recurrent acute pancreatitis and cholangitis in a patient with autosomal dominant polycystic kidney disease. Int J Prev Med. 2013; 4(2):233‐6.
  14. Ring T, and Spiegelhalter D. Risk of intracranial aneurysm bleeding in autosomal ‐ dominant polycystic kidney disease. Kidney Int. 2007; 72(11):1400‐2.
  15. Ong AC. Screening for intracranial aneurysms in ADPKD. BMJ. 2009; 339(7723):b3763.
  16. Xu HW, Yu SQ, Mei CL, and Li MH. Screening for intracranial aneurysm in 355 patients with autosomal‐dominant polycystic kidney disease. Stroke. 2011; 42(1):204‐6.
  17. Rozenfeld MN, Ansari SA, Mohan P, Shaibani A, Russell EJ, and Hurley MC. Autosomal Dominant Polycystic Kidney Disease and Intracranial Aneurysms: Is There an Increased Risk of Treatment? AJNR Am J Neuroradiol. 2016; 37(2):290‐3.
  18. Adeola T, Adeleye O, Potts JL, Faulkner M, and Oso A. Thoracic aortic dissection in a patient with autosomal dominant polycystic kidney disease. J Natl Med Assoc. 2001; 93(7‐8):282‐7.
  19. Liu D, Wang CJ, Judge DP, Halushka MK, Ni J, Habashi JP, Moslehi J, Bedja D, Gabrielson KL, Xu H, et al. A Pkd1‐Fbn1 Genetic Interaction Implicates TGF‐beta Signaling in the Pathogenesis of Vascular Complications in Autosomal Dominant Polycystic Kidney Disease. J Am SocNephrol. 2014; 25(1):81‐91.
  20. Perrone RD, Malek AM, and Watnick T. Vascular complications in autosomal dominant polycystic kidney disease. Nat Rev Nephrol. 2015; 11(10):589‐98.
  21. Guay‐Woodford LM. Autosomal recessive PKD in the early years. Nephrol News Issues. 2007; 21(12):39.
  22. Harris PC, and Torres VE. Polycystic kidney disease. Annu Rev Med. 2009;60(321‐37.
  23. Sharp AM, Messiaen LM, Page G, Antignac C, Gubler MC, Onuchic LF, Somlo S, Germino GG, and Guay‐Woodford LM. Comprehensive genomic analysis of PKHD1 mutations in ARPKD cohorts. JMed Genet. 2005; 42(4): 33 6‐49.
  24. Bergmann C, Frank V, Kupper F, Schmidt C, Senderek J, Zerres K. Functional analysis of PKHD1 splicing in autosomal recessive polycystic kidney disease. J Hum Genet 2006; 51(788‐93).
  25. Onuchic LF, Furu L, Nagasawa Y, Hou X, Eggermann T, Ren Z, Bergmann C, Senderek J, Esquivel E, Zeltner R, et al. PKHD1, the polycystic kidney and hepatic disease 1 gene, encodes a novel large protein containing multiple immune globulin‐like plexin‐ transcription‐factor domains and parallel beta‐helix 1 repeats. Am J Hum Genet. 2002; 70(5):1305‐17.
  26. Ward CJ, Hogan MC, Rossetti S, Walker D, Sneddon T, Wang X, Kubly V, Cunningham JM, Bacallao R, Ishibashi M, et al. The gene mutated in autosomal recessive polycystic kidney disease encodes a large, receptor‐like protein. Nat Genet. 2002; 30(3):259‐69.
  27. Xiong H, Chen Y, Yi Y, Tsuchiya K, Moeckel G, Cheung J, Liang D, Tham K, Xu X, Chen XZ, Pei Y, Zhao ZJ, Wu G. A Novel Gene Encoding a TIG Multiple Domain Protein Is a Positional Candidate for Autosomal Recessive Polycystic Kidney Disease. Genomics. 2002; 80(96‐104).
  28. Hu B, He X, Li A, Qiu Q, Li C, Liang D, Zhao P, Ma J, Coffey RJ, Zhan Q, et al. Cystogenesis in ARPKD results from increased apoptosis in collecting duct epithelial cells of Pkhd1 mutant kidneys. ExpCell Res. 2011; 317(2):173‐87.
  29. Lonergan GJ, Rice RR, and Suarez ES. Autosomal recessive polycystic kidney disease: radiologic‐ pathologic correlation. Radio graphics. 2000; 20(3):837‐55.
  30. Zerres K, Rudnik‐Schoneborn S, Steinkamm C, Becker J, and Mucher G. Autosomal recessive polycystic kidney disease. J Mol Med. 1998; 76(5):303‐9.
  31. Zerres K, Mucher G, Becker J, Steinkamm C, Rudnik‐Schoneborn S, Heikkila P, Rapola J, Salonen R, Germino GG, Onuchic L, et al. Prenatal diagnosis of autosomal recessive polycystic kidney disease (ARPKD): molecular genetics, clinical experience, and fetal morphology. Am J Med Genet. 1998; 76(2):137‐44.
  32. Guay‐Woodford LM, and Desmond RA. Autosomal recessive polycystic kidney disease: the clinical experience in North America. Pediatrics. 2003; 111(5 Pt 1):1072‐80.
  33. Capisonda R, Phan V, Traubuci J, Daneman A, Balfe JW, and Guay‐Woodford LM. Autosomal recessive polycystic kidney disease: outcomes from a single‐center experience. PediatrNephrol. 2003; 18(2):119‐26.
  34. Zerres K, Rudnik‐Schoneborn S, Steinkamm C, and Mucher G. Autosomal recessive polycystic kidney disease. Nephrol Dial Transplant. 1996; 11(Suppl 6):29‐33.
  35. Zerres K, Rudnik‐Schoneborn S, Senderek J, Eggermann T, and Bergmann C. Autosomal recessive polycystic kidney disease (ARPKD). J Nephrol. 2003; 16(3):453‐8.
  36. Bergmann C, Senderek J, Schneider F, Dornia C, Kupper F, Eggermann T, Rudnik‐Schoneborn S, Kirfel J, Moser M, Buttner R, Zerres K. PKHD1 mutations in families requesting prenatal diagnosis for autosomal recessive polycystic kidney disease (ARPKD). Hum Mutat. 2004; 23(487‐95).
  37. Clevers H, and Nusse R. Wnt/beta‐catenin signaling and disease. Cell. 2012; 149 (6): 1192 ‐ 205.
  38. Niehrs C. The complex world of WNT receptor signalling. Nat Rev Mol Cell Biol. 2012; 13(12):767‐ 79.
  39. Polakis P. Wnt signaling in cancer. Cold Spring HarbPerspect Biol. 2012; 4(5).
  40. Semenov MV, Habas R, Macdonald BT, and He X. SnapShot: NoncanonicalWnt Signaling Pathways. Cell. 2007; 131(7):1378.
  41. Benzing T, Simons M, and Walz G. Wnt signaling in polycystic kidney disease. J Am SocNephrol. 2007; 18(5):1389‐98.
  42. Ong AC, and Harris PC. A polycystin‐centric view of cyst formation and disease: the polycystins revisited. Kidney Int. 2015; 88(4):699‐710.
  43. Li A, Tian X, Zhang X, Huang S, Ma Y, Wu D, Moeckel G, Somlo S, and Wu G. Human Polycystin‐2 Transgene Dose‐Dependently Rescues ADPKD Phenotypes in Pkd2 Mutant Mice. Am J Pathol. 2015; 185(10):2843‐60.
  44. Li A, Fan S, Xu Y, Meng J, Shen X, Mao J, Zhang L, Zhang X, Moeckel G, Wu D, et al. Rapamycin treatment dose‐dependently improves the cystic kidney in a new ADPKD mouse model via the mTORC1 and cell‐cycle‐associated CDK1/cyclin axis. J Cell Mol Med. 2017; 21(8):1619‐35.
  45. Germino GG. Linking cilia to Wnts. Nat Genet. 2005; 37(5):455‐7.
  46. Song X, Di Giovanni V, He N, Wang K, Ingram A, Rosenblum ND, and Pei Y. Systems biology of autosomal dominant polycystic kidney disease (ADPKD): computational identification of gene expression pathways and integrated regulatory networks. Hum Mol Genet. 2009; 18(13):2328‐43.
  47. Kim S, Nie H, Nesin V, Tran U, Outeda P, Bai CX, Keeling J, Maskey D, Watnick T, Wessely O, et al. The polycystin complex mediates Wnt/Ca (2+) signalling. Nat Cell Biol. 2016; 18(7):752‐64.
  48. Luyten A, Su X, Gondela S, Chen Y, Rompani S, Takakura A, and Zhou J. Aberrant regulation of planar cell polarity in polycystic kidney disease. J Am SocNephrol. 2010; 21(9): 1521 ‐ 32.
  49. Happé H, Leonhard WN, van der Wal A, van de Water B, Lantinga‐van Leeuwen IS, Breuning MH, de Heer E, Peters DJ. Toxic tubular injury in kidneys from Pkd1‐deletion mice accelerates cystogenesis accompanied by dysregulated planar cell polarity and canonical Wnt signaling pathways. Hum Mol Genet 2009; 18(2532‐42).
  50. Karner CM, Chirumamilla R, Aoki S, Igarashi P, Wallingford JB, and Carroll TJ. Wnt9b signaling regulates planar cell polarity and kidney tubule morphogenesis. Nat Genet. 2009; 41(7):793‐9.
  51. Fischer E, Legue E, Doyen A, Nato F, Nicolas JF, Torres V, Yaniv M, and Pontoglio M. Defective planar cell polarity in polycystic kidney disease. Nat Genet. 2006; 38(1):21‐3.
  52. Saadi‐Kheddouci S, Berrebi D, Romagnolo B, Cluzeaud F, Peuchmaur M, Kahn A, Vandewalle A, and Perret C. Early development of polycystic kidney disease in transgenic mice expressing an activated mutant of the beta‐catenin gene. Oncogene. 2001; 20(42):5972‐81.
  53. Trudel M, D'Agati V, and Costantini F. C‐myc as an inducer of polycystic kidney disease in transgenic mice. Kidney Int. 1991; 39(4): 665 ‐ 71.
  54. Qian CN, Knol J, Igarashi P, Lin F, Zylstra U, Teh BT, and Williams BO. Cystic renal neoplasia following conditional inactivation of apc in mouse renal tubular epithelium. J Biol Chem. 2005; 280(5):3938‐45.
  55. Lal M, Song X, Pluznick JL, Di Giovanni V, Merrick DM, Rosenblum ND, Chauvet V, Gottardi CJ, Pei Y, and Caplan MJ. Polycystin‐1 C‐terminal tail associates with beta‐catenin and inhibits canonical Wnt signaling. Hum Mol Genet. 2008; 17(20): 3105 ‐17.
  56. Kim I, Ding T, Fu Y, Li C, Cui L, Li A, Lian P, Liang D, Wang DW, Guo C, et al. Conditional mutation of Pkd2 causes cystogenesis and upregulates beta‐catenin. J Am SocNephrol. 2009; 20(12):2556‐ 69.
  57. Miller MM, Iglesias DM, Zhang Z, Corsini R, Chu L, Murawski I, Gupta I, Somlo S, Germino GG, and Goodyer PR. T‐cell factor/beta‐catenin activity is suppressed in two different models of autosomal dominant polycystic kidney disease. Kidney Int. 2011; 80(2):146‐53.
  58. Sugiyama N, Tsukiyama T, Yamaguchi TP, and Yokoyama T. The canonical Wnt signaling pathway is not involved in renal cyst development in the kidneys of inv mutant mice. Kidney Int. 2011; 79(9):957‐65.
  59. Qin S, Taglienti M, Cai L, Zhou J, and Kreidberg JA. C-Met and NF‐kappa B‐ dependent over expression of Wnt7a and ‐7b and Pax2 promotes cystogenesis in polycystic kidney disease. JAm SocNephrol. 2012; 23(8):1309‐18.
  60. Cnossen WR, teMorsche RH, Hoischen A, Gilissen C, Venselaar H, Mehdi S, Bergmann C, Losekoot M, Breuning MH, Peters DJ, et al. LRP5 variants may contribute to ADPKD. Eur J Hum Genet. 2016; 24(2):237‐42.
  61. Lancaster MA, Louie CM, Silhavy JL, Sintasath L, Decambre M, Nigam SK, Willert K, and Gleeson JG. Impaired Wnt‐beta‐catenin signaling disrupts adult renal homeostasis and leads to cystic kidney ciliopathy. Nat Med. 2009; 15(9):1046‐54.
  62. Patel V. Balancing the Wnts in polycystic kidney disease. J Am Soc Nephrol. 2010; 21(9): 1412‐4.
  63. Wuebken A, and Schmidt‐Ott KM. WNT/ beta‐catenin signaling in polycystic kidney disease. KidneyInt. 2011; 80(2):135‐8.
  64. Li A, Xu Y, Fan S, Meng J, Shen X, Xiao Q, Li Y, Zhang L, Zhang X, Wu G, et al. Canonical Wnt inhibitors ameliorate cystogenesis in a mouse ortholog of human ADPKD. JCI Insight. 2018; 3(5).