127
Views
9
CrossRef citations to date
0
Altmetric
Review

Emerging treatments and personalised medicine for ciliopathies associated with cystic kidney disease

& ORCID Icon
Pages 785-798 | Received 14 Jul 2017, Accepted 24 Aug 2017, Published online: 31 Aug 2017

References

  • Wilson PD. Therapeutic targets for polycystic kidney disease. Expert Opin Ther Targets. 2016;20:35–45.
  • Hartung EA, Guay-Woodford LM. Autosomal recessive polycystic kidney disease: a hepatorenal fibrocystic disorder with pleiotropic effects. Pediatrics. 2014;134:e833–e845.
  • Srivastava S, Sayer JA. Nephronophthisis. J Pediatr Genet. 2014;3:103–114.
  • Hildebrandt F, Otto E. Cilia and centrosomes: a unifying pathogenic concept for cystic kidney disease? Nat Rev Genet. 2005;6:928–940.
  • Fry AM, Leaper MJ, Bayliss R. The primary cilium. Organogenesis. 2014;10:62–68.
  • Pan J, Seeger-Nukpezah T, Golemis EA. The role of the cilium in normal and abnormal cell cycles: emphasis on renal cystic pathologies. Cell Mol Life Sci. 2013;70:1849–1874.
  • Goggolidou P. Wnt and planar cell polarity signaling in cystic renal disease. Organogenesis. 2014;10:86–95.
  • Calvet JP. The role of calcium and cyclic AMP in PKD. In: Li X, editor. Polycystic kidney disease [Internet]. Brisbane (AU): Codon Publications; 2015. cited 2017 Aug 22. Available from: http://www.ncbi.nlm.nih.gov/books/NBK373395/
  • Christensen ST, Clement CA, Satir P, et al. Primary cilia and coordination of receptor tyrosine kinase (RTK) signalling. J Pathol. 2012;226:172–184.
  • Bishop GA, Berbari NF, Lewis J, et al. Type III adenylyl cyclase localizes to primary cilia throughout the adult mouse brain. J Comp Neurol. 2007;505:562–571.
  • Antal MC, Bénardais K, Samama B, et al. Adenylate cyclase type III is not a ubiquitous marker for all primary cilia during development. PLoS One. 2017;12:e0170756.
  • Händel M, Schulz S, Stanarius A, et al. Selective targeting of somatostatin receptor 3 to neuronal cilia. Neuroscience. 1999;89:909–926.
  • Raychowdhury MK, Ramos AJ, Zhang P, et al. Vasopressin receptor-mediated functional signaling pathway in primary cilia of renal epithelial cells. Am J Physiol Renal Physiol. 2009;296:F87–F97.
  • Iwanaga T, Miki T, Takahashi-Iwanaga H. Restricted expression of somatostatin receptor 3 to primary cilia in the pancreatic islets and adenohypophysis of mice. Biomed Res Tokyo Jpn. 2011;32:73–81.
  • Mick DU, Rodrigues RB, Leib RD, et al. Proteomics of primary cilia by proximity labeling. Dev Cell. 2015;35:497–512.
  • Boldt K, Van Reeuwijk J, Lu Q, et al. An organelle-specific protein landscape identifies novel diseases and molecular mechanisms. Nat Commun. 2016;7:11491.
  • Gattone VH, Maser RL, Tian C, et al. Developmental expression of urine concentration-associated genes and their altered expression in murine infantile-type polycystic kidney disease. Dev Genet. 1999;24:309–318.
  • Torres VE, Wang X, Qian Q, et al. Effective treatment of an orthologous model of autosomal dominant polycystic kidney disease. Nat Med. 2004;10:363–364.
  • Gattone VH, Wang X, Harris PC, et al. Inhibition of renal cystic disease development and progression by a vasopressin V2 receptor antagonist. Nat Med. 2003;9:1323–1326.
  • Wang X, Gattone V, Harris PC, et al. Effectiveness of vasopressin V2 receptor antagonists OPC-31260 and OPC-41061 on polycystic kidney disease development in the PCK rat. J Am Soc Nephrol. 2005;16:846–851.
  • Aihara M, Fujiki H, Mizuguchi H, et al. Tolvaptan delays the onset of end-stage renal disease in a polycystic kidney disease model by suppressing increases in kidney volume and renal injury. J Pharmacol Exp Ther. 2014;349:258–267.
  • Masyuk TV, Masyuk AI, Torres VE, et al. Octreotide inhibits hepatic cystogenesis in a rodent model of polycystic liver disease by reducing cholangiocyte adenosine 3′,5′-cyclic monophosphate. Gastroenterology. 2007;132:1104–1116.
  • Masyuk TV, Radtke BN, Stroope AJ, et al. Pasireotide is more effective than octreotide in reducing hepatorenal cystogenesis in rodents with polycystic kidney and liver diseases. Hepatology. 2013;58:409–421.
  • Sweeney WE, Chen Y, Nakanishi K, et al. Treatment of polycystic kidney disease with a novel tyrosine kinase inhibitor11See Editorial by Grantham, p. 339. Kidney Int. 2000;57:33–40.
  • Torres VE, Sweeney WE, Wang X, et al. EGF receptor tyrosine kinase inhibition attenuates the development of PKD in Han: SPRDrats. Kidney Int. 2003;64:1573–1579.
  • Sweeney WE, Hamahira K, Sweeney J, et al. Combination treatment of PKD utilizing dual inhibition of EGF-receptor activity and ligand bioavailability. Kidney Int. 2003;64:1310–1319.
  • Sweeney WE, Vigier RO, von Frost P, et al. Src inhibition ameliorates polycystic kidney disease. J Am Soc Nephrol. 2008;19:1331–1341.
  • Elliott J, Zheleznova NN, Wilson PD. c-Src inactivation reduces renal epithelial cell-matrix adhesion, proliferation, and cyst formation. Am J Physiol Cell Physiol. 2011;301:C522–C529.
  • Bukanov NO, Smith LA, Klinger KW, et al. Long-lasting arrest of murine polycystic kidney disease with CDK inhibitor roscovitine. Nature. 2006;444:949–952.
  • Bukanov NO, Moreno SE, Natoli TA, et al. CDK inhibitors R-roscovitine and S-CR8 effectively block renal and hepatic cystogenesis in an orthologous model of ADPKD. Cell Cycle Georget Tex. 2012;11:4040–4046.
  • Husson H, Moreno S, Smith LA, et al. Reduction of ciliary length through pharmacologic or genetic inhibition of CDK5 attenuates polycystic kidney disease in a model of nephronophthisis. Hum Mol Genet. 2016;25:2245–2255.
  • Tao Y, Kim J, Schrier RW, et al. Rapamycin markedly slows disease progression in a rat model of polycystic kidney disease. J Am Soc Nephrol JASN. 2005;16:46–51.
  • Wahl PR, Serra AL, Le Hir M, et al. Inhibition of mTOR with sirolimus slows disease progression in Han: sPRDrats with autosomal dominant polycystic kidney disease (ADPKD). Nephrol Dial Transplant. 2006;21:598–604.
  • Shillingford JM, Murcia NS, Larson CH, et al. The mTOR pathway is regulated by polycystin-1, and its inhibition reverses renal cystogenesis in polycystic kidney disease. Proc Natl Acad Sci. 2006;103:5466–5471.
  • Shillingford JM, Piontek KB, Germino GG, et al. Rapamycin ameliorates PKD resulting from conditional inactivation of Pkd1. J Am Soc Nephrol. 2010;21:489–497.
  • Zafar I, Ravichandran K, Belibi FA, et al. Sirolimus attenuates disease progression in an orthologous mouse model of human autosomal dominant polycystic kidney disease. Kidney Int. 2010;78:754–761.
  • Li A, Fan S, Xu Y, 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;21:1619–1635.
  • Ta MHT, Schwensen KG, Foster S, et al. Effects of TORC1 inhibition during the early and established phases of polycystic kidney disease. PLoS One. 2016;11:e0164193.
  • Gattone VH, Sinders RM, Hornberger TA, et al. Late progression of renal pathology and cyst enlargement is reduced by rapamycin in a mouse model of nephronophthisis. Kidney Int. 2009;76:178–182.
  • Tobin JL, Beales PL. Restoration of renal function in zebrafish models of ciliopathies. Pediatr Nephrol. 2008;23:2095–2099.
  • Wu M, Wahl PR, Le Hir M, et al. Everolimus retards cyst growth and preserves kidney function in a rodent model for polycystic kidney disease. Kidney Blood Press Res. 2007;30:253–259.
  • Yang B, Sonawane ND, Zhao D, et al. Small-molecule CFTR inhibitors slow cyst growth in polycystic kidney disease. J Am Soc Nephrol. 2008;19:1300–1310.
  • Takiar V, Nishio S, Seo-Mayer P, et al. Activating AMP-activated protein kinase (AMPK) slows renal cystogenesis. Proc Natl Acad Sci U S A. 2011;108:2462–2467.
  • Muto S, Aiba A, Saito Y, et al. Pioglitazone improves the phenotype and molecular defects of a targeted Pkd1 mutant. Hum Mol Genet. 2002;11:1731–1742.
  • Blazer-Yost BL, Haydon J, Eggleston-Gulyas T, et al. Pioglitazone attenuates cystic burden in the PCK rodent model of polycystic kidney disease. PPAR Res. 2010;2010:274376.
  • Yoshihara D, Kurahashi H, Morita M, et al. PPAR-gamma agonist ameliorates kidney and liver disease in an orthologous rat model of human autosomal recessive polycystic kidney disease. Am J Physiol Renal Physiol. 2011;300:F465–F474.
  • Flaig SM, Gattone VH, Blazer-Yost BL. Inhibition of cyst growth in PCK and Wpk rat models of polycystic kidney disease with low doses of peroxisome proliferator-activated receptor γ agonists. J Transl Intern Med. 2016;4:118–126.
  • Raphael KL, Strait KA, Stricklett PK, et al. Effect of pioglitazone on survival and renal function in a mouse model of polycystic kidney disease. Am J Nephrol. 2009;30:468–473.
  • Dai B, Liu Y, Mei C, et al. Rosiglitazone attenuates development of polycystic kidney disease and prolongs survival in Han: sPRDrats. Clin Sci Lond Engl. 1979;2010(119):323–333.
  • Cao Y, Semanchik N, Lee SH, et al. Chemical modifier screen identifies HDAC inhibitors as suppressors of PKD models. Proc Natl Acad Sci. 2009;106:21819–21824.
  • Xia S, Li X, Johnson T, et al. Polycystin-dependent fluid flow sensing targets histone deacetylase 5 to prevent the development of renal cysts. Development. 2010;137:1075–1084.
  • Fan LX, Li X, Magenheimer B, et al. Inhibition of histone deacetylases targets the transcription regulator Id2 to attenuate cystic epithelial cell proliferation. Kidney Int. 2012;81:76–85.
  • Cebotaru L, Liu Q, Yanda MK, et al. Inhibition of histone deacetylase 6 activity reduces cyst growth in polycystic kidney disease. Kidney Int. 2016;90:90–99.
  • Zhou X, Fan LX, Sweeney WE, et al. Sirtuin 1 inhibition delays cyst formation in autosomal-dominant polycystic kidney disease. J Clin Invest. 2013;123:3084–3098.
  • Klawitter J, Zafar I, Klawitter J, et al. Effects of lovastatin treatment on the metabolic distributions in the Han:sPRDrat model of polycystic kidney disease. BMC Nephrol. 2013;14:165.
  • Yamaguchi T, Nagao S, Kasahara M, et al. Renal accumulation and excretion of cyclic adenosine monophosphate in a murine model of slowly progressive polycystic kidney disease. Am J Kidney Dis. 1997;30:703–709.
  • Smith LA, Bukanov NO, Husson H, et al. Development of polycystic kidney disease in juvenile cystic kidney mice: insights into pathogenesis, ciliary abnormalities, and common features with human disease. J Am Soc Nephrol. 2006;17:2821–2831.
  • Starremans PG, Li X, Finnerty PE, et al. A mouse model for polycystic kidney disease through a somatic in-frame deletion in the 5′ end of Pkd1. Kidney Int. 2008;73:1394–1405.
  • Hopp K, Ward CJ, Hommerding CJ, et al. Functional polycystin-1 dosage governs autosomal dominant polycystic kidney disease severity. J Clin Invest. 2012;122:4257–4273.
  • Wallace DP. Cyclic AMP-mediated cyst expansion. Biochim Biophys Acta. 2011;1812:1291–1300.
  • Torres VE, Harris PC. Strategies targeting cAMP signaling in the treatment of polycystic kidney disease. J Am Soc Nephrol. 2014;25:18–32.
  • Torres VE, Chapman AB, Devuyst O, et al. Tolvaptan in patients with autosomal dominant polycystic kidney disease. N Engl J Med. 2012;367:2407–2418.
  • Gansevoort RT, Arici M, Benzing T, et al. Recommendations for the use of tolvaptan in autosomal dominant polycystic kidney disease: a position statement on behalf of the ERA-EDTA Working Groups on Inherited Kidney Disorders and European Renal Best Practice. Nephrol Dial Transplant. 2016;31:337–348.
  • Winkler SN, Torikai S, Levine BS, et al. Effect of somatostatin on vasopressin-induced antidiuresis and renal cyclic AMP of rats. Miner Electrolyte Metab. 1982;7:8–14.
  • Friedlander G, Amiel C. Somatostatin and alpha 2-adrenergic agonists selectively inhibit vasopressin-induced cyclic AMP accumulation in MDCK cells. FEBS Lett. 1986;198:38–42.
  • Ruggenenti P, Remuzzi A, Ondei P, et al. Safety and efficacy of long-acting somatostatin treatment in autosomal-dominant polycystic kidney disease. Kidney Int. 2005;68:206–216.
  • Ghosh AK, Hurd T, Hildebrandt F. 3D spheroid defects in NPHP knockdown cells are rescued by the somatostatin receptor agonist octreotide. Am J Physiol Ren Physiol. 2012;303:F1225–F1229.
  • Hogan MC, Masyuk TV, Page LJ, et al. Randomized clinical trial of long-acting somatostatin for autosomal dominant polycystic kidney and liver disease. J Am Soc Nephrol JASN. 2010;21:1052–1061.
  • van Keimpema L, Nevens F, Vanslembrouck R, et al. Lanreotide reduces the volume of polycystic liver: a randomized, double-blind, placebo-controlled trial. Gastroenterology. 2009;137:1661-1668-2.
  • Caroli A, Perico N, Perna A, et al. Effect of longacting somatostatin analogue on kidney and cyst growth in autosomal dominant polycystic kidney disease (ALADIN): a randomised, placebo-controlled, multicentre trial. Lancet. 2013;382:1485–1495.
  • Myint TM, Rangan GK, Webster AC. Treatments to slow progression of autosomal dominant polycystic kidney disease: systematic review and meta-analysis of randomized trials: ADPKD meta-analysis. Nephrology. 2014;19:217–226.
  • Du J, Wilson PD. Abnormal polarization of EGF receptors and autocrine stimulation of cyst epithelial growth in human ADPKD. Am J Physiol. 1995;269:C487–495.
  • Orellana SA, Sweeney WE, Neff CD, et al. Epidermal growth factor receptor expression is abnormal in murine polycystic kidney. Kidney Int. 1995;47:490–499.
  • Nakanishi K, Sweeney W, Avner ED. Segment-specific c-ErbB2 expression in human autosomal recessive polycystic kidney disease. J Am Soc Nephrol JASN. 2001;12:379–384.
  • Pugh JL, Sweeney WE, Avner ED. Tyrosine kinase activity of the EGF receptor in murine metanephric organ culture. Kidney Int. 1995;47:774–781.
  • Sweeney WE, Avner ED. Functional activity of epidermal growth factor receptors in autosomal recessive polycystic kidney disease. Am J Physiol Ren Physiol. 1998;275:F387–F394.
  • Ye M, Grant M, Sharma M, et al. Cyst fluid from human autosomal dominant polycystic kidneys promotes cyst formation and expansion by renal epithelial cells in vitro. J Am Soc Nephrol JASN. 1992;3:984–994.
  • Rohatgi R, Zavilowitz B, Vergara M, et al. Cyst fluid composition in human autosomal recessive polycystic kidney disease. Pediatr Nephrol Berl Ger. 2005;20:552–553.
  • Zheleznova NN, Wilson PD, Staruschenko A. Epidermal growth factor-mediated proliferation and sodium transport in normal and PKD epithelial cells. Biochim Biophys Acta. 2011;1812:1301–1313.
  • Torres VE, Sweeney WE, Wang X, et al. Epidermal growth factor receptor tyrosine kinase inhibition is not protective in PCK rats. Kidney Int. 2004;66:1766–1773.
  • Yamaguchi T, Pelling JC, Ramaswamy NT, et al. cAMP stimulates the in vitro proliferation of renal cyst epithelial cells by activating the extracellular signal-regulated kinase pathway1. Kidney Int. 2000;57:1460–1471.
  • Yamaguchi T, Nagao S, Wallace DP, et al. Cyclic AMP activates B-Raf and ERK in cyst epithelial cells from autosomal-dominant polycystic kidneys. Kidney Int. 2003;63:1983–1994.
  • Yamaguchi T, Wallace DP, Magenheimer BS, et al. Calcium restriction allows cAMP activation of the B-Raf/ERK pathway, switching cells to a cAMP-dependent growth-stimulated phenotype. J Biol Chem. 2004;279:40419–40430.
  • Yamaguchi T, Reif GA, Calvet JP, et al. Sorafenib inhibits cAMP-dependent ERK activation, cell proliferation, and in vitro cyst growth of human ADPKD cyst epithelial cells. Am J Physiol Renal Physiol. 2010;299:F944–F951.
  • Rastogi A, Rosner MH, Barash I, et al. Tyrosine kinase inhibitor tesevatinib for patients with autosomal dominant polycystic kidney disease. J Am Soc  Neph. 2015;26:830A.
  • Choi HJC, Lin J-R, Vannier J-B, et al. NEK8 links the ATR-regulated replication stress response and S phase CDK activity to renal ciliopathies. Mol Cell. 2013;51:423–439.
  • Slaats GG, Saldivar JC, Bacal J, et al. DNA replication stress underlies renal phenotypes in CEP290-associated Joubert syndrome. J Clin Invest. 2015;125:3657–3666.
  • Senderowicz AM. Flavopiridol: the first cyclin-dependent kinase inhibitor in human clinical trials. Invest New Drugs. 1999;17:313–320.
  • Schang LM, Rosenberg A, Schaffer PA. Roscovitine, a specific inhibitor of cellular cyclin-dependent kinases, inhibits herpes simplex virus DNA synthesis in the presence of viral early proteins. J Virol. 2000;74:2107–2120.
  • Schang LM, Bantly A, Knockaert M, et al. Pharmacological cyclin-dependent kinase inhibitors inhibit replication of wild-type and drug-resistant strains of herpes simplex virus and human immunodeficiency virus type 1 by targeting cellular, not viral, proteins. J Virol. 2002;76:7874–7882.
  • Xu H, Yu S, Liu Q, et al. Recent advances of highly selective CDK4/6 inhibitors in breast cancer. J Hematol Oncol. 2017;10:97.
  • Asghar U, Witkiewicz AK, Turner NC, et al. The history and future of targeting cyclin-dependent kinases in cancer therapy. Nat Rev Drug Discov. 2015;14:130–146.
  • Wilson PD. Polycystic kidney disease. N Engl J Med. 2004;350:151–164.
  • Siroky BJ, Towbin AJ, Trout AT, et al. Improvement in renal cystic disease of tuberous sclerosis complex after treatment with mammalian target of rapamycin inhibitor. J Pediatr. 2017;187:318–322.e2.
  • Gao X, Zhang Y, Arrazola P, et al. Tsc tumour suppressor proteins antagonize amino-acid-TOR signalling. Nat Cell Biol. 2002;4:699–704.
  • Inoki K, Li Y, Zhu T, et al. TSC2 is phosphorylated and inhibited by Akt and suppresses mTOR signalling. Nat Cell Biol. 2002;4:648–657.
  • Tee AR, Fingar DC, Manning BD, et al. Tuberous sclerosis complex-1 and −2 gene products function together to inhibit mammalian target of rapamycin (mTOR)-mediated downstream signaling. Proc Natl Acad Sci U S A. 2002;99:13571–13576.
  • Hartman TR, Liu D, Zilfou JT, et al. The tuberous sclerosis proteins regulate formation of the primary cilium via a rapamycin-insensitive and polycystin 1-independent pathway. Hum Mol Genet. 2009;18:151–163.
  • Boletta A. Emerging evidence of a link between the polycystins and the mTOR pathways. PathoGenetics. 2009;2:6.
  • Pema M, Drusian L, Chiaravalli M, et al. mTORC1-mediated inhibition of polycystin-1 expression drives renal cyst formation in tuberous sclerosis complex. Nat Commun 2016;7:ncomms10786.
  • Distefano G, Boca M, Rowe I, et al. Polycystin-1 regulates extracellular signal-regulated kinase-dependent phosphorylation of tuberin to control cell size through mTOR and its downstream effectors S6K and 4EBP1. Mol Cell Biol. 2009;29:2359–2371.
  • Zafar I, Belibi FA, He Z, et al. Long-term rapamycin therapy in the Han:SPRD rat model of polycystic kidney disease (PKD). Nephrol Dial Transplant. 2009;24:2349–2353.
  • Walz G, Budde K, Mannaa M, et al. Everolimus in patients with autosomal dominant polycystic kidney disease. N Engl J Med. 2010;363:830–840.
  • Grantham JJ, Bennett WM, Perrone RD. mTOR inhibitors and autosomal dominant polycystic kidney disease. N Engl J Med. 2011;364:286–289.
  • Serra AL, Poster D, Kistler AD, et al. Sirolimus and kidney growth in autosomal dominant polycystic kidney disease. N Engl J Med. 2010;363:820–829.
  • Jardine MJ, Liyanage T, Buxton E, et al. mTOR inhibition in autosomal-dominant polycystic kidney disease (ADPKD): the question remains open. Nephrol Dial Trans Off Publ Eur Dial Transpl Assoc Eur Ren Assoc. 2013;28:242–244.
  • Perico N, Antiga L, Caroli A, et al. Sirolimus therapy to halt the progression of ADPKD. J Am Soc Nephrol JASN. 2010;21:1031–1040.
  • Belibi F, Ravichandran K, Zafar I, et al. mTORC1/2 and rapamycin in female Han: sPRDrats with polycystic kidney disease. Am J Physiol Renal Physiol. 2011;300:F236–F244.
  • Ravichandran K, Zafar I, He Z, et al. An mTOR anti-sense oligonucleotide decreases polycystic kidney disease in mice with a targeted mutation in Pkd2. Hum Mol Genet. 2014;23:4919–4931.
  • O’Sullivan DA, Torres VE, Gabow PA, et al. Cystic fibrosis and the phenotypic expression of autosomal dominant polycystic kidney disease. Am J Kidney Dis Off J Natl Kidney Found. 1998;32:976–983.
  • Xu N, Glockner JF, Rossetti S, et al. Autosomal dominant polycystic kidney disease coexisting with cystic fibrosis. J Nephrol. 2006;19:529–534.
  • Tradtrantip L, Sonawane ND, Namkung W, et al. Nanomolar potency pyrimido-pyrrolo-quinoxalinedione CFTR inhibitor reduces cyst size in a polycystic kidney disease model. J Med Chem. 2009;52:6447–6455.
  • Nofziger C, Brown KK, Smith CD, et al. PPARγ agonists inhibit vasopressin-mediated anion transport in the MDCK-C7 cell line. Am J Physiol Ren Physiol. 2009;297:F55–F62.
  • Home P. Safety of PPAR agonists. Diabetes Care. 2011;34:S215–S219.
  • van Dijk MA, Kamper AM, van Veen S, et al. Effect of simvastatin on renal function in autosomal dominant polycystic kidney disease. Nephrol Dial Trans Off Publ Eur Dial Transpl Assoc Eur Ren Assoc. 2001;16:2152–2157.
  • Cadnapaphornchai MA, George DM, McFann K, et al. Effect of pravastatin on total kidney volume, left ventricular mass index, and microalbuminuria in pediatric autosomal dominant polycystic kidney disease. Clin J Am Soc Nephrol CJASN. 2014;9:889–896.
  • Ruiz-Perez VL, Blair HJ, Rodriguez-Andres ME, et al. Evc is a positive mediator of Ihh-regulated bone growth that localises at the base of chondrocyte cilia. Dev Camb Engl. 2007;134:2903–2912.
  • Aguilar A, Meunier A, Strehl L, et al. Analysis of human samples reveals impaired SHH-dependent cerebellar development in Joubert syndrome/Meckel syndrome. Proc Natl Acad Sci U S A. 2012;109:16951–16956.
  • Attanasio M, Uhlenhaut NH, Sousa VH, et al. Loss of GLIS2 causes nephronophthisis in humans and mice by increased apoptosis and fibrosis. Nat Genet. 2007;39:1018–1024.
  • Hynes AM, Giles RH, Srivastava S, et al. Murine Joubert syndrome reveals Hedgehog signaling defects as a potential therapeutic target for nephronophthisis. Proc Natl Acad Sci. 2014;111:9893–9898.
  • Mak IW, Evaniew N, Ghert M. Lost in translation: animal models and clinical trials in cancer treatment. Am J Transl Res. 2014;6:114–118.
  • Ehret T, Torelli F, Klotz C, et al. Translational rodent models for research on parasitic protozoa - a review of confounders and possibilities. Front Cell Infect Microbiol. 2017;7:238.
  • Ortiz A, Sanchez-Niño MD, Izquierdo MC, et al. Translational value of animal models of kidney failure. Eur J Pharmacol. 2015;759:205–220.
  • Harris PC, Torres VE. Polycystic kidney disease, autosomal dominant. In: Pagon RA, Adam MP, Ardinger HH, et al. editors. GeneReviews(®) [Internet]. Seattle (WA): University of Washington, Seattle; 1993. cited 2017 Aug 22. Available from: http://www.ncbi.nlm.nih.gov/books/NBK1246/
  • Braun DA, Hildebrandt F. Ciliopathies. Cold Spring Harb Perspect Biol. 2017;9:a028191.
  • Takahashi K, Tanabe K, Ohnuki M, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131:861–872.
  • Zhou T, Benda C, Dunzinger S, et al. Generation of human induced pluripotent stem cells from urine samples. Nat Protoc. 2012;7:2080–2089.
  • Shi L, Cui Y, Luan J, et al. Urine-derived induced pluripotent stem cells as a modeling tool to study rare human diseases. Intractable Rare Dis Res. 2016;5:192–201.
  • El Hokayem J, Cukier HN, Dykxhoorn DM. Blood derived induced pluripotent stem cells (iPSCs): benefits, challenges and the road ahead. J Alzheimers Dis Park. 2016;6:275.
  • Song B, Smink AM, Jones CV, et al. The directed differentiation of human iPS cells into kidney podocytes. PloS One. 2012;7:e46453.
  • Xia Y, Nivet E, Sancho-Martinez I, et al. Directed differentiation of human pluripotent cells to ureteric bud kidney progenitor-like cells. Nat Cell Biol. 2013;15:1507–1515.
  • Morizane R, Monkawa T, Fujii S, et al. Kidney specific protein-positive cells derived from embryonic stem cells reproduce tubular structures in vitro and differentiate into renal tubular cells. PloS One. 2014;8:e64843.
  • Takasato M, Er PX, Becroft M, et al. Directing human embryonic stem cell differentiation towards a renal lineage generates a self-organizing kidney. Nat Cell Biol. 2014;16:118–126.
  • Lam AQ, Freedman BS, Morizane R, et al. Rapid and efficient differentiation of human pluripotent stem cells into intermediate mesoderm that forms tubules expressing kidney proximal tubular markers. J Am Soc Nephrol JASN. 2014;25:1211–1225.
  • Ciampi O, Iacone R, Longaretti L, et al. Generation of functional podocytes from human induced pluripotent stem cells. Stem Cell Res. 2016;17:130–139.
  • Takasato M, Er PX, Chiu HS, et al. Kidney organoids from human iPS cells contain multiple lineages and model human nephrogenesis. Nature. 2015;526:564–568.
  • Taguchi A, Kaku Y, Ohmori T, et al. Redefining the in vivo origin of metanephric nephron progenitors enables generation of complex kidney structures from pluripotent stem cells. Cell Stem Cell. 2014;14:53–67.
  • Morizane R, Lam AQ, Freedman BS, et al. Nephron organoids derived from human pluripotent stem cells model kidney development and injury. Nat Biotechnol. 2015;33:1193–1200.
  • Freedman BS, Brooks CR, Lam AQ, et al. Modelling kidney disease with CRISPR-mutant kidney organoids derived from human pluripotent epiblast spheroids. Nat Commun. 2015;6:8715.
  • Ginn SL, Alexander IE, Edelstein ML, et al. Gene therapy clinical trials worldwide to 2012 - an update. J Gene Med. 2013;15:65–77.
  • Heikkilä P, Tibell A, Morita T, et al. Adenovirus-mediated transfer of type IV collagen alpha5 chain cDNA into swine kidney in vivo: deposition of the protein into the glomerular basement membrane. Gene Ther. 2001;8:882–890.
  • Park J, Murray GJ, Limaye A, et al. Long-term correction of globotriaosylceramide storage in Fabry mice by recombinant adeno-associated virus-mediated gene transfer. Proc Natl Acad Sci U S A. 2003;100:3450–3454.
  • Doudna JA, Charpentier E. Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science. 2014;346:1258096.
  • Yin H, Xue W, Chen S, et al. Genome editing with Cas9 in adult mice corrects a disease mutation and phenotype. Nat Biotechnol. 2014;32:551–553.
  • Long C, Amoasii L, Mireault AA, et al. Postnatal genome editing partially restores dystrophin expression in a mouse model of muscular dystrophy. Science. 2016;351:400–403.
  • Nelson CE, Hakim CH, Ousterout DG, et al. In vivo genome editing improves muscle function in a mouse model of Duchenne muscular dystrophy. Science. 2016;351:403–407.
  • Tabebordbar M, Zhu K, Cheng JKW, et al. In vivo gene editing in dystrophic mouse muscle and muscle stem cells. Science. 2016;351:407–411.
  • Xu L, Park KH, Zhao L, et al. CRISPR-mediated genome editing restores dystrophin expression and function in mdx mice. Mol Ther J Am Soc Gene Ther. 2016;24:564–569.
  • Yang Y, Wang L, Bell P, et al. A dual AAV system enables the Cas9-mediated correction of a metabolic liver disease in newborn mice. Nat Biotechnol. 2016;34:334–338.
  • Yin H, Song C-Q, Dorkin JR, et al. Therapeutic genome editing by combined viral and non-viral delivery of CRISPR system components in vivo. Nat Biotechnol. 2016;34:328–333.
  • Ohmori T, Nagao Y, Mizukami H, et al. CRISPR/Cas9-mediated genome editing via postnatal administration of AAV vector cures haemophilia B mice. Sci Rep. 2017;7:4159.
  • Yin H, Kauffman KJ, Anderson DG. Delivery technologies for genome editing. Nat Rev Drug Discov. 2017;16:387–399.
  • Cirak S, Arechavala-Gomeza V, Guglieri M, et al. Exon skipping and dystrophin restoration in patients with Duchenne muscular dystrophy after systemic phosphorodiamidate morpholino oligomer treatment: an open-label, phase 2, dose-escalation study. Lancet. 2011;378:595–605.
  • Gerard X, Perrault I, Hanein S, et al. AON-mediated exon skipping restores ciliation in fibroblasts harboring the common Leber congenital amaurosis CEP290 mutation. Mol Ther Nucleic Acids. 2012;1:e29.
  • Sayer JA, Otto EA, O’Toole JF, et al. The centrosomal protein nephrocystin-6 is mutated in Joubert syndrome and activates transcription factor ATF4. Nat Genet. 2006;38:674–681.
  • Valente EM, Silhavy JL, Brancati F, et al. Mutations in CEP290, which encodes a centrosomal protein, cause pleiotropic forms of Joubert syndrome. Nat Genet. 2006;38:623–625.
  • Coppieters F, Lefever S, Leroy BP, et al. CEP290, a gene with many faces: mutation overview and presentation of CEP290base. Hum Mutat. 2010;31:1097–1108.
  • Molinari E, Srivastava S, Sayer JA, et al. From disease modelling to personalised therapy in patients with CEP290 mutations. F1000Research. 2017;6:669.
  • Peces R, Peces C, Coto E, et al. Bilineal inheritance of type 1 autosomal dominant polycystic kidney disease (ADPKD) and recurrent fetal loss. NDT Plus. 2008;1:289–291.
  • Pei Y, Paterson AD, Wang KR, et al. Bilineal disease and trans-heterozygotes in autosomal dominant polycystic kidney disease. Am J Hum Genet. 2001;68:355–363.
  • Wu X, Gao H, Pasupathy S, et al. Systemic administration of naked DNA with targeting specificity to mammalian kidneys. Gene Ther. 2005;12:477–486.
  • Rocca CJ, Ur SN, Harrison F, et al. rAAV9 combined with renal vein injection is optimal for kidney-targeted gene delivery: conclusion of a comparative study. Gene Ther. 2014;21:618–628.
  • Oroojalian F, Rezayan AH, Mehrnejad F, et al. Efficient megalin targeted delivery to renal proximal tubular cells mediated by modified-polymyxin B-polyethylenimine based nano-gene-carriers. Mater Sci Eng C. 2017;79:770–782.
  • Wolf MTF. Nephronophthisis and related syndromes. Curr Opin Pediatr. 2015;27:201–211.
  • Lohmann K, Klein C. Next generation sequencing and the future of genetic diagnosis. Neurother J Am Soc Exp Neurother. 2014;11:699–707.
  • Besse W, Dong K, Choi J, et al. Isolated polycystic liver disease genes define effectors of polycystin-1 function. J Clin Invest. 2017;127:1772–1785.
  • Zarrinpar A, Lee D-K, Silva A, et al. Individualizing liver transplant immunosuppression using a phenotypic personalized medicine platform. Sci Transl Med. 2016;8:333ra49.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.