Disease of the Month

Nephronophthisis-Associated

Friedhelm Hildebrandt and Weibin Zhou Departments of Pediatrics and of Human Genetics, University of Michigan, Ann Arbor, Michigan

Nephronophthisis (NPHP), an autosomal recessive cystic disease, represents the most frequent genetic cause of end-stage in the first three decades of life. Contrary to polycystic kidney disease, NPHP shows normal or diminished kidney size, are concentrated at the corticomedullary junction, and tubulointerstitial fibrosis is dominant. NPHP can be associated with (Senior-Løken syndrome), fibrosis, and cerebellar vermis aplasia () in approximately 10% of patients. Positional cloning of six novel (NPHP1 through 6) as mutated in NPHP and functional characterization of their encoded have contributed to the concept of “ciliopathies.” It has helped advance a new unifying theory of cystic kidney diseases. This theory states that the products of all genes that are mutated in cystic kidney diseases in humans, mice, or zebrafish are expressed in primary cilia or of renal epithelial cells. Primary cilia are sensory organelles that connect mechanosensory, visual, osmotic, and other stimuli to mechanisms of cell-cycle control and epithelial cell polarity. The ciliary theory explains the multiple organ involvement in NPHP regarding retinitis pigmentosa, liver fibrosis, ataxia, situs inversus, and mental retardation. in NPHP genes cause defects in signaling mechanisms, including the noncanonical Wnt signaling pathway. The “” NPHP thereby is caused by defects in tissue differentiation and maintenance as a result of impaired processing of extracellular cues. Nephrocystins, the proteins that are encoded by NPHP genes, are highly conserved in evolution. Positional cloning of additional causative genes of NPHP will elucidate further signaling mechanisms that are involved, thereby establishing therapeutic approaches using animal models in mouse, zebrafish, and Caenorhabditis elegans. J Am Soc Nephrol 18: 1855–1871, 2007. doi: 10.1681/ASN.2006121344

Nephronophthisis: A Frequent Genetic which there is moderate renal enlargement), cysts seem to develop Cause of Kidney Failure in Children e vacuo by loss of normal tissue. This is in contrast to polycystic Renal Involvement kidney disease (PKD), in which cysts are evenly spread out over Nephronophthisis (NPHP) is an autosomal recessive cystic kid- the entire organ and lead to gross enlargement of the kidneys (4). ney disease that constitutes the most frequent genetic cause for NPHP is inherited in an autosomal recessive mode. This end-stage kidney disease (ESKD) in the first three decades of life includes NPHP variants with extrarenal manifestations (4). (1–4). Three clinical forms of NPHP have been distinguished by NPHP has previously been grouped together with the clinical onset of ESKD: Infantile (5), juvenile (6), and adolescent NPHP (7), entity of medullary (MCKD) (6,10) be- which manifest with ESKD at the median ages of 1, 13, and 19 yr, cause of similarities of clinical and pathologic features (12). respectively. Initial symptoms are relatively mild (except in infan- Both NPHP and MCKD feature corticomedullary cysts in kid- tile NPHP type 2) and consist of , with regular neys of normal or slightly reduced size. However, MCKD is fluid intake at nighttime, secondary enuresis, and (8). At clearly distinct from NPHP regarding multiple aspects: (1) an average age of 9 yr, a slightly raised serum creatinine is noted, MCKD follows autosomal dominant inheritance, (2) ESKD oc- before ESKD invariably develops within a few years. Renal ultra- curs in the fourth decade and later, and (3) in MCKD there is no sound reveals increased echogenicity. Later, cysts appear at the extrarenal involvement other than hyperuricemia and gout. corticomedullary junction within kidneys of normal or slightly NPHP was first described by Smith and Graham in 1945 (2) reduced size (Figure 1, A and B) (9). Renal histology reveals a and by Fanconi et al. (3), who introduced the term “familial characteristic triad of tubular basement membrane disruption, juvenile nephronophthisis.” Since then, Ͼ300 cases have been tubulointerstitial nephropathy, and corticomedullary cysts (Figure published in the literature (10). In NPHP, the earliest presenting 1C) (10,11). In NPHP, cysts arise from the corticomedullary junc- symptoms are polyuria, polydipsia, decreased urinary concen- tion of the kidneys (Figure 1, A and B). Because kidneys size is trating ability, and secondary enuresis. They occur in Ͼ80% of normal or slightly reduced (except in infantile NPHP type 2, in cases (13) and start at approximately 6 yr of age. Anemia and growth retardation develop later in the course of the disease (8). Regular fluid intake at nighttime is a characteristic feature of Published online ahead of print. Publication date available at www.jasn.org. the patient’s history and starts at approximately age 6 yr. Address correspondence to: Dr. Friedhelm Hildebrandt, Departments of Pediat- Because of the mild nature of symptoms and the lack of edema, rics and of Human Genetics, University of Michigan Health System, 8220C MSRB , or urinary tract infections, there is often a delay III, 1150 West Medical Center Drive, Ann Arbor, MI 48109-0646. Phone: 734-615- 7285 (office), 734-615-7895 or 7896 (laboratories); Fax: 734-615-1386 or 7770; in the diagnosis of NPHP. This causes a risk for sudden death E-mail: [email protected] from fluid and electrolyte imbalance. Disease recurrence has

Copyright © 2007 by the American Society of Nephrology ISSN: 1046-6673/1806-1855 1856 Journal of the American Society of Nephrology J Am Soc Nephrol 18: 1855–1871, 2007

which follows autosomal dominant inheritance, ESKD occurs later in life. MCKD types 1 and 2 show a median onset of ESKD at 62 (25) and 32 yr (26), respectively. MCKD type 2 can be positively diagnosed by detection of mutations in the UMOD that encodes uromodulin/Tamm-Horsfall .

Epidemiology NPHP has been reported from virtually all regions of the world (13). The incidence of the disease has been given as nine patients per 8.3 million (27) in the United States or one in 50,000 live births in Canada (10,28). Although a rare disorder, it represents the most frequent genetic cause of ESKD in the first three decades of life and is a major cause of ESKD in children, accounting for 10 to 25% of these patients in Europe (13,29–31). In the North American pedi- atric ESKD population, pooled data indicate a prevalence of ap- proximately 5% of all children with ESKD (32,33).

Extrarenal Manifestations of Eye, Brain, and Figure 1. Morphology of nephronophthisis (NPHP). (A) Macro- Liver scopic pathology reveals cysts that arise from the corticomed- NPHP may be associated with tapetoretinal degeneration (Se- ullary junction of normal-sized kidneys. (B) Renal ultrasound nior-Løken syndrome [SLSN] [34,35]), cerebellar vermis aplasia demonstrates increased echogenicity, loss of corticomedullary (Joubert syndrome [JBTS] [36,37]), ocular motor type differentiation, and the presence of cysts. In contrast to poly- Cogan (38), mental retardation (21,39), liver fibrosis (40), or cone- cystic kidney disease (PKD), in NPHP, cysts are concentrated at shaped epiphyses of the phalanges (Mainzer-Saldino syndrome the corticomedullary border and kidneys are not enlarged. (C) [41]). Infantile NPHP type 2 (17) can be associated with situs Renal histology in NPHP shows the characteristic triad of renal inversus (17), retinitis pigmentosa (42), or cardiac ventricular septal tubular (and glomerular) cysts, tubular membrane disruption, and tubulointerstitial cell infiltrates with interstitial fibrosis and defect (17). The ciliary theory of NPHP elucidates the pathogenic periglomerular fibrosis. Adapted from Hildebrandt et al. (9a); basis of extrarenal organ involvement regarding retinitis pigmentosa, and courtesy of D. Bockenhauer. liver fibrosis, ataxia, situs inversus, and mental retardation.

Retinal Involvement (SLSN) never been reported in kidneys that were transplanted to pa- SLSN, represented by the concomitant occurrence of NPHP tients with NPHP (14). By positional cloning, we and others with retinitis pigmentosa, was first described by Contreras (43), have identified recessive mutations in six different novel genes Senior (34), and Løken (35). Three different terms have been used as causing NPHP: NPHP1 (15,16), NPHP2/inversin (17), NPHP3 in the literature to describe the retinal findings of SLSN: Retinitis (18), NPHP4 (19), NPHP5 (20), and NPHP6 (21), defining NPHP pigmentosa, tapetoretinal degeneration, and retinal-renal dyspla- types 1, 2, 3, 4, 5, and 6, respectively. This has made definite sia. This most likely reflects a spectrum within the pathogenesis molecular genetic diagnostics possible (www.renalgenes.org). that includes developmental defects (dysplasia) as well as defects Homozygous deletions in the NPHP1 gene account for approx- of tissue maintenance (degeneration) (44). In children with reces- imately 25% of all NPHP cases, whereas the other genes con- sive mutations in the NPHP1, 2, 3, and 4 genes, retinitis pigmen- tribute Ͻ2% each. As expected in a recessive disease, pene- tosa occurs in approximately 10% of all affected families, without trance of the renal phenotype seems to be 100%. any obvious genotype/phenotype correlation. Whether patients In NPHP, chronic renal failure develops within the first three with retinal involvement carry an additional in an un- decades of life (7,22,23). Infantile NPHP, which is characterized known modifier gene is an open question. Early-onset and late- by mutations in NPHP2/inversin, leads to ESKD between birth onset types of SLSN have been distinguished. The early-onset type and age 3 yr (17,23). In a study conducted in 46 children who seems to represent a form of Leber’s congenital amaurosis, be- had juvenile NPHP type 1 caused by mutations in the NPHP1 cause children exhibit coarse nystagmus and/or blindness at birth gene, a serum creatinine of 6 mg/dl was reached at a median or develop these symptoms within the first 2 yr of life (45). It is age of 13 yr (range 4 to 20 yr) (6,22). Similarly, the median age interesting that patients with mutations in the NPHP5 or NPHP6 of ESKD in patients with mutations in the NPHP5 gene was 13 genes exhibit early-onset retinitis pigmentosa in all known cases yr (20). The median time lapse between a serum creatinine of 2 (20,21). Fundoscopic alterations are present in all patients with and 4 mg/dl was 32 mo and between 4 and 6 mg/dl was 10 mo late-onset SLSN by the age of 10 yr. The late-onset form manifests (24). In patients with adolescent NPHP as a result of mutations first with night blindness, followed by development of blindness in the NPHP3 gene, ESKD develops by 19 yr of age (18). If renal during school age. Retinal degeneration is characterized by a failure has not developed by the age of 25 yr, then the diagnosis constant and complete extinction of the electroretinogram, which of recessive NPHP should be questioned and autosomal dom- precedes the development of visual and fundoscopic signs of inant MCKD considered as a differential diagnosis. In MCKD, retinitis pigmentosa (46). The kidney involvement in SLSN is J Am Soc Nephrol 18: 1855–1871, 2007 Nephronophthisis-Associated Ciliopathies 1857 identical clinically to what is known from patients with NPHP patient with situs inversus also had a cardiac ventricular septal without ocular involvement regarding age of onset, symptoms, defect as a heterotaxy phenotype. This finding was analogous to and histology of renal disease. the randomization of heart looping that was seen in nphp2/inversin knockdown experiments in zebrafish. Recently, it has become Cerebellar Vermis Aplasia (JBTS) apparent that products of other genes that are associated with In JBTS, a developmental disorder with multiple organ involve- renal cystic disease (in addition to inversin) are important for ment, NPHP or cystic dysplasia occurs in association with left–right axis determination of the body plan (66,67). The gene coloboma of the eye (or retinal degeneration); with aplasia/hyp- PKD2, mutations in which cause autosomal dominant PKD and oplasia of the cerebellar vermis causing ataxia; and with the fac- that encodes the calcium release channel polycystin-2, had been Ϫ Ϫ ultative symptoms of psychomotor retardation, polydactyly, oc- shown in a Pkd2 / mouse model to represent a gene that regu- cipital encephalocele, and episodic neonatal tachypnea/dyspnea lates left–right axis determination, acting upstream of Nodal, Ebaf, (36,37,47–49). A pathognomonic diagnostic feature of JBTS on Leftb, and Pitx2 (68,69). axial magnetic resonance imaging of the brain is the presence of prominent superior cerebellar peduncles, termed the “molar tooth Other Syndromes with NPHP sign” of the midbrain-hindbrain junction (49,50). In patients with Bardet-Biedl syndrome (BBS) exhibits renal histology that is the association of JBTS and NPHP, mutations have been described similar to NPHP (70,71). Positional cloning of recessive genes in three different genes, NPHP1 (37,49,50), AHI (51,52) (JBTS type that are mutated in BBS has revealed that the molecular relation 3), and NPHP6 (21,53). Patients with JBTS have abnormal axonal between NPHP and BBS may lie in coexpression of the respec- decussation (crossing in the brain) that affects the corticospinal tive gene products in primary cilia, basal bodies, and centro- tract and superior cerebellar peduncles, thereby explaining the somes of renal epithelial cells (72). For BBS, an oligogenic motor and behavioral abnormalities (54). Some patients also have inheritance pattern has been described. This refers to the find- abnormal cerebral structure with cortical polymicrogyria (55). Oc- ing that mutations in more than one BBS gene may be required ular motor apraxia type Cogan, defined as the transient inability of for full penetrance of some aspects of organ involvement (73). horizontal eye movements in the first few years of life, may be Further disease variants have been described in association with associated with JBTS. This symptom has been described in pa- NPHP, including Jeune syndrome (asphyxiating thoracic dyspla- tients with mutations in the NPHP1 (38,56) (“JBTS4”) and NPHP4 sia) (74–77), Ellis van Creveld syndrome (78), RHYNS syndrome (57) genes. It may be due to defects in the nuclei of the abducens (retinitis pigmentosa, hypopituitarism, NPHP, and skeletal dys- nerve, which contain both ipsilaterally projecting motor neurons plasia) (79), Alstrom syndrome (retinitis pigmentosa, deafness, and contralaterally projecting interneurons, or supranuclear con- obesity, and diabetes without mental defect, polydactyly, or hy- trol regions such as the pontine paramedian reticular formation pogonadism) (80), and Meckel-Gruber syndrome (81,82), which in that projects to the abducens and oculomotor nuclei, which has the case of MKS3 mutations can be allelic with JBTS (83). In been postulated for other forms of horizontal gaze palsy (58). Alstrom syndrome, the single underlying gene, ALMS1, encodes a Defects in axon guidance may be related to defects in renal tubule novel protein that contains coiled-coil domains and a putative development by shared signaling pathways (see Signal Mecha- nuclear localization signal, as well as serine-rich and histidine-rich nisms Relevant for NPHP). Two additional loci for JBTS have been regions (84,85). ALMS1 forms a part of the (86,87). identified: JBTS1 on 9q34.3 (59) and JBTS2/CORS2 on This, together with the finding that BBS proteins localize to cen- chromosome 11p12-q13.3 (60). trosomes, confirms the role of centrosomal proteins in cystic kid- ney diseases that are associated with diabetes, obesity, and retini- Liver Fibrosis and Skeletal Changes tis pigmentosa (88,89). Additional NPHP-associated disorders are NPHP may be associated with liver fibrosis (40,61–63). Pa- Sensenbrenner syndrome (cranioectodermal dysplasia) (90,91) tients develop hepatomegaly and moderate portal fibrosis with and Arima syndrome (cerebro-oculo-hepato-renal syndrome) (92– mild bile duct proliferation. This pattern differs from that of 94). NPHP has also been described in association with ulcerative classical congenital hepatic fibrosis, whereby biliary dysgenesis colitis (95). is prominent. A recessive mutation in the NPHP3 gene was recently described in a patient with NPHP and liver fibrosis (18). Hepatic involvement in NPHP type 2 (infantile NPHP) Positional Cloning Reveals Seven Causative seems to involve only transient elevation of transaminases (23). Genes for NPHP The association of NPHP with cone-shaped epiphyses of the Since its first description in 1945 (2,3), the pathogenesis of phalanges (type 28 and 28A), known as Mainzer-Saldino syn- NPHP had been elusive. Positional cloning revealed novel drome, was first published by Mainzer et al. (41) in patients genes that cause NPHP when mutated. These are monogenic who also had retinal degeneration and cerebellar ataxia. recessive genes, suggesting that mutations in each single one of these genes is sufficient to cause NPHP in a patient who bears Situs Inversus these mutations, indicating that their products are necessary for The presence of situs inversus was shown in a patient with normal kidney function. Positional cloning thereby generated infantile NPHP and mutations in the NPHP2/inversin gene (17). new insights into disease mechanisms of NPHP and demon- Therefore, the role of inversin in left–right axis specification that strated that they are related to signaling mechanisms of sensory had been described in mice was confirmed in humans (64,65). The cilia, centrosomes, and planar cell polarity (1,72,96). Seven 1858 Journal of the American Society of Nephrology J Am Soc Nephrol 18: 1855–1871, 2007

NPHP-associated genes have been identified so far: NPHP1 fluid movement in the kidney tubule, where polycystin-1 trans- through 6 and AHI1 (15–21,54,55,57) (Table 1). mits the signal to polycystin-2, which is a TRP type calcium ϩ channel. This would produce sufficient Ca2 influx to induce ϩ NPHP1 Ca2 release from intracellular storage, which then regulates We identified mutations in NPHP1 as causing juvenile NPHP numerous intracellular signaling activities that are linked to the type 1 (15,16). NPHP1 encodes nephrocystin-1, a protein that regulation of cell cycle and planar cell polarity (103). In partic- interacts with components of cell–cell and cell–matrix signaling, ular, inversin/NPHP2 function has been implicated in signal- including p130Cas (97), focal adhesion kinase 2 (98), tensin, and ing mechanisms of planar cell polarity (see The Wnt Pathway) filamin A and B (99,100). It also interacts with the products of (104). Okada et al. (105) previously demonstrated that inversin other NPHP genes, such as nephrocystin-2/inversin (17), nephro- is needed to position the cilia in cells of the ventral node. cystin-3 (18), and nephrocystin-4 (57,101). Nephrocystin-1 local- izes to adherens junctions and focal adhesions of renal epithelial NPHP3 cells (99,100), which are involved in cell–cell and cell–basement By positional cloning in a large Venezuelan kindred (7), we membrane communications, respectively (101,102) (Figure 2). identified mutations in NPHP3 as responsible for adolescent NPHP (18). We demonstrated that mutations in the murine NPHP2 ortholog Nphp3 cause the renal cystic mouse mutant pcy (18), The renal cystic changes of infantile NPHP (NPHP type 2) which was recently shown to be responsive to treatment with a combine clinical features of NPHP and of PKD (5). Guided by vasopressin receptor antagonist (106). mapping of a for infantile NPHP to chromosome 9q21- q22 (23) and by the observation that a deletion in the inversin NPHP4 (Invs) gene causes renal cystic phenotype in the inv/inv mouse Mutations in NPHP4 were identified by homozygosity map- model (64,65), we identified mutations in human inversin ping and total genome search for linkage (19,57,107). The en- (INVS) as the cause of infantile NPHP (type 2) with and with- coded protein, nephrocystin-4/nephroretinin, is in a complex out situs inversus (17). Inversin interacts with nephrocystin-1 with other proteins that are involved in cell adhesion and actin and with ␤-, which constitutes the cytoskeleton organization, such as nephrocystin-1, p130Cas, of primary cilia. We demonstrated that nephrocystin-1 and Pyk2, tensin, filamin, and ␣-tubulin. In polarized epithelial inversin localize to primary cilia of renal tubular cells (17)—the cells, nephrocystin-4 localizes to primary cilia, basal bodies, same subcellular compartment that was identified as central to and the cortical actin cytoskeleton, whereas in dividing cells, it the pathogenesis of PKD (1,66,103). This was one of the first localizes to centrosomes (101). Nephrocystin-4 is conserved in findings to support a unifying theory of renal cystogenesis Caenorhabditis elegans and expressed in ciliated head and tail (1,72), which states that proteins that, when mutated, cause neurons of the nematode (108). Upon knockdown, it exhibits a renal cystic disease in humans, mice, or zebrafish (“cystopro- male mating phenotype, similar to orthologs of other genes that teins”) are expressed in primary cilia, basal bodies, or centro- are mutated in cystic kidney disease (108). somes (66,72). The interaction and co-localization to cilia of nephrocystin-1, inversin, and ␤-tubulin provided a functional NPHP5 link between the pathogenesis of NPHP, the pathogenesis of Recently, we identified another novel gene (NPHP5) as being PKD, primary cilia function, and left–right axis determination mutated in NPHP type 5 (20). It is interesting that all mutations (17). The functional relationship between ciliary expression of found were truncations of the encoded protein nephrocystin-5, these so-called “cystoproteins” (proteins mutated in cystic kid- and all patients had early-onset retinitis pigmentosa (SLSN). ney disease) and the renal cystic phenotype, however, is still Nephrocystin-5 contains two IQ domains, which directly inter- somewhat unclear. One of the first concepts for this relation- act with calmodulin (20) and is in a complex with the retinitis ship proposes that cilia may act as mechanosensors to sense pigmentosa GTPase regulator, which when defective causes

Table 1. Genetics and frequency of extrarenal associations in NPHP

NPHP Type Parameter 123456

Mutated gene NPHP1 NPHP2 NPHP3 NPHP4 NPHP5 NPHP6 AHI1 Encoded protein Nephrocystin-1 Nephrocystin-2 Nephrocystin-3 Nephrocystin-4 Nephrocystin-5 Nephrocystin-6 Jouberin Frequency of gene mutation (%)b 20 1 1 2 2 1 Ͻ2 Kidney cysts (%) 100 100 100 100 100 100 ? Retinal degeneration (%) 7 10 Ϸ40 15 100 100 ? Cerebellar vermis aplasia (%) Ϸ1— ———Ϸ90 100 Oculomotor apraxia (Cogan) (%) 1 — — Ͻ1— —?

aNPHP, nephronophthisis. bPercentage of 976 patients from different families with NPHP-associated disorders as evaluated in the authors’ worldwide cohort. Different frequencies have been reported by Saunier et al. (157). J Am Soc Nephrol 18: 1855–1871, 2007 Nephronophthisis-Associated Ciliopathies 1859

Figure 2. Cystoproteins are proteins of genes that are mutated in cystic kidney diseases of humans, mice, or zebrafish. They share the common feature of expression in primary cilia, basal bodies, or centrosomes. Depending on cell-cycle stage, some cystopro- teins localize to adherens junctions or focal adhesions. Many cystoproteins have been localized to more than one intracellular domain. The speckled arrow in the primary indicates the direction of anterograde transport along the microtubule system mediated by 2, a heterotrimeric protein that is composed of two motor units (Kif3a and Kif3b) and one nonmotor unit (KAP3). AJ, adherens junction; BB, ; Cen, ; ER, endoplasmic reticulum; FAP, focal adhesion plaque; TJ, tight junction; PC-1, polycystin-1; PC-2, polycystin-2. Modified from Watnick and Germino (72), with permission.

X-linked retinitis pigmentosa. Both nephrocystin-5 and retinitis function and tissue architecture in the pathogenesis of cystic kid- pigmentosa GTPase regulator localize to connecting cilia of pho- ney disease, retinitis pigmentosa, and central de- toreceptors and in primary cilia of renal epithelial cells (20). The velopment. Mutations in NPHP6/CEP290 have been confirmed as fact that connecting cilia of photoreceptors are the structural causing JBTS with and without renal involvement (21,53). It is equivalents of primary cilia of renal epithelial cells may explain interesting that a 300–amino acid in-frame deletion of NPHP6/ retinal involvement in the retinal-renal syndrome SLSN. CEP290 caused retinal degeneration only, without renal or cere- bellar involvement in the rds16 mouse model (109). This is in NPHP6 accordance with the recent finding that a hypomorphic mutation Very recently, we identified by positional cloning of recessive of NPHP6/CEP290 represents the most frequent cause of Leber’s truncating mutations in a novel gene NPHP6/CEP290, which en- congenital amaurosis (110). codes a centrosomal protein, as the cause of NPHP type 6 and JBTS type 5 (21). We demonstrated that abrogation of NPHP6 AHI1 function in zebrafish causes planar cell polarity (convergent ex- Finally, mutations in AHI1 have been detected in patients tension) defects and recapitulates the human phenotype of NPHP with JBTS with and without renal involvement (51,52,54,55). type 6, including renal cysts, retinitis pigmentosa, and cerebellar Taken together, these findings indicate that the nephrocystin defects (21). In addition, a defect in cell size control and morpho- proteins are involved in functions of sensory cilia, cell polarity, genesis was found upon nphp6 knockdown in the nonvertebrate and cell division (101). Ciona intestinalis. Nephrocystin-6 modulates the activity of ATF4/ CREB2, a transcription factor that is implicated in cAMP-depen- Cilia: A Unifying Theory for Cystic dent renal formation (106). Nephrocystin-6 is expressed in Kidney Disease centrosomes and the mitotic spindle in a cell cycle–dependent The demonstration that nephrocystin-1 and inversin/ manner. Its identification establishes a link between centrosome NPHP-2 localize to primary cilia of renal tubular cells (17) was 1860 Journal of the American Society of Nephrology J Am Soc Nephrol 18: 1855–1871, 2007 among the first findings to support a new unifying theory of renal cystogenesis (72). This theory states that proteins that, when mutated, cause renal cystic disease in humans, mice, or zebrafish (“cystoproteins”) are expressed in primary cilia, basal bodies, or centrosomes (66,72) (Figure 3).

Cilia Structure and Function The cilium is a hair-like structure that extends from the cell surface into the extracellular space. Virtually all vertebrate cell types have cilia in developing or mature tissue. Cilia consist of a microtubule-based axoneme covered by a specialized plasma membrane. The axoneme has nine peripheral microtubule dou- blets arranged around a central core. There may be two central (9 ϩ 2or9ϩ 0 axoneme; Figure 3). 9 ϩ 2 cilia usually have arms that link the microtubule doublets and are motile, whereas most 9 ϩ 0 cilia lack dynein arms and are nonmotile (“primary cilia”). The ciliary axoneme is an- chored in the basal body, a microtubule-organizing center de- rived from the older of the two . The transition zone at the junction of the basal body acts as a filter for the molecules that can pass into or out of the cilium. Nephrocystin-1 is local- ized at the transition zone of epithelial cells (111). During generation of cilia (), cilia elongate from the basal body by the addition of new axonemal subunits to the distal tip, the plus end of the microtubules. Axonemal and membrane components are transported in raft macromolecular particles (complex A and B) by so-called (IFT) along the axonemal doublet microtubules (Figure 3) (112). An- terograde transport toward the tip is driven by heterotrimeric kinesin 2, which contains motor subunits Kif3a and Kif3b and a nonmotor subunit. Mutations of Kif3a cause renal cysts in mice (113). also help form signaling complexes within the ciliary membrane. Retrograde transport back to the cell body occurs via the motor protein cytoplasmic dynein 1B (114). The unexpected convergence in primary cilia of proteins that underlie cystic kidney diseases is supported by the following findings: (1) there is high evolutionary conservation of the proteins involved (Figure 4, A through D), (2) most of these proteins interact with each other (Figure 4C), and (3) clinical phenotypes are related to tissue-specific expression in the sen- sory cilia (Figure 4, E through G).

Cilia: Conserved Modules to Sense Cell External Signals Figure 3. Cilia structure and intraflagellar transport (IFT). (A) A typ- It is becoming apparent that primary cilia are highly con- ical cilium consists of an axoneme of nine doublet microtubules. Each served structures for sensing of a wide variety of extracellular doublet arises from the inner two microtubules of the basal body cues by a broad variety of specialized tissues. A common microtubule triplets. The axoneme is surrounded by a specialized ciliary membrane that is separated from the cell membrane by a zone “theme” conserved through evolution seems to suggest that of transition fibers. (B) A cross-section of 9 ϩ 2 and 9 ϩ 0 cilium. Cilia many times when cells are to receive cues from the outside of are broadly divided into two types on the basis of the presence or the cell, they use a primary cilium. A broad range of cues can be absence of a central pair of microtubule singlets in the axoneme (9 ϩ received by specific ciliary receptors. These include photosen- 2or9ϩ 0 structure, respectively). Inner and outer dynein arms, sation (rhodopsin), mechanosensation (polycystin-1 and -2), which are usually associated with 9 ϩ 2 cilia, can be present in either osmosensation, and olfactory sensation (seven-membrane type of cilium and are important for ciliary motility. Ciliary assembly spanning olfactory receptors). The decision rules by which cells and maintenance is accomplished by IFT, which relies on the micro- place a specific receptor molecule into the cilium are unknown. tubule motor proteins kinesin 2 and cytoplasmic dynein to transport In general, it seems that the pathogenesis of ciliopathies is IFT protein complexes and their associated cargo up and down the based on an inability of epithelial cells to sense or process length of the cilium (depicted in A). Eb1, end-binding protein 1. extracellular cues (115). Adapted from Bisgrove and Yost (110a). J Am Soc Nephrol 18: 1855–1871, 2007 Nephronophthisis-Associated Ciliopathies 1861

Figure 4. The cilia/basal body hypothesis of renal cystic disease and related disorders. This illustration summarizes the new unifying pathogenic theory of cystic kidney disease, which states that virtually all proteins that are mutated in cystic kidney disease of humans, mice, or zebrafish (“cystoproteins”) show expression of their encoded proteins in primary cilia, basal bodies, or centrosomes. The horizontal axis symbolizes the flow of genetic information from genes over proteins to disease phenotypes. The vertical axis represents evolutionary time. (A) On the evolutionary scale, genes that are responsible for renal cystic disease in vertebrates are shown, including the unicellular organism Chlamydomonas reinhardtii. (B) Many of the orthologs of human cystic kidney disease genes are expressed in ciliated neurons of head and tail of the nematode Caenorhabditis elegans, where they lead to a male mating phenotype when knocked down (e.g., lov-1 or nph-4) (see E). Most proteins that are mutated in Bardet-Biedl syndrome (BBS) are conserved as basal body components of motile cilia (flagella) C. reinhardtii, where mutations in these genes lead to a phenotype of defective IFT or propulsion (see E). (C) Many cystoproteins directly interact with each other (e.g., NPHP1 and NPHP2/inversin). (D) Recently, it was discovered that the products of all genes that are mutated in cystic kidney diseases of humans, mice, or zebrafish show expression of their encoded proteins in primary cilia, basal bodies, or centrosomes. These findings placed primary cilia and centrosomes at the center of these disease processes (D has no vertical dimension). (E through G) The convergence of the pathogenesis at sensory cilia that serve distinct functions in different tissues may explain the broad organ involvement (pleiotropy) of NPHP and BBS that includes many different organ systems through defects of sensory cilia. Pleiotropic phenotypes in NPHP or BBS include cystic kidney disease, retinitis pigmentosa, infertility, diabetes, and other diseases of premature aging of organs.

Evolutionary Conservation of Cystoproteins in the lifespan of the worm has been demonstrated (121). In C. elegans, For many “cystoproteins,” the renal cystic phenotype is con- bbs-7 and bbs-8 are required for the correct localization/motility of the served among vertebrates. For example, mutations in inversin IFT proteins osm-5/polaris and che-11 (122). A bioinformatics ap- lead to NPHP type 2 in humans, mice, and zebrafish (Figure 4) proach based on the ciliary/basal body hypothesis of BBS pathogen- (17). At least two nephrocystins are conserved even in the nema- esis also helped to identify the cystoprotein BBS5 (123). Using bioin- tode C. elegans by amino acid sequence, functional features, and formatic screens for ciliary genes in combination with data from their expression patterns and knockdown phenotypes: Nphp-1 positional cloning, mutations in ARL6 were identified as being re- and nphp-4 are expressed in head (amphid) and tail (phasmid) sponsible for BBS type 3 (124,125). ARL6, a small GTPase, is specifi- neurons, which are ciliated osmosensor neurons of C. elegans (Figure 4A) (108,116). Localization of nephrocystin-1 and -4 to cally expressed in ciliated cells and undergoes IFT. some of these ciliated neurons overlaps with localization of the Evolutionary conservation of cystoproteins goes even fur- Ͼ cystoprotein orthologs polycystin-1 (lov-1) and polycystin-2 ther: Some cystoproteins have been conserved over 1.5 billion (pkd-2) and with many orthologs of BBS proteins (117). Knock- years of evolution from the unicellular organism Chlamydomo- down of nphp-1 and nphp-4 leads to impaired male mating behav- nas reinhardtii to vertebrates (Figure 4, A and B). C. reinhardtii ior (108), similar to what has been described for lov-1 and pkd-2 uses two motor cilia (flagella) for locomotion. Strikingly, mutants (118). These data have been confirmed and refined for nephrocystin-4 and at least six proteins that are mutated in BBS specific neuronal cell type (119,120). In addition, a role for nphp-4 are conserved in C. reinhardtii where they are part of the basal 1862 Journal of the American Society of Nephrology J Am Soc Nephrol 18: 1855–1871, 2007 body proteome (117,126). Defects of cystoprotein orthologs in Retinal Degeneration. The renal-retinal involvement in C. reinhardtii have deficient IFT and flagellar propulsion (127). SLSN can be explained by the fact that the primary cilium of renal epithelial cells is a structural equivalent of the connecting Nephrocystin Complexes cilium of photoreceptor cells in the retina. We have shown that Many cystoproteins participate in protein–protein interaction nephrocystin-5 and nephrocystin-6 are expressed in the con- complexes (Figure 4C). These interactions may partially explain necting cilia of photoreceptors (20,21). In analogy to motor why mutations in different NPHP genes lead to similar pheno- transport along the axoneme of primary cilia in kidney epithe- types. Some of the domains that occur in cystoproteins are shared lial cells, in the connecting cilia of photoreceptors, cargo is between different proteins. Coiled-coil domains, for example, oc- trafficked along microtubule tracks from the photoreceptor in- cur in six of eight proteins that are defective in NPHP-like diseases ner segment to the outer segment via a motor protein complex (nephrocystin-1, -2, -3, -5, and -6 and ALMS1). Nephrocystin-1 is that contains kinesin 2 and back to the cell body via a cytoplas- targeted to the transition zone of motile and primary cilia (111), mic dynein (Figure 5) (129). In this way, 10 billion molecules of and casein kinase 2–mediated phosphorylation of three critical the visual pigment rhodopsin are trafficked up and down the serine residues within a cluster of acidic amino acids in nephro- connecting cilia per human retina per day. cystin mediates phosphofurin acidic cluster sorting protein Liver Fibrosis. Both NPHP and BBS can be associated with (PACS)-1 binding and is essential for co-localization of nephrocys- liver fibrosis (40), whereas autosomal recessive PKD is associ- tin with PACS-1 at the base of cilia (128). The basal body/centro- ated with bile duct ectasia. Bile duct involvement in these cystic somal expression of proteins that are involved in NPHP has led to kidney diseases may be explained by the ciliary theory, because identification of mutations in NPHP6/CEP290 as the cause of the epithelial cells lining bile ducts (cholangiocytes) possess NPHP type 6 (JBTS type 5). Its gene product nephrocystin-6/ primary cilia (Figure 4, F and G). Cep290 (21) is part of the centrosomal proteome (86). Central Nervous System. Recent findings suggest that oc- Extrarenal Organ Involvement by Ciliary Dysfunction ulomotor apraxia type Cogan (associated with NPHP1 and A prominent feature of NPHP is that in a certain percentage NPHP4 mutations), cerebellar vermis hypoplasia (in JBTS), and of cases, there can be involvement of multiple organs (pleiot- mental retardation (in NPHP type 6) may be due to defects in ropy) other than the kidney. In some instances, there seems to microtubule-associated functions during neurite outgrowth be a genotype/phenotype correlation regarding pleiotropy. For and axonal guidance. Mechanotransport along microtubules instance, there is involvement of the retina in all known cases plays a role not only in intraciliary but also in axonal transport with mutations of NPHP5 or NPHP6. In other instances, such as (130). An example is the motor protein KIF3A, which is mu- NPHP1 mutations, the molecular basis of eye involvement is tated in a renal cystic mouse model and also plays a role in unknown. The pleiotropy of NPHP has now found a potential axonal transport (113). Because the malformations of the cere- explanation in the ciliary hypothesis of cystic kidney diseases bellum that occur in JBTS (37) consist of abnormal “wiring” of (Figure 4, E through G). The extrarenal organ involvement in decussating (crossing) neurons, impaired axonal outgrowth NPHP by organ system is discussed as follows. and axon guidance may be central to the neurologic defects in

Figure 5. Primary (nonmotile) cilia of renal epithelial cells and connecting cilia of retinal photoreceptors are analogous structures. In the primary cilium (A) of renal epithelial cells (B), “cargo” proteins are trafficked along the microtubule tracks from the region of the Golgi stack to the tip of the cilia via the motorprotein kinesin 2 and back down via cytoplasmic dynein 1b. (C) In an analogous manner, approximately 109 molecules of the visual pigment rhodopsin are transferred up and down the connecting cilia per human retina per day. Adapted from references (66,129). J Am Soc Nephrol 18: 1855–1871, 2007 Nephronophthisis-Associated Ciliopathies 1863

JBTS, in analogy to the lissencephaly phenotype that is caused caused by mutations in uromodulin, which has so far not been by the centrosomal proteins LIS1 and doublecortin (131). detected in cilia, basal bodies, or centrosomes. Congenital Cardiac Malformations. In a patient with infan- tile NPHP, we observed a ventricular septal defect as a congenital cardiac malformation (17). This developmental defect may be Focal Adhesion Hypothesis viewed as a “heterotaxy” (left–right orientation) phenotype that is When NPHP1 was first identified (15), we proposed a patho- caused by the same mechanism (68) that leads to situs inversus in genic hypothesis that tied in nephrocystin-1 with defects of this patient. The phenotypic combination of NPHP, situs inversus, cell–cell and cell–matrix signaling (102,138). This was based on and cardiac septal defect on the basis of inversin mutations is the finding that nephrocystin-1 contains an SH3 domain, local- observed in humans, mice, and zebrafish (17). izes to adherens junctions and focal adhesions of renal epithe- Obesity. Obesity is part of the clinical spectrum of the lial cells, and interacts with integral components of these struc- ciliopathy BBS, and excessive obesity has been described in tures, such as p130CAS (99,100). This “adherens junction/focal children with NPHP6 mutations after renal transplantation (B. adhesion hypothesis” of NPHP pathogenesis (102,138) recently Hoppe, University of Cologne, Germany, personal communi- was partially reconciled with the “cilia/centrosome” hypothe- cation, 2006). It is interesting that in the Bbs6 knockout mouse sis in an integrative hypothesis by showing that nephrocystin- model, obesity was associated with hyperphagia and decreased 4/nephroretinin in polarized epithelial cells co-localizes with ␤ activity of the mice (132). -catenin at cell–cell contact sites and to primary cilia, whereas in dividing cells, it localizes to centrosomes (101) (Figure 6, A Signaling Mechanisms Relevant for NPHP and B). Nephrocystins and other cystoproteins are expressed in dif- ferent subcellular compartments in a cell cycle–dependent The Wnt Pathway manner (21,133). These subcellular compartments include focal Recent results on inversin/NPHP2 shed light on the mech- adhesions, adherens junctions, cilia, basal bodies, centrosomes, anosensory hypothesis of bending of primary cilia by tubular and the mitotic spindle (Figure 6). It is still mostly unclear flow. They have provided data on downstream signaling events which of these subcellular localizations are most proximal to that are necessary to maintain normal tubular development and the pathogenesis of NPHP or other cystic kidney diseases. In morphology (104): In this model (Figure 6,C, F, and H) the canon- relation to these subcellular localizations, many hypotheses on ical Wnt signaling occurs primarily through ␤-catenin–dependent mechanisms of cystogenesis have been put forward. Among pathways in the absence of tubular flow. Stimulation of the pri- them are (1) the mechanosensory hypothesis of renal cilia func- mary cilium by flow, however, increases expression of inversin, tion, (2) participation in signaling at focal adhesions and adhe- which then reduces levels of cytoplasmic disheveled through pro- rens junctions, (3) a role in the maintenance of planar cell teasomal degradation and subsequently switches off the canonical polarity within the noncanonical Wnt signaling pathway, and pathway by allowing activation of the ␤-catenin destruction com- (4) a role in centrosome-related functions of cell-cycle regula- plex (96). When inversin is defective (as in NPHP type 2), the tion. These hypotheses are discussed next (Figure 6). canonical Wnt pathway will prevail and disrupt apical-basolateral polarity of the renal epithelium (96). Because planar cell polarity The Mechanosensory Hypothesis signaling is important for oriented cell division, it seems logical On the basis of the initial finding that bending the primary that Fisher et al. (139) recently were able to demonstrate abnormal 2ϩ cilium elicits Ca influx (134–136), it was shown that cilia can orientation of the mitotic spindle in two different rodent models of act as mechanosensors to sense fluid movement in the kidney cystic kidney disease. tubule, in cooperation with polycystins. In this model, polycys- tin-1 transmits the signal to polycystin-2, which is a TRP cal- ϩ cium channel (103). This produces sufficient Ca2 influx to Centrosomes ϩ induce Ca2 release from intracellular stores, which then reg- Nephrocystin-6, which is mutated in JBTS, is a component of ulates numerous signaling activities inside the cell that are the centrosomal proteome (86). In addition, BBS4 is instru- linked to cell-cycle regulation. It is thought that defects in mental in recruiting proteins to the pericentrosomal matrix, cell-cycle regulation may be ultimately responsible for the de- implicating the centrosome and its relation to cell-cycle control velopment of kidney cysts (103). In support of the ciliary hy- in the pathogenesis of BBS (117,140). The products of the genes pothesis of cystic kidney diseases, a motor protein of the kine- mutated in the ciliopathy Meckel-Gruber syndrome MKS1 (81) sin II family, KIF3A, which is involved in intraciliary transport, and MKS3 (82) recently were shown to localize to basal bodies was shown to cause a murine renal cystic disease when mu- and centrosomes (141) (Figure 6D). tated (113). Kramer-Zucker et al. (137) recently showed that cilia of larval zebrafish kidney tubules have a 9 ϩ 2 configuration Cell Cycle and are motile. Disruption of cilia structure or motility resulted A balance between hyperproliferation and apoptosis may in pronephric cyst formations, with left–right asymmetry de- play an important role in the pathogenesis of cystic kidney fects. Despite many data in support of the ciliary hypothesis, diseases (Figure 6, F and G). For example, whereas in PKD, some data are still hard to reconcile with this model; for exam- kidneys are grossly enlarged, in NPHP and BBS, kidney size ple, the autosomal dominant variant of NPHP, MCKD2, is remains normal and cysts grow at the expense of normal tissue 1864 Journal of the American Society of Nephrology J Am Soc Nephrol 18: 1855–1871, 2007

Figure 6. Nephrocystins localize to different subcellular compartments in a cell cycle–dependent manner and participate in multiple signaling pathways together with other “cystoproteins” (proteins that are mutated in cystic kidney disease). Functional complexes that play a role in the planar cell polarity (PCP) pathway are highlighted for focal adhesion (A), adherens junction (B), cilium (C), basal body (D), centrosome (E), nucleus (F), mitotic spindle (G), and the Wnt pathways (H). Cystoproteins are shown on colored background and in bold type using blue for nephrocystins (NPHP), orange for BBS proteins, green for polycystins (PKD) and (PKHD1), and yellow for cystoproteins of renal cystic mouse models. Proteins that are not bold have been described in the context of the pathogenesis of cystic kidney disease (e.g., as a binding partner to a bona fide cystoprotein). Associated proteins with no known role in cystic kidney diseases are shown on gray background. Black dots connect proteins that directly interact. (A) Focal adhesions. (1) Nephrocystin-1 directly interacts with the focal adhesion adapter protein p130Cas (“crk-associated substrate”) (99,100,102), which is a major mediator of focal adhesion assembly, binds to focal adhesion kinase, and mediates stress fiber formation (149). Nephrocystin-1 competes for binding to p130Cas with the proto-oncogene products Src and Fyn (99). (2) Nephrocystin-1 is in a protein complex with the focal adhesion proteins Pyk2/Fak2 (focal adhesion kinase 2), tensin (98), and filamin A and B. Its overexpression leads to activation of extracellular signal–regulated kinases 1 and 2 (ERK1 and ERK2) (98). (3) In children with NPHP, overexpression of ␣5␤1 integrin was described in proximal tubules, which most likely results from defective ␣6-integrin expression (150). (4) The knockout mouse models for tensin (151) and for the Rho GDI␣ gene (152) both exhibit an NPHP-like phenotype, thereby implicating further proteins of the focal adhesion signaling cascade in the pathogenesis of NPHP-like diseases. (B) Adherens junctions. (1) Nephrocystin-1 co-localizes with E-cadherin and p130Cas to adherens junctions. (2) The C-terminal half of nephrocystin, the “nephrocystin homology domain,” is able to promote nephrocystin self-association and epithelial adherens junctional targeting (100). (3) Disruption of this targeting leads to reduced transepithelial resistance (100). (4) Nephrocystin-4 is in a protein complex with nephrocystin-1, NPHP2/inversin, p130Cas, and Pyk2/Fak2 and has been J Am Soc Nephrol 18: 1855–1871, 2007 Nephronophthisis-Associated Ciliopathies 1865

(e vacuo). It seems that hyperproliferation may be the predom- NPHP: Defects of Tissue Differentiation inant mechanism in PKD-like diseases (142), whereas apoptosis and Maintenance is predominant in diseases of the NPHP and BBS group. A role It is striking that in renal cystic diseases (and in the associ- Ϫ/Ϫ Ϫ/Ϫ of apoptosis was in fact confirmed in the Bbs2 and Bbs4 ated extrarenal organ involvement), mutations of monogenic mouse models (143,144). Polycystin-1 and -2 signaling and the disease genes may lead to developmental defects (dysplasia), in renal cystic phenotype may be linked by a function of these which a structural organ defect is present at birth, but also to proteins in cell growth regulation. Polycystin-1 expression ac- degenerative defects (degeneration), in which organ structure tivates the JAK-STAT pathway, thereby upregulating and function are normal at birth but deteriorate over time. p21(waf1) and inducing cell-cycle arrest in G0/G1 (145). The Examples of this phenomenon from the side of cystic kidney cell-cycle arrest requires polycystin-2. Involvement of polycys- disease are autosomal recessive PKD, in which there is a struc- tin-1/2 signaling in the JAK/STAT pathway might explain how tural defect of the kidney present at birth, as opposed to NPHP, mutations of either gene can result in dysregulated growth in which kidneys are normal at birth (with the exception of (145). Very recently, this hypothesis was confirmed by demon- NPHP2) but degeneration leads to loss of renal function over stration that two mouse models of PKD (jck and cpk) can be the course of years. Regarding retinal involvement, NPHP can efficiently treated with the cyclin-dependent kinase inhibitor be associated with the developmental defect of retinal (R) roscovitine (146). coloboma (lack of retinal tissue) in JBTS but also with the

localized to adherens junctions in confluent MDCK cells (101). (C) Primary cilia. Recently, the development of a unifying hypothesis of renal cystogenesis was established (72). This hypothesis states that proteins that, when mutated, cause renal cystic disease in humans, mice, or zebrafish are part of a functional module, as defined by their subcellular localization to primary cilia, basal bodies, or centrosomes. This applies to polycystin-1 and -2; fibrocystin/polyductin; nephrocystin-1, -2 (inversin), -3, -4, and -5; BBS-associated proteins; cystin; polaris; ALMS1; and many others. Because nephrocystins interact, they are represented here as the “nephrocystin complex.” On the basis of positional cloning, mutations in the inversin gene were identified as causing infantile NPHP (type 2) (17). This established a link between the pathogenesis of NPHP and disease mechanisms of PKD (17), in which nephrocystin-1 interacts with both inversin and ␤-tubulin. Because ␤-tubulin is a major component of primary cilia, this led to demonstration of co-localization of all three proteins in the primary renal cilia of epithelial cells (17). The complex also contains NPHP3, which is mutated in adolescent NPHP (type 3) and in the renal cystic mouse model pcy. The ciliary hypothesis of cystic kidney disease was confirmed by revealing that Nphp3 mRNA was expressed in kidney, retinal connecting cilia, ciliated bile ducts, and the node, which regulates left–right body axis in mice (18), and by identifying mutations in nephrocystin-5 (IQCB1), which co-localizes with calmodulin to primary cilia of renal epithelial cells and retinal connecting cilia (20). (D) Basal bodies. Basal bodies are short cylindrical arrays of microtubules and other proteins that are found at the base of cilia and that organize the assembly of the ciliary axoneme. They are analogous to centrosomes. Nephrocystins localize to the transition zone of basal bodies (111). Proteins that are mutated in BBS are components of the basal body transitional zone and are highly conserved in evolution. (E) Centrosomes. For a protein that is mutated in the related disease BBS4, it was shown that BBS4 is instrumental in recruiting proteins (e.g., PCM1) to the pericentrosomal matrix, confirming the role of centrosomal function in the pathogenesis of BBS (116). (F) Transcriptional programs. The transcription factor HNF1␤ regulates transcription of multiple genes that are mutated in cystic kidney disease–related genes (153). (G) Cell-cycle regulation. The hypotheses of functional involvement of the nephrocystin complex at focal adhesions and adherens junction on the one hand and the ciliary/centrosomal hypotheses on the other hand may be integrated by demonstration that different locations of the complex may predominate in a cell cycle–dependent manner. This is evidenced by the findings that inversin/nephrocystin-2 (17), nephrocystin-4 (101), and nephrocystin-6 (21) expression occurs in a cell cycle–dependent manner. Inversin exhibits a dynamic expression pattern in MDCK cells that show expression at centrosomes in early prophase, at spindle poles in metaphase and anaphase, and at the midbody in cells that undergo cytokinesis (133,154). Nephrocystin-4 was detected in MDCK cells at centrosomes of dividing cells and in polarized cells close at the cytoskeleton and in the vicinity of the cortical actin cytoskeleton, with co-localization of p130Cas, Pyk2, and ␤-catenin (101). (H) Wnt pathways. Recent data demonstrated that in renal tubules, inversin/NPHP2 may induce switching from the canonical to the noncanonical Wnt signaling pathway in response to flow sensing by primary cilia of renal tubular cells (104). It is thought that this function of inversin is important to maintain renal epithelial cell polarity (96). The hypothesis that the renal cystic disease phenotype is due to defects in the maintenance of planar cell polarity seems plausible for multiple reasons: (1) It would reconcile previous functional hypotheses, because focal adhesions, adherens junctions, cilia, centrosomes, basal bodies, and regulation of the cell cycle all play a pivotal role in the regulation and maintenance of planar cell polarity (155); (2) because planar cell polarity plays an important role in developmental morphogenesis and also in the regeneration of differentiated tissue, a defect in planar cell polarity may explain both the occurrence of cysts during organogenesis and degenerative cystogenesis as it occurs in NPHP; and (3) the mechanism of convergent extension, which may be central to renal tubular morphology, was shown to be disturbed in many renal cystic diseases (156). APC, adenomatous polyposis coli protein; APC2, anaphase promoting complex subunit 2; CBP, CREB-binding protein; CREB, cAMP response element–binding protein; CK1␧, casein kinase ␧; DKK, Dickkopf-related protein; CK2, casein kinase II; GAP, RhoGTPase activating protein; GEF, guanine nucleotide exchange factor; GSK3, glycogen synthase kinase 3; JUNK, JUN kinase; PCM1, pericentriolar material 1; p150, p150glued/dynactin-1; PKC, protein kinase C; ROCK, Rho-associated protein kinase. 1866 Journal of the American Society of Nephrology J Am Soc Nephrol 18: 1855–1871, 2007 degenerative processes of tapetoretinal degeneration/retinitis dorf F, Reis A, Hildebrandt F: Identification of a new gene pigmentosa (21,44,109). The joint occurrence of developmental locus for adolescent nephronophthisis, on chromosome and degenerative defect may be explained by the fact that many 3q22 in a large Venezuelan pedigree. Am J Hum Genet 66: developmental transcriptional programs are reinitiated for 118–127, 2000 mechanisms of tissue maintenance and repair. Because planar 8. Ala-Mello S, Kivivuori SM, Ronnholm KA, Koskimies O, Siimes MA: Mechanism underlying early anaemia in chil- cell polarity plays an important role in developmental morpho- dren with familial juvenile nephronophthisis. Pediatr Neph- genesis and also in the regeneration of differentiated tissue, a rol 10: 578–581, 1996 defect in planar cell polarity may explain both occurrence of 9. Blowey DL, Querfeld U, Geary D, Warady BA, Alon U: cysts during organogenesis and degenerative cystogenesis as it Ultrasound findings in juvenile nephronophthisis. Pediatr occurs in NPHP. Nephrol 10: 22–24, 1996 9a. Hildebrandt F, Sayer JA, Jungers P, Gru¨nfield J-P: Therapeutic Approaches to NPHP Nephronophthisis—medullary cystic and medullary No effective prophylaxis or treatment is available for NPHP. sponge kidney disease. In: Diseases of the Kidney and Urinary The only therapeutic options are supportive treatment once Tract, edited by Schrier WB, Philadelphia, Lippincott, Wil- chronic renal failure has developed and and transplan- liams & Wilkins, 2007, pp 478–501 tation for terminal renal failure. An important future challenge 10. Waldherr R, Lennert T, Weber HP, Fodisch HJ, Scharer K: The nephronophthisis complex. A clinicopathologic study will be the development of therapies that capitalize on what we in children. Virchows Arch A Pathol Anat Histol 394: 235– have learned about the biology of NPHP and other cystic 254, 1982 diseases of the kidney. Gattone et al. (106) recently showed that 11. Zollinger HU, Gaboardi F, Imbasciati E, Lennert T: the renal cystic phenotype of pcy mice, which is the equivalent Nephronophthisis (medullary cystic disease of the kidney). of human NPHP type 3, can be strongly mitigated or even A study using electron microscopy, immunofluorescence, reversed by treatment with the vasopressin V2 receptor antag- and a review of the morphological findings. Helv Paediatr onist OPC31260. Similar results were obtained using a pkd2 Acta 35: 509–530, 1980 mouse model (147). This effect is thought to be mediated by a 12. Gardner KD Jr: Juvenile nephronophthisis and renal med- reduction in intracellular cAMP levels, a finding that awaits ullary cystic disease. Perspect Nephrol Hypertens 4: 173–185, reconciliation with the ciliary/centrosome hypothesis of 1976 NPHP. In this context, it is interesting that nephrocystin-6 13. Kleinknecht C: The inheritance of nephronophthisis. In: Inheritance of Kidney and Urinary Tract Diseases, Vol 9, edited directly interacts with the transcription factor ATF4/CREB2, by Avner ED, Boston, Kluwer Academic Publishers, 1989, which plays a role in the regulation of intracellular cAMP levels pp 299–305 (21). Additional therapeutic approaches are being considered 14. Steel BT, Lirenman DS, Battie CW: Nephronophthisis. Am J for PKD (148). Med 68: 531–538, 1980 15. Hildebrandt F, Otto E, Rensing C, Nothwang HG, Vollmer Disclosures M, Adolphs J, Hanusch H, Brandis M: A novel gene en- None. coding an SH3 domain protein is mutated in nephronoph- thisis type 1. Nat Genet 17: 149–153, 1997 16. Saunier S, Calado J, Heilig R, Silbermann F, Benessy F, References Morin G, Konrad M, Broyer M, Gubler MC, Weissenbach J, 1. Hildebrandt F, Otto E: Cilia and centrosomes: A unifying Antignac C: A novel gene that encodes a protein with a pathogenic concept for cystic kidney disease? Nat Rev putative src homology 3 domain is a candidate gene for Genet 6: 928–930, 2005 familial juvenile nephronophthisis. Hum Mol Genet 6: 2317– 2. Smith C, Graham J: Congenital medullary cysts of the 2323, 1997 kidneys with severe refractory anemia. Am J Dis Child 69: 17. Otto EA, Schermer B, Obara T, O’Toole JF, Hiller KS, 369–377, 1945 Mueller AM, Ruf RG, Hoefele J, Beekmann F, Landau D, 3. Fanconi G, Hanhart E, von Albertini A, Uhlinger E, Dolivo Foreman JW, Goodship JA, Strachan T, Kispert A, Wolf G, Prader A: Familial, juvenile nephronophthisis (idio- MT, Gagnadoux MF, Nivet H, Antignac C, Walz G, Drum- pathic parenchymal contracted kidney) [in German]. Helv mond IA, Benzing T, Hildebrandt F: Mutations in INVS Pediatr Acta 6: 1–49, 1951 encoding inversin cause nephronophthisis type 2, linking 4. Hildebrandt F: Juvenile nephronophthisis. In: Pediatric Ne- renal cystic disease to the function of primary cilia and phrology, edited by Harmon WE, Baltimore, Williams & left-right axis determination. Nat Genet 34: 413–420, 2003 Wilkins, 2004, pp 665–673 18. Olbrich H, Fliegauf M, Hoefele J, Kispert A, Otto E, Volz A, 5. Gagnadoux MF, Bacri JL, Broyer M, Habib R: Infantile Wolf MT, Sasmaz G, Trauer U, Reinhardt R, Sudbrak R, chronic tubulo-interstitial nephritis with cortical micro- Antignac C, Gretz N, Walz G, Schermer B, Benzing T, cysts: Variant of nephronophthisis or new disease entity. Hildebrandt F, Omran H: Mutations in a novel gene, Pediatr Nephrol 3: 50–55, 1989 NPHP3, cause adolescent nephronophthisis, tapeto-retinal 6. Hildebrandt F, Waldherr R, Kutt R, Brandis M: The degeneration and hepatic fibrosis Nat Genet 34: 455–459, nephronophthisis complex: Clinical and genetic aspects. 2003 Clin Investig 70: 802–808, 1992 19. Otto E, Hoefele J, Ruf R, Mueller AM, Hiller KS, Wolf MT, 7. Omran H, Fernandez C, Jung M, Haffner K, Fargier B, Schuermann MJ, Becker A, Birkenhager R, Sudbrak R, Villaquiran A, Waldherr R, Gretz N, Brandis M, Ruschen- Hennies HC, Nurnberg P, Hildebrandt F: A gene mutated J Am Soc Nephrol 18: 1855–1871, 2007 Nephronophthisis-Associated Ciliopathies 1867

in nephronophthisis and retinitis pigmentosa encodes a nal insufficiency in children and adolescents. The 1995 novel protein, nephroretinin, conserved in evolution. Am J Annual Report of the North American Pediatric Renal Hum Genet 71: 1167–1171, 2002 Transplant Cooperative Study. Pediatr Nephrol 11: 49–64, 20. Otto EA, Loeys B, Khanna H, Hellemans J, Sudbrak R, Fan 1997 S, Muerb U, O’Toole JF, Helou J, Attanasio M, Utsch B, 33. Avner ED: Medullary cystic disease and medullary sponge Sayer JA, Lillo C, Jimeno D, Coucke P, De Paepe A, Rein- kidney. In: Primer on Kidney Diseases, edited by Greenberg hardt R, Klages S, Tsuda M, Kawakami I, Kusakabe T, A, Boston, Academic Press, 1994, pp 255–262 Omran H, Imm A, Tippens M, Raymond PA, Hill J, Beales 34. Senior B, Friedmann AI, Braudo JL: Juvenile familial ne- P, He S, Kispert A, Margolis B, Williams DS, Swaroop A, phropathy with tapetoretinal degeneration: A new oculo- Hildebrandt F: Nephrocystin-5, a ciliary IQ domain pro- renal dystrophy. Am J Ophthalmol 52: 625–633, 1961 tein, is mutated in Senior-Loken syndrome and interacts 35. Loken AC, Hanssen O, Halvorsen S, Jolster NJ: Hereditary with RPGR and calmodulin. Nat Genet 37: 282–288, 2005 renal dysplasia and blindness. Acta Paediatr 50: 177–184, 21. Sayer JA, Otto EA, O’Toole JF, Nurnberg G, Kennedy MA, 1961 Becker C, Hennies HC, Helou J, Attanasio M, Fausett BV, 36. Saraiva JM, Baraitser M: Joubert syndrome: A review. Am J Utsch B, Khanna H, Liu Y, Drummond I, Kawakami I, Med Genet 43: 726–731, 1992 Kusakabe T, Tsuda M, Ma L, Lee H, Larson RG, Allen SJ, 37. Valente EM, Marsh SE, Castori M, Dixon-Salazar T, Bertini Wilkinson CJ, Nigg EA, Shou C, Lillo C, Williams DS, E, Al-Gazali L, Messer J, Barbot C, Woods CG, Boltshauser Hoppe B, Kemper MJ, Neuhaus T, Parisi MA, Glass IA, E, Al-Tawari AA, Salpietro CD, Kayserili H, Sztriha L, Petry M, Kispert A, Gloy J, Ganner A, Walz G, Zhu X, Gribaa M, Koenig M, Dallapiccola B, Gleeson JG: Distin- Goldman D, Nurnberg P, Swaroop A, Leroux MR, Hilde- guishing the four genetic causes of Jouberts syndrome- brandt F: The centrosomal protein nephrocystin-6 is mu- related disorders. Ann Neurol 57: 513–519, 2005 tated in Joubert syndrome and activates transcription fac- 38. Saunier S et al.: Large deletions of the NPH1 region in tor ATF4. Nat Genet 38: 674–681, 2006 Cogan syndrome (CS) associated with familial juvenile 22. Hildebrandt F, Strahm B, Nothwang HG, Gretz N, nephronophthisis (NPH) [Abstract]. Am J Hum Genet 61: Schnieders B, Singh-Sawhney I, Kutt R, Vollmer M, Bran- A346, 1997 dis M: Molecular genetic identification of families with 39. Baris H, Bejjani BA, Tan WH, Coulter DL, Martin JA, Storm juvenile nephronophthisis type 1: Rate of progression to AL, Burton BK, Saitta SC, Gajecka M, Ballif BC, Irons MB, renal failure. APN Study Group. Arbeitsgemeinschaft fur Shaffer LG, Kimonis VE: Identification of a novel polymor- Padiatrische Nephrologie. Kidney Int 51: 261–269, 1997 phism: The duplication of the NPHP1 (nephronophthisis 1) 23. Haider NB, Carmi R, Shalev H, Sheffield VC, Landau D: A gene. Am J Med Genet A 140: 1876–1879, 2006 Bedouin kindred with infantile nephronophthisis demon- 40. Boichis H, Passwell J, David R, Miller H: Congenital he- strates linkage to chromosome 9 by homozygosity map- patic fibrosis and nephronophthisis. A family study. Q ping. Am J Hum Genet 63: 1404–1410, 1998 J Med 42: 221–233, 1973 24. Gretz N: Rate of deterioration of renal function in juvenile 41. Mainzer F, Saldino RM, Ozonoff MB, Minagi H: Familial nephronophthisis. Pediatr Nephrol 3: 56–60, 1989 nephropathy associated with retinitis pigmentosa, cerebel- 25. Christodoulou K, Tsingis M, Stavrou C, Eleftheriou A, lar ataxia and skeletal abnormalities. Am J Med 49: 556–562, Papapavlou P, Patsalis PC, Ioannou P, Pierides A, Con- 1970 stantinou Deltas C: Chromosome 1 localization of a gene 42. O’Toole JF, Otto E, Frishberg Y, Hildebrandt F: Retinitis for autosomal dominant medullary cystic kidney disease. pigmentosa and renal failure in a patient with mutations in Hum Mol Genet 7: 905–911, 1998 inversin [Abstract]. J Am Soc Nephrol 15: 215A, 2004 26. Scolari F, Ghiggeri GM, Amoroso A, Caridi GL, Aridon P: 43. Contreras DB, Espinoza JS: Discussion clinica y anato- Genetic heterogeneity for autosomal dominant medullary mopatologica de enfermos que presentaron un problema cystic kidney disease (ADMCKD) [Abstract]. JAmSoc diagnostico [in Spanish]. Pediatrica (Santiago) 3: 271–282, Nephrol 9: 393A, 1998 1960 27. Potter DE, Holliday MA, Piel CF, Feduska NJ, Belzer FO, 44. Saraux H, Dhermy P, Fontaine JL, Boulesteix J, Lasfargue Salvatierra O Jr: Treatment of end-stage renal disease in G, Grenet P, NЈghiem M, Laplane R: Senior-Loken retino- children: A 15-year experience. Kidney Int 18: 103–109, 1980 tubular degeneration [in French]. Arch Ophtalmol Rev Gen 28. Pistor K, Scharer K, Olbing H, Tamminen-Mobius T: Chil- Ophtalmol 30: 683–696, 1970 dren with chronic renal failure in the Federal Republic of 45. Medhioub M, Cherif D, Benessy F, Silbermann F, Gubler Germany: II. Primary renal diseases, age and intervals MC, Le Paslier D, Cohen D, Weissenbach J, Beckmann J, from early renal failure to renal death. Arbeitsgemein- Antignac C: Refined mapping of a gene (NPH1) causing schaft fur Padiatrische Nephrologie. Clin Nephrol 23: 278– familial juvenile nephronophthisis and evidence for ge- 284, 1985 netic heterogeneity. Genomics 22: 296–301, 1994 29. Cantani A, Bamonte G, Ceccoli D: Familial juvenile 46. Biersdorf WR: The clinical utility of the foveal electroreti- nephronophthisis. Clin Pediatr 25: 90–95, 1986 nogram: A review. Doc Ophthalmol 73: 313–325, 1989 30. Betts PR, Forest-Hay I: Juvenile nephronophthisis. Lancet 2: 47. Joubert M, Eisenring JJ, Robb JP, Andermann F: Familial 475–478, 1973 agenesis of the cerebellar vermis. A syndrome of episodic 31. Green A, Allos M, Donohoe J, Carmody M, Walshe J: hyperpnea, abnormal eye movements, ataxia, and retarda- Prevalence of hereditary renal disease. Ir Med J 83: 11–13, tion. Neurology 19: 813–825, 1969 1990 48. Parisi MA, Dobyns WB: Human malformations of the mid- 32. Warady BA, Hebert D, Sullivan EK, Alexander SR, Tejani brain and hindbrain: Review and proposed classification A: Renal transplantation, chronic dialysis, and chronic re- scheme. Mol Genet Metab 80: 36–53, 2003 1868 Journal of the American Society of Nephrology J Am Soc Nephrol 18: 1855–1871, 2007

49. Gleeson JG, Keeler K, Parisi MA, Marsh SE, Chance PF, Al-Gazali L, Gururaj A, Gleeson JG: Linkage analysis in Glass IA, Graham Jr JM, Maria BL, Barkovich AJ, Dobyns families with Joubert syndrome plus oculo-renal involve- WB: Molar tooth sign of the midbrain-hindbrain junction: ment identifies the CORS2 locus on chromosome 11p12– Occurrence in multiple distinct syndromes. Am J Med Genet q13.3. Am J Hum Genet 73: 656–662, 2003 125A: 125–134, discussion 117, 2004 61. Delaney V, Mullaney J, Bourke E: Juvenile nephronophthi- 50. Castori M, Valente EM, Donati MA, Salvi S, Fazzi E, Pro- sis, congenital hepatic fibrosis and retinal hypoplasia in copio E, Galluccio T, Emma F, Dallapiccola B, Bertini E; twins. Q J Med 47: 281–290, 1978 Italian MTS Study Group: NPHP1 gene deletion is a rare 62. Proesmans W, Van Damme B, Macken J: Nephronophthisis cause of Joubert syndrome related disorders. J Med Genet and tapetoretinal degeneration associated with liver fibro- 42: e9, 2005 sis. Clin Nephrol 3: 160–164, 1975 51. Utsch B, Sayer JA, Attanasio M, Pereira RR, Eccles M, 63. Rayfield EJ, McDonald FD: Red and blonde hair in renal Hennies HC, Otto EA, Hildebrandt F: Identification of the medullary cystic disease. Arch Intern Med 130: 72–75, 1972 first AHI1 gene mutations in nephronophthisis-associated 64. Mochizuki T, Saijoh Y, Tsuchiya K, Shirayoshi Y, Takai S, Joubert syndrome. Pediatr Nephrol 21: 32–35, 2006 Taya C, Yonekawa H, Yamada K, Nihei H, Nakatsuji N, 52. Parisi MA, Doherty D, Eckert ML, Shaw DW, Ozyurek H, Overbeek PA, Hamada H, Yokoyama T: Cloning of inv,a Aysun S, Giray O, Al Swaid A, Al Shahwan S, Dohayan N, gene that controls left/right asymmetry and kidney devel- Bakhsh E, Indridason OS, Dobyns WB, Bennett CL, Chance opment. Nature 395: 177–181, 1998 PF, Glass IA: AHI1 mutations cause both retinal dystrophy 65. Morgan D, Turnpenny L, Goodship J, Dai W, Majumder K, and renal cystic disease in Joubert syndrome. J Med Genet Matthews L, Gardner A, Schuster G, Vien L, Harrison W, Elder 43: 334–339, 2006 FF, Penman-Splitt M, Overbeek P, Strachan T: Inversin, a novel 53. Valente EM, Silhavy JL, Brancati F, Barrano G, Krish- gene in the vertebrate left-right axis pathway, is partially deleted naswami SR, Castori M, Lancaster MA, Boltshauser E, in the inv mouse. Nat Genet 20: 149–156, 1998 Boccone L, Al-Gazali L, Fazzi E, Signorini S, Louie CM, 66. Igarashi P, Somlo S: Genetics and pathogenesis of polycys- Bellacchio E; International Joubert Syndrome Related Dis- tic kidney disease. J Am Soc Nephrol 13: 2384–2398, 2002 orders Study Group; Bertini E, Dallapiccola B, Gleeson JG: 67. Hirokawa N, Noda Y, Okada Y: Kinesin and dynein su- Mutations in CEP290, which encodes a centrosomal pro- perfamily proteins in organelle transport and cell division. tein, cause pleiotropic forms of Joubert syndrome. Nat Curr Opin Cell Biol 10: 60–73, 1998 Genet 38: 623–625, 2006 68. McGrath J, Somlo S, Makova S, Tian X, Brueckner M: Two 54. Ferland RJ, Eyaid W, Collura RV, Tully LD, Hill RS, Al- populations of node monocilia initiate left-right asymme- Nouri D, Al-Rumayyan A, Topcu M, Gascon G, Bodell A, try in the mouse. Cell 114: 61–73, 2003 Shugart YY, Ruvolo M, Walsh CA: Abnormal cerebellar 69. Pennekamp P, Karcher C, Fischer A, Schweickert A, Skry- development and axonal decussation due to mutations in abin B, Horst J, Blum M, Dworniczak B: The ion channel AHI1 in Joubert syndrome. Nat Genet 36: 1008–1013, 2004 polycystin-2 is required for left-right axis determination in 55. Dixon-Salazar T, Silhavy JL, Marsh SE, Louie CM, Scott LC, mice. Curr Biol 12: 938–943, 2002 Gururaj A, Al-Gazali L, Al-Tawari AA, Kayserili H, Sztriha 70. Alton DJ, McDonald P: Urographic findings in the Bardet- L, Gleeson JG: Mutations in the AHI1 gene, encoding jou- Biedl syndrome, formerly the Laurence-Moon-Biedl syn- berin, cause Joubert syndrome with cortical polymicro- gyria. Am J Hum Genet 75: 979–987, 2004 drome. Radiology 109: 659–663, 1973 56. Betz R, Rensing C, Otto E, Mincheva A, Zehnder D, Lichter 71. Green JS, Parfrey PS, Harnett JD, Farid NR, Cramer BC, P, Hildebrandt F: Children with ocular motor apraxia type Johnson G, Heath O, McManamon PJ, O’Leary E, Pryse- Cogan carry deletions in the gene (NPHP1) for juvenile Phillips W: The cardinal manifestations of Bardet-Biedl nephronophthisis. J Pediatr 136: 828–831, 2000 syndrome, a form of Laurence-Moon-Biedl syndrome. 57. Mollet G, Salomon R, Gribouval O, Silbermann F, Bacq D, N Engl J Med 321: 1002–1009, 1989 Landthaler G, Milford D, Nayir A, Rizzoni G, Antignac C, 72. Watnick T, Germino G: From cilia to cyst. Nat Genet 34: Saunier S: The gene mutated in juvenile nephronophthisis 355–356, 2003 type 4 encodes a novel protein that interacts with nephro- 73. Badano JL, Katsanis N: Beyond Mendel: An evolving cystin. Nat Genet 32: 300–305, 2002 view of human genetic disease transmission. Nat Rev Genet 58. Jen JC, Chan WM, Bosley TM, Wan J, Carr JR, Rub U, 3: 779–789, 2002 Shattuck D, Salamon G, Kudo LC, Ou J, Lin DD, Salih MA, 74. Donaldson MD, Warner AA, Trompeter RS, Haycock GB, Kansu T, Al Dhalaan H, Al Zayed Z, MacDonald DB, Chantler C: Familial juvenile nephronophthisis, Jeune’s Stigsby B, Plaitakis A, Dretakis EK, Gottlob I, Pieh C, syndrome, and associated disorders. Arch Intern Med 60: Traboulsi EI, Wang Q, Wang L, Andrews C, Yamada K, 426–434, 1985 Demer JL, Karim S, Alger JR, Geschwind DH, Deller T, 75. Jeune M, Beraud C, Carron R: Asphyxiating thoracic dys- Sicotte NL, Nelson SF, Baloh RW, Engle EC: Mutations in trophy with familial characteristics [in French]. Arch Fr a human ROBO gene disrupt hindbrain axon pathway Pediatr 12: 886–891, 1955 crossing and morphogenesis. Science 304: 1509–1513, 2004 76. Amirou M, Bourdat-Michel G, Pinel N, Huet G, Gaultier J, 59. Saar K, Al-Gazali L, Sztriha L, Rueschendorf F, Nur-E- Cochat P: Successful renal transplantation in Jeune syn- Kamal M, Reis A, Bayoumi R: Homozygosity mapping in drome type 2. Pediatr Nephrol 12: 293–294, 1998 families with Joubert syndrome identifies a locus on chro- 77. Sarimurat N, Elcioglu N, Tekant GT, Elicevik M, Yeker D: mosome 9q34.3 and evidence for genetic heterogeneity. Jeune’s asphyxiating thoracic dystrophy of the newborn. Am J Hum Genet 65: 1666–1671, 1999 Eur J Pediatr Surg 8: 100–101, 1998 60. Keeler LC, Marsh SE, Leeflang EP, Woods CG, Sztriha L, 78. Moudgil A, Bagga A, Kamil ES, Rimoin DL, Lachman RS, J Am Soc Nephrol 18: 1855–1871, 2007 Nephronophthisis-Associated Ciliopathies 1869

Cohen AH, Jordan SC: Nephronophthisis associated with physiological, and genetic aspects of the disorder. Medicine Ellis-van Creveld syndrome. Pediatr Nephrol 12: 20–22, 1998 (Baltimore) 52: 53–71, 1973 79. Di Rocco M, Picco P, Arslanian A, Restagno G, Perfumo F, 90. Tsimaratos M, Sarles J, Sigaudy S, Philip N: Renal and Buoncompagni A, Gattorno M, Borrone C: Retinitis pig- retinal involvement in the Sensenbrenner syndrome. Am J mentosa, hypopituitarism, nephronophthisis, and mild Med Genet 77: 337, 1998 skeletal dysplasia (RHYNS): A new syndrome Am J Med 91. Costet C, Betis F, Berard E, Tsimaratos M, Sigaudy S, Genet 73: 1–4, 1997 Antignac C, Gastaud P: Pigmentosum retinis and tubulo- 80. Marshall JD, Bronson RT, Collin GB, Nordstrom AD, interstitial nephronophtisis in Sensenbrenner syndrome: A Maffei P, Paisey RB, Carey C, Macdermott S, Russell-Eggitt case report [in French]. J Fr Ophtalmol 23: 158–160, 2000 I, Shea SE, Davis J, Beck S, Shatirishvili G, Mihai CM, 92. Kumada S, Hayashi M, Arima K, Nakayama H, Sugai K, Sasaki Hoeltzenbein M, Pozzan GB, Hopkinson I, Sicolo N, Nag- M, Kurata K, Nagata M: Renal disease in Arima syndrome is gert JK, Nishina PM: New Alstrom syndrome phenotypes nephronophthisis as in other Joubert-related cerebello-oculo-re- based on the evaluation of 182 cases. Arch Intern Med 165: nal syndromes. Am J Med Genet A 131: 71–76, 2004 675–683, 2005 93. Satran D, Pierpont ME, Dobyns WB: Cerebello-oculo-renal 81. Kyttala M, Tallila J, Salonen R, Kopra O, Kohlschmidt N, syndromes including Arima, Senior-Loken and COACH Paavola-Sakki P, Peltonen L, Kestila M: MKS1, encoding a syndromes: More than just variants of Joubert syndrome. component of the flagellar apparatus basal body proteome, is Am J Med Genet 86: 459–469, 1999 mutated in Meckel syndrome. Nat Genet 38: 155–157, 2006 94. Chance PF, Cavalier L, Satran D, Pellegrino JE, Koenig M, 82. Smith UM, Consugar M, Tee LJ, McKee BM, Maina EN, Dobyns WB: Clinical nosologic and genetic aspects of Jou- Whelan S, Morgan NV, Goranson E, Gissen P, Lilliquist S, bert and related syndromes. J Child Neurol 14: 660–666, Aligianis IA, Ward CJ, Pasha S, Punyashthiti R, Malik discussion 669–672, 1999 Sharif S, Batman PA, Bennett CP, Woods CG, McKeown C, 95. Ala-Mello S, Kaariainen H, Koskimies O: Nephronophthi- Bucourt M, Miller CA, Cox P, Algazali L, Trembath RC, sis and ulcerative colitis in siblings: A new association. Torres VE, Attie-Bitach T, Kelly DA, Maher ER, Gattone Pediatr Nephrol 16: 507–509, 2001 VH 2nd, Harris PC, Johnson CA: The transmembrane pro- 96. Germino GG: Linking cilia to Wnts. Nat Genet 37: 455–457, tein meckelin (MKS3) is mutated in Meckel-Gruber syn- 2005 drome and the wpk rat. Nat Genet 38: 191–196, 2006 97. Otto E, Kispert A, Schatzle, Lescher B, Rensing C, Hilde- 83. Baala L, Romano S, Khaddour R, Saunier S, Smith UM, brandt F: Nephrocystin: and sequence Audollent S, Ozilou C, Faivre L, Laurent N, Foliguet B, conservation between human, mouse, and Caenorhabditis Munnich A, Lyonnet S, Salomon R, Encha-Razavi F, elegans. J Am Soc Nephrol 11: 270–282, 2000 Gubler MC, Boddaert N, de Lonlay P, Johnson CA, Veke- 98. Benzing T, Gerke P, Hopker K, Hildebrandt F, Kim E, Walz mans M, Antignac C, Attie-Bitach T: The Meckel-Gruber G: Nephrocystin interacts with Pyk2, p130(Cas), and tensin syndrome gene, MKS3, is mutated in Joubert syndrome. and triggers phosphorylation of Pyk2. Proc Natl Acad Sci Am J Hum Genet 80: 186–194, 2007 USA98: 9784–9789, 2001 84. Collin GB, Marshall JD, Ikeda A, So WV, Russell-Eggitt I, 99. Donaldson JC, Dempsey PJ, Reddy S, Bouton AH, Coffey Maffei P, Beck S, Boerkoel CF, Sicolo N, Martin M, Nishina RJ, Hanks SK: Crk-associated substrate p130(Cas) interacts PM, Naggert JK: Mutations in ALMS1 cause obesity, type with nephrocystin and both proteins localize to cell-cell 2 diabetes and neurosensory degeneration in Alstrom syn- contacts of polarized epithelial cells. Exp Cell Res 256: 168– drome. Nat Genet 31: 74–78, 2002 178, 2000 85. Hearn T, Renforth GL, Spalluto C, Hanley NA, Piper K, Brick- 100. Donaldson JC, Dise RS, Ritchie MD, Hanks SK: Nephro- wood S, White C, Connolly V, Taylor JF, Russell-Eggitt I, Bon- cystin-conserved domains involved in targeting to epithe- neau D, Walker M, Wilson DI: Mutation of ALMS1, a large gene lial cell-cell junctions, interaction with filamins, and estab- with a tandem repeat encoding 47 amino acids, causes Alstrom lishing cell polarity. J Biol Chem 277: 29028–29035, 2002 syndrome. Nat Genet 31: 79–83, 2002 101. Mollet G, Silbermann F, Delous M, Salomon R, Antignac C, 86. Andersen JS, Wilkinson CJ, Mayor T, Mortensen P, Nigg Saunier S: Characterization of the nephrocystin/nephro- EA, Mann M: Proteomic characterization of the human cystin-4 complex and subcellular localization of nephro- centrosome by protein correlation profiling. Nature 426: cystin-4 to primary cilia and centrosomes. Hum Mol Genet 570–574, 2003 14: 645–656, 2005 87. Hearn T, Spalluto C, Phillips VJ, Renforth GL, Copin N, 102. Hildebrandt F, Otto E: Molecular genetics of nephronoph- Hanley NA, Wilson DI: Subcellular localization of ALMS1 thisis and medullary cystic kidney disease. J Am Soc Neph- supports involvement of centrosome and basal body dys- rol 11: 1753–1761, 2000 function in the pathogenesis of obesity, insulin resistance, 103. Nauli SM, Alenghat FJ, Luo Y, Williams E, Vassilev P, Li X, and type 2 diabetes. Diabetes 54: 1581–1587, 2005 Elia AE, Lu W, Brown EM, Quinn SJ, Ingber DE, Zhou J: 88. Alstrom CH, Hallgren B, Nilsson LB, Asander H: Retinal Polycystins 1 and 2 mediate mechanosensation in the pri- degeneration combined with obesity, diabetes mellitus and mary cilium of kidney cells. Nat Genet 33: 129–137, 2003 neurogenous deafness: A specific syndrome (not hitherto 104. Simons M, Gloy J, Ganner A, Bullerkotte A, Bashkurov M, described) distinct from the Laurence-Moon-Biedl syn- Kronig C, Schermer B, Benzing T, Cabello OA, Jenny A, drome. A clinical endocrinological and genetic examina- Mlodzik M, Polok B, Driever W, Obara T, Walz G: Inversin, tion based on a large pedigree. Acta Psychiatr Neurol Scand the gene product mutated in nephronophthisis type II, 34[Suppl 129]: 1–35, 1959 functions as a molecular switch between Wnt signaling 89. Goldstein JL, Fialkow PJ: The Alstrom syndrome. Report of pathways. Nat Genet 37: 537–543, 2005 three cases with further delineation of the clinical, patho- 105. Okada Y, Nonaka S, Tanaka Y, Saijoh Y, Hamada H, Hi- 1870 Journal of the American Society of Nephrology J Am Soc Nephrol 18: 1855–1871, 2007

rokawa N: Abnormal nodal flow precedes situs inversus in General and cell-type specific mechanisms target TRPP2/ iv and inv mice. Mol Cell 4: 459–468, 1999 PKD-2 to cilia. Development 133: 3859–3870, 2006 106. Gattone VH 2nd, Wang X, Harris PC, Torres VE: Inhibition of 120. Jauregui AR, Barr MM: Functional characterization of the renal cystic disease development and progression by a vaso- C. elegans nephrocystins NPHP-1 and NPHP-4 and their pressin V2 receptor antagonist. Nat Med 9: 1323–1326, 2003 role in cilia and male sensory behaviors. Exp Cell Res 305: 107. Schuermann MJ, Otto E, Becker A, Saar K, Ruschendorf F, 333–342, 2005 Polak BC, Ala-Mello S, Hoefele J, Wiedensohler A, Haller M, 121. Winkelbauer ME, Schafer JC, Haycraft CJ, Swoboda P, Omran H, Nurnberg P, Hildebrandt F: Mapping of gene loci Yoder BK: The C. elegans homologs of nephrocystin-1 and for nephronophthisis type 4 and Senior-Loken syndrome, to nephrocystin-4 are cilia transition zone proteins involved chromosome 1p36. Am J Hum Genet 70: 1240–1246, 2002 in chemosensory perception. J Cell Sci 118: 5575–5587, 2005 108. Wolf MT, Lee J, Panther F, Otto EA, Guan KL, Hildebrandt 122. Blacque OE, Reardon MJ, Li C, McCarthy J, Mahjoub MR, Ans- F: Expression and phenotype analysis of the nephrocys- ley SJ, Badano JL, Mah AK, Beales PL, Davidson WS, Johnsen tin-1 and nephrocystin-4 homologs in Caenorhabditis el- RC, Audeh M, Plasterk RH, Baillie DL, Katsanis N, Quarmby egans. J Am Soc Nephrol 16: 676–687, 2005 LM, Wicks SR, Leroux MR: Loss of C. elegans BBS-7 and BBS-8 109. Chang B, Khanna H, Hawes N, Jimeno D, He S, Lillo C, protein function results in cilia defects and compromised in- Parapuram SK, Cheng H, Scott A, Hurd RE, Sayer JA, Otto traflagellar transport. Genes Dev 18: 1630–1642, 2004 EA, Attanasio M, O’Toole JF, Jin G, Shou C, Hildebrandt F, 123. Li JB, Gerdes JM, Haycraft CJ, Fan Y, Teslovich TM, May- Williams DS, Heckenlively JR, Swaroop A: In-frame dele- Simera H, Li H, Blacque OE, Li L, Leitch CC, Lewis RA, tion in a novel centrosomal/ciliary protein CEP290/ Green JS, Parfrey PS, Leroux MR, Davidson WS, Beales PL, NPHP6 perturbs its interaction with RPGR and results in Guay-Woodford LM, Yoder BK, Stormo GD, Katsanis N, early-onset retinal degeneration in the rd16 mouse. Hum Dutcher SK: Comparative genomics identifies a flagellar Mol Genet 15: 1847–1857, 2006 and basal body proteome that includes the BBS5 human 110. den Hollander AI, Koenekoop RK, Yzer S, Lopez I, Arends disease gene. Cell 117: 541–552, 2004 ML, Voesenek KE, Zonneveld MN, Strom TM, Meitinger T, 124. Fan Y, Esmail MA, Ansley SJ, Blacque OE, Boroevich K, Brunner HG, Hoyng CB, van den Born LI, Rohrschneider Ross AJ, Moore SJ, Badano JL, May-Simera H, Compton DS, Green JS, Lewis RA, van Haelst MM, Parfrey PS, Baillie K, Cremers FP: Mutations in the CEP290 (NPHP6) gene are DL, Beales PL, Katsanis N, Davidson WS, Leroux MR: a frequent cause of Leber congenital amaurosis. Am J Hum Mutations in a member of the Ras superfamily of small Genet 79: 556–561, 2006 GTP-binding proteins causes Bardet-Biedl syndrome. Nat 110a. Bisgrove BW, Yost HJ: The roles of cilia in developmental Genet 36: 989–993, 2004 disorders and diseases. Development 133: 4131–4143, 2006 125. Chiang AP, Nishimura D, Searby C, Elbedour K, Carmi R, 111. Fliegauf M, Horvath J, von Schnakenburg C, Olbrich H, Ferguson AL, Secrist J, Braun T, Casavant T, Stone EM, Muller D, Thumfart J, Schermer B, Pazour GJ, Neumann Sheffield VC: Comparative genomic analysis identifies an HP, Zentgraf H, Benzing T, Omran H: Nephrocystin spe- ADP-ribosylation factor-like gene as the cause of Bardet- cifically localizes to the transition zone of renal and respi- Biedl syndrome (BBS3). Am J Hum Genet 75: 475–484, 2004 ratory cilia and photoreceptor connecting cilia. JAmSoc 126. Mykytyn K, Sheffield VC: Establishing a connection be- Nephrol 17: 2424–2433, 2006 tween cilia and Bardet-Biedl Syndrome. Trends Mol Med 10: 112. Kozminski KG, Johnson KA, Forscher P, Rosenbaum JL: A 106–109, 2004 motility in the eukaryotic unrelated to flagellar 127. Efimenko E, Bubb K, Mak HY, Holzman T, Leroux MR, beating. Proc Natl Acad Sci U S A 90: 5519–5523, 1993 Ruvkun G, Thomas JH, Swoboda P: Analysis of xbx genes 113. Lin F, Hiesberger T, Cordes K, Sinclair AM, Goldstein LS, in C. elegans. Development 132: 1923–1934, 2005 Somlo S, Igarashi P: Kidney-specific inactivation of the 128. Schermer B, Hopker K, Omran H, Ghenoiu C, Fliegauf M, KIF3A subunit of kinesin-II inhibits renal ciliogenesis and Fekete A, Horvath J, Kottgen M, Hackl M, Zschiedrich S, produces polycystic kidney disease. Proc Natl Acad Sci Huber TB, Kramer-Zucker A, Zentgraf H, Blaukat A, Walz USA100: 5286–5291, 2003 G, Benzing T: Phosphorylation by casein kinase 2 induces 114. Pazour GJ, Dickert BL, Witman GB: The DHC1b (DHC2) PACS-1 binding of nephrocystin and targeting to cilia. isoform of cytoplasmic dynein is required for flagellar EMBO J 24: 4415–4424, 2005 assembly. J Cell Biol 144: 473–481, 1999 129. Pazour GJ, Rosenbaum JL: Intraflagellar transport and cil- 115. Benzing T, Walz G: Cilium-generated signaling: A cellular ia-dependent diseases. Trends Cell Biol 12: 551–555, 2002 GPS? Curr Opin Nephrol Hypertens 15: 245–249, 2006 130. Hirokawa N, Takemura R: Biochemical and molecular 116. Ansley SJ, Badano JL, Blacque OE, Hill J, Hoskins BE, Leitch characterization of diseases linked to motor proteins. CC, Kim JC, Ross AJ, Eichers ER, Teslovich TM, Mah AK, Trends Biochem Sci 28: 558–565, 2003 Johnsen RC, Cavender JC, Lewis RA, Leroux MR, Beales PL, 131. Tanaka T, Serneo FF, Higgins C, Gambello MJ, Wynshaw- Katsanis N: Basal body dysfunction is a likely cause of pleio- Boris A, Gleeson JG: Lis1 and doublecortin function with tropic Bardet-Biedl syndrome. Nature 425: 628–633, 2003 dynein to mediate coupling of the nucleus to the centro- 117. Badano JL, Teslovich TM, Katsanis N: The centrosome in some in neuronal migration. J Cell Biol 165: 709–721, 2004 human genetic disease. Nat Rev Genet 6: 194–205, 2005 132. Fath MA, Mullins RF, Searby C, Nishimura DY, Wei J, 118. Barr MM, DeModena J, Braun D, Nguyen CQ, Hall DH, Rahmouni K, Davis RE, Tayeh MK, Andrews M, Yang B, Sternberg PW: The Caenorhabditis elegans autosomal dominant Sigmund CD, Stone EM, Sheffield VC: Mkks-null mice polycystic kidney disease gene homologs lov-1 and pkd-2 act have a phenotype resembling Bardet-Biedl Syndrome. in the same pathway. Curr Biol 11: 1341–1346, 2001 Hum Mol Genet 14: 1109–1118, 2005 119. Bae YK, Qin H, Knobel KM, Hu J, Rosenbaum JL, Barr MM: 133. Nurnberger J, Bacallao RL, Phillips CL: Inversin forms a J Am Soc Nephrol 18: 1855–1871, 2007 Nephronophthisis-Associated Ciliopathies 1871

complex with catenins and N-cadherin in polarized epithe- regulation of the cell cycle via direct activation of the lial cells. Mol Biol Cell 13: 3096–3106, 2002 JAK-STAT signaling pathway in a process requiring PKD2. 134. Praetorius HA, Spring KR: Bending the MDCK cell pri- Cell 109: 157–168, 2002 mary cilium increases intracellular calcium. J Membr Biol 146. Bukanov NO, Smith LA, Klinger KW, Ledbetter SR, 184: 71–79, 2001 Ibraghimov-Beskrovnaya O: Long-lasting arrest of murine 135. Praetorius HA, Praetorius J, Nielsen S, Frokiaer J, Spring polycystic kidney disease with CDK inhibitor roscovitine. KR: Beta1-integrins in the primary cilium of MDCK cells Nature 444: 949–952, 2006 potentiate fibronectin-induced Ca2ϩ signaling. Am J 147. Torres VE, Wang X, Qian Q, Somlo S, Harris PC, Gattone Physiol Renal Physiol 287: F969–F978, 2004 VH 2nd: Effective treatment of an orthologous model of 136. Praetorius HA, Spring KR: The renal cell primary cilium autosomal dominant polycystic kidney disease. Nat Med functions as a flow sensor. Curr Opin Nephrol Hypertens 12: 10: 363–364, 2004 517–520, 2003 148. Torres VE, Harris PC: Mechanisms of disease: Autosomal 137. Kramer-Zucker AG, Olale F, Haycraft CJ, Yoder BK, Schier dominant and recessive polycystic kidney diseases. Nat AF, Drummond IA: Cilia-driven fluid flow in the zebrafish Clin Pract Nephrol 2: 40–55; quiz 55, 2006 pronephros, brain and Kupffer’s vesicle is required for 149. Brugge JS: Casting light on focal adhesions. Nat Genet 19: normal organogenesis. Development 132: 1907–1921, 2005 309–311, 1998 138. Hildebrandt F: Identification of a gene for nephronophthi- 150. Rahilly MA, Fleming S: Abnormal integrin receptor ex- sis. Nephrol Dial Transplant 13: 1334–1336, 1998 pression in two cases of familial nephronophthisis. Histo- 139. Fischer E, Legue E, Doyen A, Nato F, Nicolas JF, Torres V, pathology 26: 345–349, 1995 Yaniv M, Pontoglio M: Defective planar cell polarity in 151. Lo SH, Yu QC, Degenstein L, Chen LB, Fuchs E: Progres- polycystic kidney disease. Nat Genet 38: 21–23, 2006 sive kidney degeneration in mice lacking tensin. J Cell Biol 140. Kim JC, Badano JL, Sibold S, Esmail MA, Hill J, Hoskins 136: 1349–1361, 1997 BE, Leitch CC, Venner K, Ansley SJ, Ross AJ, Leroux MR, 152. Togawa A, Miyoshi J, Ishizaki H, Tanaka M, Takakura A, Katsanis N, Beales PL: The Bardet-Biedl protein BBS4 tar- Nishioka H, Yoshida H, Doi T, Mizoguchi A, Matsuura N, gets cargo to the pericentriolar region and is required for Niho Y, Nishimune Y, Nishikawa S, Takai Y: Progressive microtubule anchoring and cell cycle progression. Nat impairment of kidneys and reproductive organs in mice Genet 36: 462–470, 2004 lacking Rho GDIalpha. Oncogene 18: 5373–5380, 1999 141. Dawe HR, Smith UM, Cullinane AR, Gerrelli D, Cox P, 153. Hiesberger T, Shao X, Gourley E, Reimann A, Pontoglio M, Badano JL, Blair-Reid S, Sriram N, Katsanis N, Attie-Bitach T, Afford SC, Copp AJ, Kelly DA, Gull K, Johnson CA: The Igarashi P: Role of the hepatocyte nuclear factor-1beta Meckel-Gruber Syndrome proteins MKS1 and meckelin (HNF-1beta) C-terminal domain in Pkhd1 (ARPKD) gene interact and are required for primary cilium formation. transcription and renal cystogenesis. J Biol Chem 280: Hum Mol Genet 16: 173–186, 2007 10578–10586, 2005 142. Ong AC, Harris PC: Molecular pathogenesis of ADPKD: 154. Morgan D, Eley L, Sayer J, Strachan T, Yates LM, Craig- The polycystin complex gets complex. Kidney Int 67: 1234– head AS, Goodship JA: Expression analyses and interac- 1247, 2005 tion with the anaphase promoting complex protein Apc2 143. Nishimura DY, Fath M, Mullins RF, Searby C, Andrews M, suggest a role for inversin in primary cilia and involve- Davis R, Andorf JL, Mykytyn K, Swiderski RE, Yang B, ment in the cell cycle. Hum Mol Genet 11: 3345–3350, 2002 Carmi R, Stone EM, Sheffield VC: Bbs2-null mice have 155. Keller R: Shaping the vertebrate body plan by polarized neurosensory deficits, a defect in social dominance, and embryonic cell movements. Science 298: 1950–1954, 2002 associated with mislocalization of rhodopsin. 156. Ross AJ, May-Simera H, Eichers ER, Kai M, Hill J, Jagger Proc Natl Acad Sci U S A 101: 16588–16593, 2004 DJ, Leitch CC, Chapple JP, Munro PM, Fisher S, Tan PL, 144. Mykytyn K, Mullins RF, Andrews M, Chiang AP, Swider- Phillips HM, Leroux MR, Henderson DJ, Murdoch JN, ski RE, Yang B, Braun T, Casavant T, Stone EM, Sheffield Copp AJ, Eliot MM, Lupski JR, Kemp DT, Dollfus H, Tada VC: Bardet-Biedl syndrome type 4 (BBS4)-null mice impli- M, Katsanis N, Forge A, Beales PL: Disruption of Bardet- cate Bbs4 in flagella formation but not global cilia assem- Biedl syndrome ciliary proteins perturbs planar cell polar- bly. Proc Natl Acad Sci U S A 101: 8664–8669, 2004 ity in vertebrates. Nat Genet 37: 1135–1140, 2005 145. Bhunia AK, Piontek K, Boletta A, Liu L, Qian F, Xu PN, 157. Saunier S, Salomon R, Antignac C: Nephronophthisis. Curr Germino FJ, Germino GG: PKD1 induces p21(waf1) and Opin Genet Dev 15: 324–331, 2005

Access to UpToDate on-line is available for additional clinical information at http://www.jasn.org/