nephronophthisis: disease mechanisms of a ciliopathy

13
Nephronophthisis: Disease Mechanisms of a Ciliopathy Friedhelm Hildebrandt,* †‡ Massimo Attanasio,* and Edgar Otto* Departments of *Pediatrics and Human Genetics, University of Michigan, and Howard Hughes Medical Institute, Ann Arbor, Michigan Nephronophthisis (NPHP) is an autoso- mal recessive cystic kidney disease that constitutes the most frequent genetic cause for ESRD in the first three decades of life. 1– 4 Renal failure manifests at a me- dian age of 13 yr. Initial symptoms are rel- atively mild, start at approximately age 6 yr, and consist of polyuria, polydipsia, sec- ondary enuresis, and anemia. 5 Regular fluid intake at nighttime is a characteristic feature of the patients’ history. Renal ultra- sound reveals normal kidney size, in- creased echogenicity, and corticomedul- lary cysts (Figure 1A). 6 Renal histology exhibits a characteristic triad of cortico- medullary cysts, tubular basement mem- brane disruption, and tubulointerstitial nephropathy (Figure 1B). 7,8 Disease recur- rence has never been reported in kidneys transplanted into patients with NPHP. 9 NPHP is inherited in an autosomal reces- sive mode. 4 It was first described by Smith and Graham in 1945 2 and by Fanconi et al., 3 who introduced the term “familial ju- venile nephronophthisis.” In juvenile NPHP, renal failure develops within the first three decades of life. 10 –12 In contrast, infantile NPHP, which is characterized by mutations in NPHP2/inversin, leads to re- nal failure between birth and age 3 yr. 12,13 In more than 10% of cases, NPHP is asso- ciated with extrarenal involvement, pri- marily including retinal degeneration (Se- nior-Løken syndrome), cerebellar vermis aplasia (Joubert syndrome), liver fibrosis, and cone-shaped epiphyses. These clinical features are discussed here in light of the cilia/centrosome theory of NPHP. More than 300 cases of NPHP have been pub- lished in the literature. 7 It has been re- ported from virtually all regions of the world. 14 The incidence of the disease is es- timated at nine patients per 8.3 million 15 in the United States or one in 50,000 live births in Canada. 7,16 In the North Ameri- can pediatric population with ESRD, pooled data indicate a prevalence of ap- proximately 5% of all children with renal failure. 17,18 Positional cloning has identified nine genes causing nephronophthisis when mutated. 13,19 –29 These are monogenic re- cessive genes, suggesting that mutations in each single one is sufficient by itself to cause NPHP in a patient bearing muta- tions and indicating their gene products are necessary for normal kidney func- tion. Positional cloning generated new insights into disease mechanisms of NPHP by revealing the mechanism is re- lated to the signaling pathways of pri- mary cilia, centrosomes, and planar cell polarity. 1,13,30,31 The demonstration that nephrocystin-1 and inversin/NPHP2 lo- calize to primary cilia of renal tubular cells 13 was among the first findings to support a new unifying theory of renal Published online ahead of print. Publication date available at www.jasn.org. Correspondence: Dr. Friedhelm Hildebrandt, Howard Hughes Medical Institute, Departments of Pediatrics and of Human Genetics, University of Michigan Health System, 8220C MSRB III, 1150 West Medical Center Drive, Ann Arbor, MI 48109- 5646. Phone: 734-615-7895; Fax: 734-615-1386; E-mail: [email protected] Copyright 2009 by the American Society of Nephrology ABSTRACT Nephronophthisis (NPHP), a recessive cystic kidney disease, is the most frequent genetic cause of end-stage kidney disease in children and young adults. Positional cloning of nine genes (NPHP1 through 9) and functional characterization of their encoded proteins (nephrocystins) have contributed to a unifying theory that defines cystic kidney diseases as “ciliopathies.” The theory is based on the finding that all proteins mutated in cystic kidney diseases of humans or animal models are expressed in primary cilia or centrosomes of renal epithelial cells. Primary cilia are sensory organelles that connect mechanosensory, visual, and other stimuli to mechanisms of epithelial cell polarity and cell-cycle control. Mutations in NPHP genes cause defects in signaling mechanisms that involve the noncanonical Wnt signaling pathway and the sonic hedgehog signaling pathway, resulting in defects of planar cell polarity and tissue maintenance. The ciliary theory explains the multiple organ involvement in NPHP, which includes retinal degeneration, cerebellar hypoplasia, liver fibrosis, situs inversus, and mental retardation. Positional cloning of dozens of unknown genes that cause NPHP will elucidate further signaling mechanisms involved. Nephrocystins are highly conserved in evolution, thereby allowing the use of animal models to develop future therapeutic approaches. J Am Soc Nephrol 20: 23–35, 2009. doi: 10.1681/ASN.2008050456 BRIEF REVIEW www.jasn.org J Am Soc Nephrol 20: 23–35, 2009 ISSN : 1046-6673/2001-23 23

Upload: others

Post on 12-Sep-2021

5 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Nephronophthisis: Disease Mechanisms of a Ciliopathy

Nephronophthisis: Disease Mechanisms of a Ciliopathy

Friedhelm Hildebrandt,*†‡ Massimo Attanasio,* and Edgar Otto*

Departments of *Pediatrics and †Human Genetics, University of Michigan, and ‡Howard Hughes Medical Institute,Ann Arbor, Michigan

Nephronophthisis (NPHP) is an autoso-mal recessive cystic kidney disease thatconstitutes the most frequent geneticcause for ESRD in the first three decadesof life.1–4 Renal failure manifests at a me-dian age of 13 yr. Initial symptoms are rel-atively mild, start at approximately age 6 yr,and consist of polyuria, polydipsia, sec-ondary enuresis, and anemia.5 Regularfluid intake at nighttime is a characteristicfeature of the patients’ history. Renal ultra-sound reveals normal kidney size, in-creased echogenicity, and corticomedul-lary cysts (Figure 1A).6 Renal histologyexhibits a characteristic triad of cortico-medullary cysts, tubular basement mem-brane disruption, and tubulointerstitialnephropathy (Figure 1B).7,8 Disease recur-rence has never been reported in kidneys

transplanted into patients with NPHP.9

NPHP is inherited in an autosomal reces-sive mode.4 It was first described by Smithand Graham in 19452 and by Fanconi etal.,3 who introduced the term “familial ju-venile nephronophthisis.” In juvenileNPHP, renal failure develops within thefirst three decades of life.10–12 In contrast,infantile NPHP, which is characterized bymutations in NPHP2/inversin, leads to re-nal failure between birth and age 3 yr.12,13

In more than 10% of cases, NPHP is asso-ciated with extrarenal involvement, pri-marily including retinal degeneration (Se-nior-Løken syndrome), cerebellar vermisaplasia (Joubert syndrome), liver fibrosis,and cone-shaped epiphyses. These clinicalfeatures are discussed here in light of thecilia/centrosome theory of NPHP. More

than 300 cases of NPHP have been pub-lished in the literature.7 It has been re-ported from virtually all regions of theworld.14 The incidence of the disease is es-timated at nine patients per 8.3 million15 inthe United States or one in 50,000 livebirths in Canada.7,16 In the North Ameri-can pediatric population with ESRD,pooled data indicate a prevalence of ap-proximately 5% of all children with renalfailure.17,18

Positional cloning has identified ninegenes causing nephronophthisis whenmutated.13,19 –29 These are monogenic re-cessive genes, suggesting that mutationsin each single one is sufficient by itself tocause NPHP in a patient bearing muta-tions and indicating their gene productsare necessary for normal kidney func-tion. Positional cloning generated newinsights into disease mechanisms ofNPHP by revealing the mechanism is re-lated to the signaling pathways of pri-mary cilia, centrosomes, and planar cellpolarity.1,13,30,31 The demonstration thatnephrocystin-1 and inversin/NPHP2 lo-calize to primary cilia of renal tubularcells13 was among the first findings tosupport a new unifying theory of renal

Published online ahead of print. Publication dateavailable at www.jasn.org.

Correspondence: Dr. Friedhelm Hildebrandt,Howard Hughes Medical Institute, Departments ofPediatrics and of Human Genetics, University ofMichigan Health System, 8220C MSRB III, 1150West Medical Center Drive, Ann Arbor, MI 48109-5646. Phone: 734-615-7895; Fax: 734-615-1386;E-mail: [email protected]

Copyright � 2009 by the American Society ofNephrology

ABSTRACTNephronophthisis (NPHP), a recessive cystic kidney disease, is the most frequentgenetic cause of end-stage kidney disease in children and young adults. Positionalcloning of nine genes (NPHP1 through 9) and functional characterization of theirencoded proteins (nephrocystins) have contributed to a unifying theory that definescystic kidney diseases as “ciliopathies.” The theory is based on the finding that allproteins mutated in cystic kidney diseases of humans or animal models are expressedin primary cilia or centrosomes of renal epithelial cells. Primary cilia are sensoryorganelles that connect mechanosensory, visual, and other stimuli to mechanisms ofepithelial cell polarity and cell-cycle control. Mutations in NPHP genes cause defects insignaling mechanisms that involve the noncanonical Wnt signaling pathway and thesonic hedgehog signaling pathway, resulting in defects of planar cell polarity andtissue maintenance. The ciliary theory explains the multiple organ involvement inNPHP, which includes retinal degeneration, cerebellar hypoplasia, liver fibrosis, situsinversus, and mental retardation. Positional cloning of dozens of unknown genes thatcause NPHP will elucidate further signaling mechanisms involved. Nephrocystins arehighly conserved in evolution, thereby allowing the use of animal models to developfuture therapeutic approaches.

J Am Soc Nephrol 20: 23–35, 2009. doi: 10.1681/ASN.2008050456

BRIEF REVIEW www.jasn.org

J Am Soc Nephrol 20: 23–35, 2009 ISSN : 1046-6673/2001-23 23

Page 2: Nephronophthisis: Disease Mechanisms of a Ciliopathy

cystogenesis.30 This theory states that thecystoproteins mutated32 in renal cysticdisease in humans, mice, or zebrafish areexpressed in primary cilia, basal bodies,or centrosomes.30,33 Basal bodies are thefoundations from which cilia are assem-bled (Figure 2). After mitosis and cell di-vision are completed, basal bodies derivefrom the mother centriole of the centriolepair that had organized the mitotic spin-dle during cell division. As cilia areformed from the basal body, the daughtercentriole is placed on the side of the nu-cleus opposite to the basal body, thusspecifying cell polarity. The structureand function of primary cilia and basalbodies are delineated in Figure 2.

Primary cilia are highly conservedstructures that sense a wide variety of ex-tracellular cues in a wide spectrum of ep-ithelial tissues. There is a broad range ofcues that can be received by specific cili-ary receptors, including photosensation,mechanosensation, osmosensation, andolfactory sensation. In general, thepathogenesis of ciliopathies is based onan inability of epithelial cells to sense orprocess extracellular cues.34

POSITIONAL CLONING REVEALSNEPHRONOPHTHISIS AS A“CILIOPATHY”

By positional cloning, we and othersidentified recessive mutations in ninenovel genes as causing NPHP: NPHP1,19,20

NPHP2/inversin,13 NPHP3,22 NPHP4,21,27

NPHP5,23 NPHP6/CEP290,24,35 NPHP7/GLIS2,28 NPHP8/RPGRIP1L,29,36,37 and

NPHP9/NEK8,38 defining NPHP types 1through 9, respectively. This collectionof genes has made diagnostic testing pos-sible (http://www.renalgenes.org). Ho-mozygous deletions in the NPHP1 geneaccount for approximately 21% of allNPHP cases, whereas the other genescontribute less than 3% each (Figure 3).As determined in more than 1000 fami-lies with NPHP, the causative genes are

Figure 1. Morphology of nephronophthisis. (A) Renal ultrasound demonstrates in-creased echogenicity, loss of corticomedullary differentiation, and the presence of cor-ticomedullary cysts. In contrast to PKD, kidneys are not enlarged. (B) Renal histology inNPHP shows the characteristic triad of renal tubular cysts, tubular membrane disruption,and tubulointerstitial cell infiltrates with interstitial fibrosis and periglomerular fibrosis (Bis courtesy of D. Bockenhauer, London).

Tipcomplex

Ciliaryaxoneme

Ciliarymembrane

Transitionfibers

Basalbody

Nucleus

Centrosome

Eb1

Dynein

Kinesin-2

Pc1/Pc2complex

IFT cargo

Retrogradetransport

Anterogradetransport

IFTcomplexes

AB

Cell membrane

Figure 2. Cilia structure and intraflagellar transport. The cilium is ahair-like structure that extends from the cell surface into the extra-cellular space. Virtually all vertebrate cell types can produce cilia.Cilia consist of a microtubule-based axoneme covered by a special-ized plasma membrane. The axoneme has nine peripheral microtu-bule doublets. There may be two central microtubules (9 � 2 versus9 � 0 axoneme). 9 � 2 cilia usually have dynein arms that link themicrotubule doublets and are motile, whereas most 9 � 0 cilia lackdynein arms and are nonmotile (“primary cilia”) with a few excep-tions. The ciliary axoneme is anchored in the basal body, a micro-tubule-organizing center derived from the mother centriole. Thetransition zone at the junction of the basal body acts as a filter forthe molecules that can pass into or out of the cilium. Nephrocystin-1is localized at the transition zone of epithelial cells.47 During cilio-genesis, cilia elongate from the basal body by the addition of newaxonemal subunits to the distal tip, the plus end of the microtu-bules. Axonemal and membrane components are transported in raftmacromolecular particles (complex A and B) by so-called intraflagel-lar transport (IFT) along the axonemal doublet microtubules.129

Anterograde transport toward the tip is driven by heterotrimerickinesin 2, which contains motor subunits Kif3a and Kif3b and anonmotor subunit. Mutations of Kif3a cause renal cysts and cere-bellar vermis aplasia in mice.130 Retrograde transport back to thecell body occurs via the motor protein cytoplasmic dynein 1B.131

Adapted from Bisgrove and Yost.132

BRIEF REVIEW www.jasn.org

24 Journal of the American Society of Nephrology J Am Soc Nephrol 20: 23–35, 2009

Page 3: Nephronophthisis: Disease Mechanisms of a Ciliopathy

still unknown in approximately 70% ofpatients, indicating that additional genesare involved in the pathogenesis ofNPHP (Figure 3). Evidence has beengenerated that more than one recessivegene may be mutated in individual pa-tients with NPHP, as has been proposedfor the related disorder Bardet-Biedl syn-drome (BBS).39 In this situation, muta-tions in an additional NPHP gene maymodify the clinical picture of NPHP inthe direction of a more severe extrarenalinvolvement.40,41 Here, disease mecha-nisms of NPHP are discussed in the con-text of the discovery of each of the genesencoding NPHP. We describe how re-sulting insights into the function of theirgene products, the nephrocystins, helpedrefine the cilia/centrosome theory of re-nal cystic diseases.

NPHP1 Locates to Cell Contactsand the Cilia Transition ZoneMutations in NPHP1 were identified ascausing juvenile nephronophthisis type1.19,20 NPHP1 encodes nephrocystin-1, aprotein that interacts with components ofcell–cell and cell–matrix signaling, includ-ing p130Cas,42 focal adhesion kinase 2,43

tensin, and filamin A and B.44,45 Nephro-cystin-1 is located at adherens junctionsand focal adhesions of renal epithelialcells,44,45 which are involved in cell–celland cell–basement membrane contacts,respectively (Figure 4). It also interactswith the product of other nephronophthi-sis genes such as nephrocystin-2/inver-sin,13 nephrocystin-3,22 and nephrocystin-4.21,46 More recently, it was shown thatnephrocystin-1 is targeted to the transitionzone of motile and primary cilia by the pro-tein phosphofurin acidic cluster sortingprotein 1 (PACS-1).47 This is initiated bycasein kinase 2–mediated phosphorylationof three critical serine residues within acluster of acidic amino acids in nephrocys-tin, leading to PACS-1 binding and co-lo-calization of nephrocystin with PACS-1 atthe base of cilia.48

When NPHP1 was first identified,19

we proposed a pathogenic hypothesisthat tied nephrocystin-1 in with defectsof cell– cell and cell–matrix signaling.49,50

This was based on the finding thatnephrocystin-1 contains an SH3 do-

main, localizes to adherens junctions andfocal adhesions of renal epithelial cells,and interacts with integral componentsof these structures, such as p130CAS.44,45

This “adherens junction/focal adhesionhypothesis” of NPHP pathogenesis49,50

was partially reconciled with the “cilia/centrosome” hypothesis in an integrativehypothesis by showing that nephrocys-tin-4 in polarized epithelial cells co-lo-calizes with �-catenin at cell– cell contactsites and to primary cilia, whereas, in di-viding cells, it localizes to centrosomes46

(Figure 4).

NPHP2/Inversin Implicates Cilia andPlanar Cell Polarity in CystogenesisOn the basis of positional cloning12 andcandidate gene data,51,52 we identifiedmutations in human inversin (INVS) asthe cause of infantile NPHP (type 2) withand without situs inversus.13 The renalcystic changes of infantile nephronoph-thisis (NPHP type 2) combine clinicalfeatures of NPHP and of polycystic kid-ney disease (PKD).53 We demonstratedthat nephrocystin-1 and NPHP2/inver-sin interact with �-tubulin, which con-stitutes the microtubule axoneme of pri-mary cilia, and that they localize atprimary cilia of renal tubular cells.13 Thiswas one of the first findings to support aunifying theory of renal cystogen-esis1,30,33,54 which states that cystopro-teins, which are mutated in renal cysticdisease in humans, mice, or zebrafish, are

expressed in primary cilia, basal bodies,or centrosomes.13,30 The interaction andco-localization to cilia and basal bodiesof nephrocystin-1, inversin, and �-tubu-lin provide a functional link between thepathogenesis of NPHP, the pathogenesisof PKD, primary cilia function, and left–right axis determination.13 Okada et al.55

previously demonstrated that inversin isneeded to position cilia in cells of theventral node. Inversin was then shown tolocalize to different subcellular locations,in a cell cycle– dependent manner (Fig-ure 5). Specifically, it is found at the mi-totic spindle in mitosis, at the mid-bodyin cytokinesis, and in cilia at the basalbody and centrosome in interphase (Fig-ure 5). All of these subcellular organellesare involved in regulation of planar cellpolarity or the cell cycle (Figure 4).

A major breakthrough was also madein the understanding of the pathogenesisof renal cystic diseases when Simons etal.56 demonstrated a role for inversin/NPHP2 in signaling mechanisms of pla-nar cell polarity necessary to maintainnormal tubular development and mor-phology, as outlined in Figure 6.31,56 As aconsequence of this model, when inver-sin is defective (as in NPHP type 2), ca-nonical Wnt pathway will prevail anddisrupt the apical-basolateral polarity ofrenal epithelium.31 Because planar cellpolarity signaling is important for ori-ented cell division, it seems logical thatFisher et al.57 were able to demonstrate

NPHP21%

NPHP31%

NPHP53%

NPHP61%

NPHP7/ GLIS20.1%

NPHP8/ RPGRIP1L0.5%

NPHP9/ NEK80.1%

No mutation70%

NPHP121%

NPHP42%

Figure 3. Distribution of causative mutations in the NPHP1 through NPHP9 genes in aworldwide cohort of 1079 families with NPHP. Note that whereas mutations in NPHP1account for �21% of NPHP, all other genes are in the range of �3%.

BRIEF REVIEWwww.jasn.org

J Am Soc Nephrol 20: 23–35, 2009 Mechanisms of Nephronophthisis 25

Page 4: Nephronophthisis: Disease Mechanisms of a Ciliopathy

abnormal orientation of mitotic spindlesin two different rodent models of cystickidney disease.

NPHP3: A Doorstep to Treatment?By positional cloning, we identified mu-tations in NPHP3 as responsible for ado-lescent nephronophthisis in a large Ven-ezuelan kindred.11,22 We demonstratedthat mutations in the murine orthologNphp3 cause the renal cystic mouse mu-tant pcy,22 which was demonstrated to beresponsive to treatment with a vasopres-sin receptor antagonist.58 Recently, it wasshown that complete loss of Nphp3 func-tion results in situs inversus, congenitalheart defects, and embryonic lethality inmice.59 In addition, truncating muta-tions of NPHP3 in humans causes abroad clinical spectrum of early embry-onic patterning defects that resemblesMeckel syndrome. This includes situs in-versus, polydactyly, central nervous sys-tem malformations, structural heart de-fects, preauricular fistulae, and a widerange of congenital anomalies of the kid-ney and urinary tract.59

NPHP4 Is Conserved inCaenorhabditis elegansMutations in the novel gene NPHP4were identified by homozygosity map-ping and total genome search for link-age.21,60,61 Nephrocystin-4, like inversin,localizes to primary cilia, basal bodies,centrosomes, and the cortical actin cy-toskeleton46 (Figure 4). Nephrocystin-4is conserved in Caenorhabditis elegansand expressed in ciliated head and tailneurons of the nematode.62 Uponknockdown, it exhibits a male matingphenotype, similar to the phenotypefound upon knockdown of the polycys-tin-1 and polycystin-2 orthologs.62 Lo-calization of nphp-1 and nphp-4 to someof these ciliated neurons also overlapswith localization of the cystoprotein or-thologs polycystin-1 (lov-1), polycys-tin-2 (pkd-2) and with many orthologs ofBBS proteins,62,63 similar to what hasbeen described for lov-1 and pkd-2 mu-tants.64 These data were refined for spe-cific neuronal cell types,65,66 and the ne-cessity of nphp-1 and nphp-4 formorphologic integrity of ciliated neu-

rons in C. elegans was observed.67,68

nphp-4 has an additional role in deter-mining the life span of the worm.69

Evolutionary conservation of cys-toproteins goes even further: Some cys-toproteins have been conserved overmore than 1.5 billion years of evolutionfrom the unicellular organism Chlamy-domonas reinhardtii to vertebrates. Ch.

Reinhardtii uses two motor cilia (flagella)for locomotion. Striking, nephrocystin-4and at least six proteins mutated in BBSare conserved in Ch. reinhardtii, wherethey are part of its basal body pro-teome.63,70 Defects of cystoprotein or-thologs in Ch. reinhardtii have deficientintraflagellar transport and flagellar pro-pulsion.71

1

NEPHROCYSTINS SIGNALING

NPHP1 =

2 =

3 =

4 =

5 =

6 =

7 =

NPHP2/Inversin

NPHP3

NPHP4

NPHP5

NPHP6/CEP290

NPHP7/GLIS2

PKD

Jeune

Nephronophthisis

Bardet-Biedl

MKS

Nucleus

LOCALIZATION CILIOPATHY

Receptor

Adherensjunction

Cargo

Motor protein

Transition zone

Basal body

Golgi

Mitotic spindle

Centrosome(orientation)

PC1,2

IFT80

KIF3A

NPHP1

2-4-5-6-8-9

1–5

Wnt Shh

7

2

1

1

2

PC2

PACS-1

BBS1-12

Joubert

1

BBS1-12

2-4-5-6-8-9

7

8 =

9 =

NPHP8/RPGRIP1L

NPHP9/NEK8

8

8 =

9 =

NPHP8/RPGRIP1L

NPHP9/NEK8

A B C D

Figure 4. Subcellular localization of nephrocystins to primary cilia, basal bodies, themitotic spindle, focal adhesions and adherens junctions, and functional interaction withother proteins mutated in renal “ciliopathies.” “Cystoproteins” are proteins of genesmutated in cystic kidney diseases of humans, mice, or zebrafish. Depending on cell-cyclestage, cystoproteins are localized at different subcellular organelles (shown in gray orbrown),24,133 including primary cilia, basal bodies, endoplasmic reticulum, the mitoticspindle, centrosomes, adherens junctions, or focal adhesions. Arrows in the primarycilium indicate the direction of anterograde transport along the microtubule systemmediated by kinesin-2 and retrograde transport by cytoplasmic dynein 1b. (A) Mostnephrocystins (blue) are located at cilia, the basal body, and centrosome in a cellcycle–dependent manner. NPHP1 is also at the transition zone, focal adhesions, andadherens junctions. (B) Sensory cilia perceive and process cell external signals, and“cystoproteins” are involved in signaling mechanisms downstream of cilial signal recog-nition. Downstream of cilia (pink), Wnt signaling (Figure 6) and hedgehog signaling(Figure 8) play a role in planar cell polarity, which is mediated (C) partially throughorientation of centrosomes and the mitotic spindle poles (Figure 7). (D) Cilia-dependentmechanisms of planar cell polarity seem to be central to the pathogenesis of theciliopathies, the most prominent of which are listed on the right. Wnt, Wnt signalingpathway; Shh, sonic hedgehog signaling pathway.

BRIEF REVIEW www.jasn.org

26 Journal of the American Society of Nephrology J Am Soc Nephrol 20: 23–35, 2009

Page 5: Nephronophthisis: Disease Mechanisms of a Ciliopathy

NPHP5: The Senior-LøkenSyndrome GeneWhen the novel gene NPHP5 was identi-fied as mutated in nephronophthisis type5,23 all mutations detected were trunca-tions of the encoded protein nephrocys-tin-5, and all patients had early-onsetretinitis pigmentosa (Senior-Løken syn-drome). Nephrocystin-5 contains an IQdomain, which directly interacts withcalmodulin,23 and is in a complex withthe retinitis pigmentosa GTPase regula-tor (RPGR), which when defective causesX-linked retinitis pigmentosa. Bothnephrocystin-5 and RPGR are localizedin connecting cilia of photoreceptors andin primary cilia of renal epithelial cells23

(Figure 4). That connecting cilia of pho-toreceptors are the structural equivalentsof primary cilia of renal epithelial cellsrenders an explanation for retinal in-volvement in the retinal-renal syndromeSenior-Løken syndrome.

CYSTOGENESIS: DEFECTIVEREGULATION OF PLANAR CELLPOLARITY?

NPHP6/CEP290: Centrosomes as aCentral Hub for Planar Cell PolarityRegulationWe identified by positional cloning re-cessive, truncating mutations in a novelgene NPHP6/CEP290 as the cause ofNPHP type 6 and Joubert syndrome type5.24 Its gene product nephrocystin-6/Cep29024 is part of the centrosomal pro-teome.24 Like NPHP2/inversin (Figure5) and NPHP4, NPHP6/CEP290 is ex-

pressed in centrosomes and the mitoticspindle in a cell cycle– dependent man-ner. Abrogation of NPHP6 function inzebrafish causes planar cell polarity de-fects and recapitulates the human phe-

notype of NPHP type 6, including renalcysts, retinal degeneration, and cerebel-lar defects.24 Nephrocystin-6 modulatesthe activity of ATF4/CREB2, a transcrip-tion factor implicated in cAMP-depen-

Figure 5. Inversin/NPHP2 localizes to cilia, centrosomes, and the mitotic spindle in a cell cycle–dependent manner. (A) Inversin/NPHP2is found in interphase in the cilial axoneme (data not shown), close to the centrioles of the basal body complex (arrow), and at thecentrosome (arrow head). (B through D) In metaphase (B) and anaphase (C), it is at the mitotic spindle (arrows) and in telophase (D) atthe midbody (arrow) of the separating cells and in the nucleus. This reflects a centriole-associated function of inversin/NPHP2throughout the cell cycle. Reprinted from Morgan et al.,134 with permission.

Nucleus

Basal body

Wnt

Frizzled

Dvl

Dvl Dvl

Dvl

Apc

RhoROK

GSK3β

?

?

Ca2+

?

Inv

Axin

Axin

DvlAxin

Lrp5/6

Wnt

Frizzled

Lrp5/6

β-catenin

β-catenindestruction complex

Proteasomaldegradation

Lef/Tcf

MECHANOSENSORYCILIUM AT REST

MECHANOSENSORYCILIUM BENT BY URINE FLOW

C-MycC-Myc

Dvl

Inv

Inv

Lef/Tcf

A B

FLOW

Figure 6. Inversin/NPHP2 mediates a switch from the canonical the noncanonical Wntsignaling pathway, which plays a role in planar cell polarity maintenance.56 (A) This illustrationof a renal tubular epithelial cell shows how Wnt signaling occurs primarily through �-catenin–dependent pathways in the absence of urine flow. Ligand binding by the frizzled receptorresults in inactivation of the �-catenin destruction complex through the presence of dishev-eled (Dvl), increased �-catenin levels, and upregulation of effector gene expression of thecanonical Wnt signaling pathway. (B) Stimulation of the primary cilium (e.g., by urine flow)results in increased expression of inversin (Inv), which then reduces levels of cytoplasmic Dvlby increasing its proteasomal degradation. This allows reassembly and activation of the�-catenin destruction complex, thereby switching from the canonical to the noncanonicalWnt signaling pathway. The model is consistent with the finding that overexpression of�-catenin (equivalent to canonical Wnt signaling) leads to renal cysts in a mouse model.135

Adapted from Germino.31

BRIEF REVIEWwww.jasn.org

J Am Soc Nephrol 20: 23–35, 2009 Mechanisms of Nephronophthisis 27

Page 6: Nephronophthisis: Disease Mechanisms of a Ciliopathy

dent renal cyst formation.58 Interestingly, a300–amino acid in-frame deletion ofNPHP6/CEP290 causes retinal degenera-tion only, without renal or cerebellar in-volvement in the rds16 mouse model.24

This is in accordance with the recent find-ing that a hypomorphic mutation ofNphp6/Cep290 represents the most fre-quent cause of Leber’s congenital amauro-sis.24 Mutations in NPHP6/CEP290 havebeen confirmed as causing Joubert syn-drome with and without renal involve-ment.35 Furthermore, truncating muta-tions in NPHP6 were shown to causeMeckel-Gruber syndrome.72

Correct orientation of the mitotic spin-dle and centrosomes with respect to thelongitudinal axis of the tubule is critical forproper apical-basolateral polarity (Figure7). Noncanonical Wnt signaling (see Fig-ure 6) is involved in these processes duringrenal tubular morphogenesis, when in ro-dent renal tubules still elongate postnatally.The structure that would result from dis-ruption of the longitudinal growth wouldbe a dilated tubule or cyst. Recently, evi-dence for a role of planar cell polarity inrenal cystic diseases57 was advanced bymeasuring orientation of the mitotic spin-dle through three-dimensional imaging ofrenal tubules. Comparison of the distribu-tion of the mitotic angles in wild-type ani-mals and rodent cystic kidney diseasemodels suggest that mitotic angles of tworodent models of cystic kidneys, theHNF1�-deficient mouse model and thepck rat model, were clearly different fromwild-type littermates.57

NPHP7/GLIS2: The Link toHedgehog SignalingRecently, we identified mutations in thegene NPHP7/GLIS2 in a Cree Indian kin-dred encoding the transcription factorGli-similar protein 2 as the cause ofNPHP type 728 (Figure 4). Starting at 8wk of age, Glis2 mutant mice show severerenal atrophy and fibrosis resemblinghuman nephronophthisis.28 Differentialgene expression studies on Glis2 mutantkidneys demonstrated that genes pro-moting epithelial-to-mesenchymal tran-sition and fibrosis are upregulated in theabsence of Glis2.28 Striking, there wasalso prominent apoptosis present in dis-

tal tubular segments of the kidney, whichmight provide an explanation for whyPKD kidneys are enlarged with hyper-proliferation prevailing, whereas inNPHP, kidney size is reduced. GLIS2 isrelated to the GLI transcription factor,and these findings implicate the hedge-hog pathway in the pathogenesis of cystickidney diseases (Figure 8). It is a signal-ing pathway that controls cell determina-tion and tissue patterning during embry-ogenesis. The other known role of

hedgehog is the maintenance of stem cellpools in postembryonic tissues.

NPHP8: A Clinical Spectrum fromMeckel Syndrome to JoubertSyndromeRecently, missense and truncating muta-tions in the RPGRIP1L gene were identifiedby positional cloning as the cause of a Jou-bert syndrome-like phenotype (cerebro-oculo-renal syndrome [CORS]) andMeckel syndrome (Figure 4).29,36 It was

Mitotic spindle orientationalong the longitudinal axis:

Directional cell division

Disrupted planar orientation:Non-directional cell division,

cyst development

Planarasymmetry

PKD

Figure 7. Defects of cystoproteins lead to disruption of planar cell polarity and thereby torenal cysts through to malorientation of the centrosome or mitotic spindle complex. Correctorientation of the mitotic spindle and centrosomes with respect to the longitudinal axis of thetubule is critical for proper planar cell polarity (i.e., the orientation of an epithelial cell layer inthree-dimensional space). Noncanonical Wnt signaling (see Figure 6) is involved in regulationof planar cell polarity during renal tubular morphogenesis; in rodents 2 wk post partum, thetubules still elongate. The structure that would result from disruption of this longitudinalorientation is a dilated tubule or cyst. Adapted from Germino.31

Cilium

Gli1 Gli2

GliAct

Gli3

Gli3R

PKA

PromoteGli3

processingPlasmamembrane

Ptch1

Intracellularvesicle

Smo

Cos2 ?

Sufu

Iguana

Cilium

Gli1 Gli2

GliAct

Gli3

Gli3R

PKA

Ptch1

Smo

Cos2

Sufu

Iguana

Hedgehog

Figure 8. The hedgehog signaling pathway may be involved in renal cystogenesis. Inthe hedgehog pathway of vertebrates, upon binding of the Shh ligand to the patched(Ptch1) receptor, repression of the smoothened (Smo) receptor to be inserted into thecilium membrane is relieved, and posttranslational modification of the Gli transcriptionfactors within the cilium induces both their activator (GliAct) and repressor (GliR) functions.Several studies in mice have demonstrated that ciliary proteins are needed for hedgehogsignaling. Recently, the related transcription factor Gli-similar 2 (GLIS2) was found to bemutated in NPHP type 7. Adapted from Huangfu and Anderson.136

BRIEF REVIEW www.jasn.org

28 Journal of the American Society of Nephrology J Am Soc Nephrol 20: 23–35, 2009

Page 7: Nephronophthisis: Disease Mechanisms of a Ciliopathy

shown that defects in the mouse orthologRpgrip1l (Ftm) recapitulate the cerebral,renal, and hepatic defects of CORS andMeckel syndrome. RPGRIP1L co-localizesat the basal body and centrosomes with theprotein products of both NPHP6 andNPHP4.29 RPGRIP1L missense mutationsfound in individuals with CORS diminishthe interaction between RPGRIP1L andnephrocystin-4. Missense mutations wereseen in patients with Joubert syndrome.29,36

These findings confirmed there is a contin-uum for the multiorgan phenotypic abnor-malities found in Meckel syndrome, Joubertsyndrome/CORS, and nephronophthisison the basis of distinct mutations of iden-tical genes (multiple allelism).

NPHP9: The Link From Cilia to Cell-Cycle ControlWe recently identified three differenthighly conserved amino acid changes inthe gene NEK8 (never in mitosis kinase 8)as causing NPHP type 9.73 One of the mu-tations identified is positioned in the sameRCC1 domain in which a missense muta-tion causes renal cystic disease in jckmice.74,75 Upon expression in medullarycollecting duct cells, all three mutant formsof NEK8 showed defects in ciliary and cen-trosomal localization to varying degrees,supporting the notion that mutations inNEK8 cause NPHP type 938 (Figure 4). AsNEK8 plays a major role in cell-cycle regu-lation, these findings provide a direct linkbetween a protein defective in renal cysticdisease and the role of centrosomes for cell-cycle regulation (Figure 4). In this contextit is interesting that also for polycystin-1and -2 signaling, the renal cystic phenotypehas been linked to cell growth regulation.Polycystin-1 expression activates the JAK-STAT pathway, thereby upregulatingp21(waf1) and inducing cell-cycle arrest inG0/G1.76 Cell-cycle arrest required poly-cystin-2. Involvement of polycystin-1 and-2 signaling in the JAK/STAT pathwaymight explain how mutations of eithergene can result in dysregulated growth.76

Involvement of cell-cycle regulation in re-nal cystic disease was confirmed by dem-onstration that two mouse models of PKD(jck and cpk) can be efficiently treated withthe cyclin-dependent kinase inhibitorroscovitine.77

THE CILIARY THEORY EXPLAINSEXTRARENAL INVOLVEMENT OFEYE, BRAIN, AND LIVER IN NPHP

A prominent feature of NPHP is in-volvement of multiple organs (pleiot-ropy) outside the kidney. Defects inother organs are usually of a degenera-tive or developmental nature (Figure9). Specifically, NPHP may be associ-ated with tapetoretinal degeneration(Senior-Løken syndrome78,79), cere-bellar vermis aplasia (Joubert syn-drome80,81), ocular motor apraxia typeCogan,82 mental retardation,24 liver fi-brosis,83 or cone-shaped epiphyses ofthe phalanges (Mainzer-Saldino syn-drome84). Infantile NPHP type 213 canbe associated with situs inversus,13 reti-nitis pigmentosa,85 or cardiac ven-

tricular septal defect.13 In some in-stances, there seems to be a genotype/phenotype correlation regardingpleiotropy. For instance, there is in-volvement of the retina in all knowncases with mutations of NPHP5 orNPHP6. In other instances, such asNPHP1 mutations, the molecular basisof eye involvement is unknown. Thepleiotropy of NPHP has now found apotential explanation in the ciliary hy-pothesis of cystic kidney diseases (Fig-ure 9). The extrarenal organ involve-ment in NPHP is discussed by organsystem as follows (Figure 9).

Retinal Involvement (Senior-LøkenSyndrome)The renal-retinal involvement in Senior-Løken syndrome can be explained by the

Figure 9. Ciliopathies feature a broad spectrum of organ involvement, shown herefor the nephronophthisis-related ciliopathies. There is overlap between differentsyndromes: Exclusive kidney involvement is called nephronophthisis. Associatedretinal degeneration in known as Senior-Løken syndrome. Involvement of the cere-bellum represents Joubert syndrome. In the most severe form, Meckel syndrome,brain malformations, liver fibrosis, heart defects, polydactyly, and perinatal mortalityare associated. It has recently become evident that the spectrum can vary by at leasttwo mechanisms: First, multiple allelism, in which a hypomorphic mutation may causea milder phenotype. For example, a splice site mutation of NPHP6 may cause Lebercongenital optical atrophy (LCA) only. In another example, the presence of onenontruncating mutation in NPHP8 can rescue the phenotype from Meckel syndrometo Joubert syndrome. Second, NPHP genes can modify each other. For instance,NPHP6 and AHI1 modify recessive NPHP1 mutations to express a more severephenotype.137

BRIEF REVIEWwww.jasn.org

J Am Soc Nephrol 20: 23–35, 2009 Mechanisms of Nephronophthisis 29

Page 8: Nephronophthisis: Disease Mechanisms of a Ciliopathy

fact that the primary cilium of renal epi-thelial cells is a structural equivalent ofthe connecting cilium of photoreceptorcells in the retina.86 We have shown thatnephrocystin-5 and nephrocystin-6 areexpressed in the connecting cilia of pho-toreceptors.72,87

Cerebellar Vermis Aplasia (JoubertSyndrome)In Joubert syndrome, NPHP is associ-ated with coloboma of the eye, with apla-sia/hypoplasia of the cerebellar vermiscausing ataxia, and with the inconstantsymptoms of psychomotor retardationand episodic neonatal tachypnea/dys-pnea (Figure 9).80,81,88 –90 The radio-graphic feature of Joubert syndrome onaxial magnetic resonance brain imagingis the so-called “molar tooth sign” of themidbrain-hindbrain junction.90,91 It isdue to abnormal axonal decussation(nerve tract crossing) in the corticospinaltract and the superior cerebellar pe-duncles as the basis of the motor and be-havioral abnormalities of Joubert syn-drome.25 Ocular motor apraxia typeCogan, defined as the transient inabilityof horizontal eye movements in the firstfew years of life, may also be associatedwith Joubert syndrome (JBTS). Thissymptom has been described in patientswith mutations in the NPHP182,92

(“JBTS4”) and NPHP421 genes. Threedifferent recessive genes, NPHP1,81,90,91

AHI93,94 (JBTS type 3), and NPHP6,24,35

have been found mutated in JBTS. Threeadditional loci for JBTS have been identi-fied: JBTS1 on chromosome 9q34.395 andJBTS2/CORS2 on chromosome 11p12-q13.3.96 In addition, mutations of NPHP8/RPGRIP1L can cause Joubert syndrome ifat least one mutation is not truncating.29,36

Liver FibrosisNPHP and the related disorder BBS can beassociated with periductal liver fibro-sis,83,97–99 as has been described for a pa-tient with NPHP3 mutation.22 Patients de-velop hepatomegaly and moderate portalfibrosis with mild bile duct proliferation.This pattern differs from that of classicalcongenital hepatic fibrosis, where biliarydysgenesis is prominent, and from hepaticinvolvement in autosomal recessive PKD,

Arima syndrome (cerebro-oculo-hepato-renal syndrome),100–102 and Meckel syn-drome, which appears as bile duct prolifer-ation. Bile duct involvement in these cystickidney diseases may be explained by theciliary theory because the epithelial cellslining bile ducts (cholangiocytes) possessprimary cilia.

Brain Malformations (MeckelSyndrome)Meckel syndrome (MKS) features theassociation of renal cystic dysplasiawith occipital encephalocele, polydac-tyly, and biliary digenesis (Figure 9).Two recessive genes have been identi-fied, MKS1103 and MKS3,104 and an-other gene locus, MKS2,105 has beenmapped. Recently, a Meckel-like phe-notype was described for truncatingmutations of NPHP3,59 NPHP6/CEP290,72 and NPHP8/RPGRIP1L.29,36

Within the spectrum of NPHP-associ-ated ciliopathies, MKS is the most se-vere, leading to perinatal mortality.Consequently, MKS represents the cili-opathy of the group that encompassesdefects in most organs, and involvementis most severe and of developmentalrather than degenerative nature. For in-stance, organ defects reveal cystic dyspla-sia rather than NPHP in the kidneys, mi-crophthalmia of the eyes, bile ductdysgenesis in the liver, and occipital en-cephalocele in the brain, and bones areinvolved by postaxial polydactyly.106 Thenotion that MKS is at the most pro-nounced end of the clinical spectrum issupported by the finding that the pres-ence of two truncating mutations inNPHP8/RPGRIP1L causes MKS, whereasone “mild” mutation (missense ratherthan truncating) may cause the less severephenotype of Joubert syndrome.29 In addi-tion, the presence of two truncating muta-tions in NPHP6/CEP290 may cause anMKS-like phenotype (MKS4).72

Cardiac Defects and Situs InversusIn a patient with mutation of NPHP2,we observed a ventricular septal defect asa congenital cardiac malformation.13

Thus, the role of inversin for left–rightaxis specification that had been de-scribed in mice was confirmed in hu-

mans.51,52 The patient with situs inversusalso had a cardiac ventricular septal de-fect, which may be viewed as a “hetero-taxy” (left–right orientation) phenotypecaused by the same mechanism107 thatlead to situs inversus in this patient. Weconfirmed the phenotypic combinationof cystic kidney disease, situs inversus,and cardiac septal defect on the basis ofinversin mutations is observed in hu-mans, mice, and zebrafish.13 PKD2 genemutations that cause autosomal domi-nant PKD also represent a gene that reg-ulates left–right axis determination, act-ing upstream of Nodal, Ebaf, Leftb, andPitx2.107,108

Skeletal DefectsMultiple disease variants that are associ-ated with NPHP include skeletal defects,strongly suggesting a role of primary ciliafunction in skeletal development. Theseinclude Jeune syndrome (asphyxiatingthoracic dysplasia),109 –112 Ellis van Crev-eld syndrome,113 RHYNS syndrome (ret-initis pigmentosa, hypopituitarism,NPHP, skeletal dysplasia),114 Meckel-Gruber syndrome,103,104 and Sensen-brenner syndrome (cranioectodermaldysplasia).115,116 The association ofNPHP with cone-shaped epiphyses ofthe phalanges (types 28 and 28A) isknown as Mainzer-Saldino syndromeand occurs in patients who also have ret-inal degeneration and cerebellar ataxia.84

Interestingly, mutations in the orthologof the intraflagellar transport proteinIFT80 of Ch. reinhardtii is also a cause ofJeune syndrome117 (Figure 4), which em-phasizes again the strong evolutionaryconservation of “ciliopathy genes.”

BBS and Alstrøm SyndromeBBS exhibits renal histology similar toNPHP118,119 (Figure 4). Positional clon-ing of recessive genes mutated in BBS re-veals the molecular relationship betweenNPHP and BBS may lie in coexpressionof the respective gene products in pri-mary cilia, basal bodies, or centrosomesof renal epithelial cells.30 Alstrom syn-drome exhibits some phenotypic overlapwith BBS (NPHP, retinitis pigmentosa,deafness, obesity, and diabetes withoutmental defect, polydactyly, or hypogo-

BRIEF REVIEW www.jasn.org

30 Journal of the American Society of Nephrology J Am Soc Nephrol 20: 23–35, 2009

Page 9: Nephronophthisis: Disease Mechanisms of a Ciliopathy

nadism).120 The single underlying reces-sive gene, ALMS1, encodes a novel pro-tein that is a molecular component of thecentrosome.121–124 This finding, togetherwith the finding that BBS proteins local-ize to centrosomes, confirms the role ofcentrosomal proteins in cystic kidneydiseases that are associated with diabetes,obesity, and retinitis pigmentosa.125,126

Obesity is also a part of the clinical spec-trum of the ciliopathies BBS and Alstromsyndrome. Interestingly, in the Bbs6knockout mouse model, obesity is asso-ciated with hyperphagia and decreasedactivity of the mice.127

THERAPEUTIC APPROACHES TONPHP

No effective prophylaxis or treatment isavailable for NPHP other than support-ive care once chronic renal failure devel-ops and dialysis and transplantation forterminal renal failure. Gattone et al.58

showed the renal cystic phenotype of pcymice, which is the equivalent of humanNPHP type 3, can be strongly mitigatedor even reversed by treatment with thevasopressin V2 receptor antagonistOPC31260. Similar results were ob-tained using a pkd2 mouse model.128

This effect is thought to be mediated by areduction in intracellular cAMP levels.24

An important future challenge will be thedevelopment of therapies that capitalizeon what we have learned about the biol-ogy of NPHP and other cystic diseases ofthe kidney.

ACKNOWLEDGMENTS

This research was supported by grants from

the National Institutes of Health to F.H.

(R01-DK69274, R01-DK68306, and R01-

DK064614). F.H. is the Frederick G.L.

Huetwell Professor, a Doris Duke Distin-

guished Clinical Scientist, and an Investigator

of the Howard Hughes Medical Institute.

DISCLOSURESNone.

REFERENCES

1. Hildebrandt F, Otto EA: Primary cilia: Aunifying pathogenic concept for cystic kid-ney disease? Nat Rev Genet 6: 928–940,2005

2. Smith C, Graham J: Congenital medullarycysts of the kidneys with severe refractoryanemia. Am J Dis Child 69: 369–377, 1945

3. Fanconi G, Hanhart E, von Albertini A, Uh-linger E, Dolivo G, Prader A: Familial, juve-nile nephronophthisis (idiopathic paren-chymal contracted kidney) [undeterminedlanguage]. Helv Paediatr Acta 6: 1–49,1951

4. Hildebrandt F: Juvenile nephronophthisis.In: Pediatric Nephrology, edited by BarrattTM, Avner ED, Harmon WE, Baltimore, Wil-liams & Wilkins, 1999, pp. 453–458

5. Ala-Mello S, Kivivuori SM, Ronnholm KA,Koskimies O, Siimes MA: Mechanism un-derlying early anaemia in children with fa-milial juvenile nephronophthisis. PediatrNephrol 10: 578–581, 1996

6. Blowey DL, Querfeld U, Geary D, WaradyBA, Alon U: Ultrasound findings in juvenilenephronophthisis. Pediatr Nephrol 10: 22–24, 1996

7. Waldherr R, Lennert T, Weber HP, FodischHJ, Scharer K: The nephronophthisis com-plex: A clinicopathologic study in children.Virchows Arch A Pathol Anat Histol 394:235–254, 1982

8. Zollinger HU, Mihatsch MJ, Edefonti A, Ga-boardi F, Imbasciati E, Lennert T: Nephron-ophthisis (medullary cystic disease of thekidney): A study using electron microscopy,immunofluorescence, and a review of themorphological findings. Helv Paediatr Acta35: 509–530, 1980

9. Steel BT, Lirenman DS, Battie CW:Nephronophthisis. Am J Med 68: 531–538,1980

10. Hildebrandt F, Strahm B, Nothwang HG,Gretz N, Schnieders B, Singh-Sawhney I,Kutt R, Vollmer M, Brandis M: Moleculargenetic identification of families with juve-nile nephronophthisis type 1: Rate of pro-gression to renal failure. APN Study Group.Arbeitsgemeinschaft fur Padiatrische Neph-rologie. Kidney Int 51: 261–269, 1997

11. Omran H, Fernandez C, Jung M, Haffner K,Fargier B, Villaquiran A, Waldherr R, GretzN, Brandis M, Ruschendorf F, Reis A, Hilde-brandt F: Identification of a new gene locusfor adolescent nephronophthisis, on chro-mosome 3q22 in a large Venezuelan ped-igree. Am J Hum Genet 66: 118–127, 2000

12. Haider NB, Carmi R, Shalev H, SheffieldVC, Landau D: A Bedouin kindred with in-fantile nephronophthisis demonstrateslinkage to chromosome 9 by homozygositymapping. Am J Hum Genet 63: 1404–1410, 1998

13. Otto EA, Schermer B, Obara T, O’Toole JF,Hiller KS, Mueller AM, Ruf RG, Hoefele J,

Beekmann F, Landau D, Foreman JW,Goodship JA, Strachan T, Kispert A, WolfMT, Gagnadoux MF, Nivet H, Antignac C,Walz G, Drummond IA, Benzing T, Hilde-brandt F: Mutations in INVS encoding in-versin cause nephronophthisis type 2, link-ing renal cystic disease to the function ofprimary cilia and left-right axis determina-tion. Nat Genet 34: 413–420, 2003

14. Kleinknecht C: The inheritance of nephron-ophthisis. In Inheritance of Kidney and Uri-nary Tract Diseases, Vol. 9, edited bySpitzer A, Avner ED, Boston, Kluwer Aca-demic Publishers, 1989, pp. 111–113

15. Potter DE, Holliday MA, Piel CF, FeduskaNJ, Belzer FO, Salvatierra O Jr: Treatmentof end-stage renal disease in children: A15-year experience. Kidney Int 18: 103–109, 1980

16. Pistor K, Scharer K, Olbing H, Tamminen-Mobius T: Children with chronic renal fail-ure in the Federal Republic of Germany: II.Primary renal diseases, age and intervalsfrom early renal failure to renal death. Ar-beitsgemeinschaft fur Padiatrische Neph-rologie. Clin Nephrol 23: 278–284, 1985

17. Warady BA, Hebert D, Sullivan EK, Alex-ander SR, Tejani A: Renal transplantation,chronic dialysis, and chronic renal insuffi-ciency in children and adolescents. The1995 Annual Report of the North AmericanPediatric Renal Transplant CooperativeStudy. Pediatr Nephrol 11: 49–64, 1997

18. Avner ED: Medullary cystic disease andmedullary sponge kidney. In: Primer onKidney Diseases, edited by Greenberg A,Boston, Academic Press, 1994, pp. 178–181

19. Hildebrandt F, Otto E, Rensing C, Noth-wang HG, Vollmer M, Adolphs J, HanuschH, Brandis M: A novel gene encoding anSH3 domain protein is mutated in nephron-ophthisis type 1. Nat Genet 17: 149–153,1997

20. Saunier S, Calado J, Heilig R, Silbermann F,Benessy F, Morin G, Konrad M, Broyer M,Gubler MC, Weissenbach J, Antignac C: Anovel gene that encodes a protein with aputative src homology 3 domain is a can-didate gene for familial juvenile nephron-ophthisis. Hum Mol Genet 6: 2317–2323,1997

21. Mollet G, Salomon R, Gribouval O, Silber-mann F, Bacq D, Landthaler G, Milford D,Nayir A, Rizzoni G, Antignac C, Saunier S:The gene mutated in juvenile nephronoph-thisis type 4 encodes a novel protein thatinteracts with nephrocystin. Nat Genet 32:300–305, 2002

22. Olbrich H, Fliegauf M, Hoefele J, Kispert A,Otto E, Volz A, Wolf MT, Sasmaz G, TrauerU, Reinhardt R, Sudbrak R, Antignac C,Gretz N, Walz G, Schermer B, Benzing T,Hildebrandt F, Omran H: Mutations in anovel gene, NPHP3, cause adolescentnephronophthisis, tapeto-retinal degener-

BRIEF REVIEWwww.jasn.org

J Am Soc Nephrol 20: 23–35, 2009 Mechanisms of Nephronophthisis 31

Page 10: Nephronophthisis: Disease Mechanisms of a Ciliopathy

ation and hepatic fibrosis. Nat Genet 34:455–459, 2003

23. Otto E, Loeys B, Khanna H, Hellemans J,Sudbrak R, Fan S, Muerb U, O’Toole JF,Helou J, Attanasio M, Utsch B, Sayer JA,Lillo C, Jimeno D, Coucke P, De Paepe A,Reinhardt R, Klages S, Tsuda M, KawakamiI, Kusakabe T, Omran H, Imm A, TippensM, Raymond PA, Hill J, Beales P, He S,Kispert A, Margolis B, Williams DS, Swa-roop A, Hildebrandt F: Nephrocystin-5, aciliary IQ domain protein, is mutated inSenior-Løken syndrome and interacts withRPGR and calmodulin. Nat Genet 37: 282–288, 2005

24. Sayer JA, Otto EA, O’Toole JF, NurnbergG, Kennedy MA, Becker C, Hennies HC,Helou J, Attanasio M, Fausett BV, Utsch B,Khanna H, Liu Y, Drummond I, Kawakami I,Kusakabe T, Tsuda M, Ma L, Lee H, LarsonRG, Allen SJ, Wilkinson CJ, Nigg EA, ShouC, Lillo C, Williams DS, Hoppe B, KemperMJ, Neuhaus T, Parisi MA, Glass IA, PetryM, Kispert A, Gloy J, Ganner A, Walz G,Zhu X, Goldman D, Nurnberg P, SwaroopA, Leroux MR, Hildebrandt F: The centro-somal protein nephrocystin-6 is mutated inJoubert syndrome and activates transcrip-tion factor ATF4. Nat Genet 38: 674–681,2006

25. Ferland RJ, Eyaid W, Collura RV, Tully LD,Hill RS, Al-Nouri D, Al-Rumayyan A, TopcuM, Gascon G, Bodell A, Shugart YY, RuvoloM, Walsh CA: Abnormal cerebellar devel-opment and axonal decussation due tomutations in AHI1 in Joubert syndrome.Nat Genet 36: 1008–1013, 2004

26. Dixon-Salazar T, Silhavy JL, Marsh SE,Louie CM, Scott LC, Gururaj A, Al-Gazali L,Al-Tawari AA, Kayserili H, Sztriha L, Glee-son JG: Mutations in the AHI1 gene, en-coding jouberin, cause Joubert syndromewith cortical polymicrogyria. Am J HumGenet 75: 979–987, 2004

27. Otto E, Hoefele J, Ruf R, Mueller AM, HillerKS, Wolf MT, Schuermann MJ, Becker A,Birkenhager R, Sudbrak R, Hennies HC,Nurnberg P, Hildebrandt F: A gene mu-tated in nephronophthisis and retinitis pig-mentosa encodes a novel protein, nephro-retinin, conserved in evolution. Am J HumGenet 71: 1167–1171, 2002

28. Attanasio M, Uhlenhaut NH, Sousa VH,O’Toole JF, Otto E, Anlag K, Klugmann C,Treier AC, Helou J, Sayer JA, Seelow D,Nurnberg G, Becker C, Chudley AE, Nurn-berg P, Hildebrandt F, Treier M: Loss ofGLIS2 causes nephronophthisis in humansand mice by increased apoptosis and fibro-sis. Nat Genet 39: 1018–1024, 2007

29. Delous M, Baala L, Salomon R, Laclef C,Vierkotten J, Tory K, Golzio C, Lacoste T,Besse L, Ozilou C, Moutkine I, Hellman NE,Anselme I, Silbermann F, Vesque C, Ger-hardt C, Rattenberry E, Wolf MT, GublerMC, Martinovic J, Encha-Razavi F, Bod-

daert N, Gonzales M, Macher MA, Nivet H,Champion G, Bertheleme JP, Niaudet P,McDonald F, Hildebrandt F, Johnson CA,Vekemans M, Antignac C, Ruther U,Schneider-Maunoury S, Attie-Bitach T,Saunier S: The ciliary gene RPGRIP1L is mu-tated in cerebello-oculo-renal syndrome(Joubert syndrome type B) and Meckel syn-drome. Nat Genet 39: 875–881, 2007

30. Watnick T, Germino G: From cilia to cyst.Nat Genet 34: 355–356, 2003

31. Germino GG: Linking cilia to Wnts. NatGenet 37: 455–457, 2005

32. Arts HH, Doherty D, van Beersum SE, ParisiMA, Letteboer SJ, Gorden NT, Peters TA,Marker T, Voesenek K, Kartono A, OzyurekH, Farin FM, Kroes HY, Wolfrum U, BrunnerHG, Cremers FP, Glass IA, Knoers NV, Ro-epman R: Mutations in the gene encodingthe basal body protein RPGRIP1L, anephrocystin-4 interactor, cause Joubertsyndrome. Nat Genet 39: 882–888, 2007

33. Igarashi P, Somlo S: Genetics and patho-genesis of polycystic kidney disease. J AmSoc Nephrol 13: 2384–2398, 2002

34. Benzing T, Walz G: Cilium-generated sig-naling: A cellular GPS? Curr Opin NephrolHypertens 15: 245–249, 2006

35. Valente EM, Silhavy JL, Brancati F, BarranoG, Krishnaswami SR, Castori M, LancasterMA, Boltshauser E, Boccone L, Al-Gazali L,Fazzi E, Signorini S, Louie CM, Bellacchio E,Bertini E, Dallapiccola B, Gleeson JG: Mu-tations in CEP290, which encodes a centro-somal protein, cause pleiotropic forms ofJoubert syndrome. Nat Genet 38:623–625, 2006

36. Wolf MT, Saunier S, O’Toole JF, Wanner N,Groshong T, Attanasio M, Salomon R, Stall-mach T, Sayer JA, Waldherr R, Griebel M,Oh J, Neuhaus TJ, Josefiak U, Antignac C,Otto EA, Hildebrandt F: Mutational analy-sis of the RPGRIP1L gene in patients withJoubert syndrome and nephronophthisis.Kidney Int 72: 1520–1526, 2007

37. Roepman R, Bernoud-Hubac N, Schick DE,Maugeri A, Berger W, Ropers HH, CremersFP, Ferreira PA: The retinitis pigmentosaGTPase regulator (RPGR) interacts withnovel transport-like proteins in the outersegments of rod photoreceptors. Hum MolGenet 9: 2095–2105, 2000

38. Otto EA, Trapp ML, Schultheiss UT, HelouJ, Quarmby LM, Hildebrandt F: NEK8 mu-tations affect ciliary and centrosomal local-ization and may cause nephronophthisis.J Am Soc Nephrol 19: 587–592, 2008

39. Beales PL, Badano JL, Ross AJ, Ansley SJ,Hoskins BE, Kirsten B, Mein CA, Froguel P,Scambler PJ, Lewis RA, Lupski JR, KatsanisN: Genetic interaction of BBS1 mutationswith alleles at other BBS loci can result innon-Mendelian Bardet-Biedl syndrome.Am J Hum Genet 72: 1187–1199, 2003

40. Hoefele J, Wolf MT, O’Toole JF, Otto EA,Schultheiss U, Deschenes G, Attanasio M,

Utsch B, Antignac C, Hildebrandt F: Evi-dence of oligogenic inheritance innephronophthisis. J Am Soc Nephrol 18:2789–2795, 2007

41. Badano JL, Kim JC, Hoskins BE, Lewis RA,Ansley SJ, Cutler DJ, Castellan C, BealesPL, Leroux MR, Katsanis N: Heterozygousmutations in BBS1, BBS2 and BBS6 have apotential epistatic effect on Bardet-Biedlpatients with two mutations at a secondBBS locus. Hum Mol Genet 12: 1651–1659,2003

42. Otto E, Kispert A, Schatzle, Lescher B,Rensing C, Hildebrandt F: Nephrocystin:Gene expression and sequence conserva-tion between human, mouse, and Caeno-rhabditis elegans. J Am Soc Nephrol 11:270–282, 2000

43. Benzing T, Gerke P, Hopker K, HildebrandtF, Kim E, Walz G: Nephrocystin interactswith Pyk2, p130(Cas), and tensin and trig-gers phosphorylation of Pyk2. Proc NatlAcad Sci U S A 98: 9784–9789, 2001

44. Donaldson JC, Dempsey PJ, Reddy S, Bou-ton AH, Coffey RJ, Hanks SK: Crk-associ-ated substrate p130(Cas) interacts withnephrocystin and both proteins localize tocell-cell contacts of polarized epithelialcells. Exp Cell Res 256: 168–178, 2000

45. Donaldson JC, Dise RS, Ritchie MD, HanksSK: Nephrocystin-conserved domains in-volved in targeting to epithelial cell-celljunctions, interaction with filamins, and es-tablishing cell polarity. J Biol Chem 277:29028–29035, 2002

46. Mollet G, Silbermann F, Delous M, SalomonR, Antignac C, Saunier S: Characterization ofthe nephrocystin/nephrocystin-4 complexand subcellular localization of nephrocystin-4to primary cilia and centrosomes. Hum MolGenet 14: 645–656, 2005

47. Fliegauf M, Horvath J, von SchnakenburgC, Olbrich H, Muller D, Thumfart J,Schermer B, Pazour GJ, Neumann HP,Zentgraf H, Benzing T, Omran H: Nephro-cystin specifically localizes to the transitionzone of renal and respiratory cilia and pho-toreceptor connecting cilia. J Am SocNephrol 17: 2424–2433, 2006

48. Schermer B, Hopker K, Omran H, GhenoiuC, Fliegauf M, Fekete A, Horvath J, Kott-gen M, Hackl M, Zschiedrich S, Huber TB,Kramer-Zucker A, Zentgraf H, Blaukat A,Walz G, Benzing T: Phosphorylation by ca-sein kinase 2 induces PACS-1 binding ofnephrocystin and targeting to cilia. EMBOJ 24: 4415–4424, 2005

49. Hildebrandt F: Identification of a gene fornephronophthisis. Nephrol Dial Transplant13: 1334–1336, 1998

50. Hildebrandt F, Otto E: Molecular geneticsof nephronophthisis and medullary cystickidney disease. J Am Soc Nephrol 11:1753–1761, 2000

51. Mochizuki T, Saijoh Y, Tsuchiya K, Shiray-oshi Y, Takai S, Taya C, Yonekawa H,

BRIEF REVIEW www.jasn.org

32 Journal of the American Society of Nephrology J Am Soc Nephrol 20: 23–35, 2009

Page 11: Nephronophthisis: Disease Mechanisms of a Ciliopathy

Yamada K, Nihei H, Nakatsuji N, OverbeekPA, Hamada H, Yokoyama T: Cloning ofinv, a gene that controls left/right asymme-try and kidney development. Nature 395:177–181, 1998

52. Morgan D, Turnpenny L, Goodship J, DaiW, Majumder K, Matthews L, Gardner A,Schuster G, Vien L, Harrison W, Elder FF,Penman-Splitt M, Overbeek P, Strachan T:Inversin, a novel gene in the vertebrateleft-right axis pathway, is partially deletedin the inv mouse. Nat Genet 20: 149–156,1998

53. Gagnadoux MF, Bacri JL, Broyer M, HabibR: Infantile chronic tubulo-interstitial ne-phritis with cortical microcysts: Variant ofnephronophthisis or new disease entity?Pediatr Nephrol 3: 50–55, 1989

54. Nauli SM, Alenghat FJ, Luo Y, Williams E,Vassilev P, Li X, Elia AE, Lu W, Brown EM,Quinn SJ, Ingber DE, Zhou J: Polycystins 1and 2 mediate mechanosensation in theprimary cilium of kidney cells. Nat Genet33: 129–137, 2003

55. Okada Y, Nonaka S, Tanaka Y, Saijoh Y,Hamada H, Hirokawa N: Abnormal nodalflow precedes situs inversus in iv and invmice. Mol Cell 4: 459–468, 1999

56. Simons M, Gloy J, Ganner A, Bullerkotte A,Bashkurov M, Kronig C, Schermer B, Ben-zing T, Cabello OA, Jenny A, Mlodzik M,Polok B, Driever W, Obara T, Walz G: In-versin, the gene product mutated innephronophthisis type II, functions as amolecular switch between Wnt signalingpathways. Nat Genet 37: 537–543, 2005

57. Fischer E, Legue E, Doyen A, Nato F, Nico-las JF, Torres V, Yaniv M, Pontoglio M:Defective planar cell polarity in polycystickidney disease. Nat Genet 38: 21–23, 2006

58. Gattone VH 2nd, Wang X, Harris PC, TorresVE: Inhibition of renal cystic disease devel-opment and progression by a vasopressinV2 receptor antagonist. Nat Med 9: 1323–1326, 2003

59. Bergmann C, Fliegauf M, Bruchle NO,Frank V, Olbrich H, Kirschner J, SchermerB, Schmedding I, Kispert A, Kranzlin B,Nurnberg G, Becker C, Grimm T, GirschickG, Lynch SA, Kelehan P, Senderek J, Neu-haus TJ, Stallmach T, Zentgraf H, NurnbergP, Gretz N, Lo C, Lienkamp S, Schafer T,Walz G, Benzing T, Zerres K, Omran H:Loss of nephrocystin-3 function can causeembryonic lethality, Meckel-Gruber-likesyndrome, situs inversus, and renal-hepat-ic-pancreatic dysplasia. Am J Hum Genet82: 959–970, 2008

60. Otto E, Hoefele J, Ruf R, Mueller AM, HillerKS, Wolf MT, Schuermann MJ, Becker A,Birkenhager R, Sudbrak R, Hennies HC,Nurnberg P, Hildebrandt F: A gene mu-tated in nephronophthisis and retinitis pig-mentosa encodes a novel protein, nephro-retinin, conserved in evolution. Am J HumGenet 71: 1161–1167, 2002

61. Schuermann MJ, Otto E, Becker A, Saar K,Ruschendorf F, Polak BC, Ala-Mello S,Hoefele J, Wiedensohler A, Haller M, Om-ran H, Nurnberg P, Hildebrandt F: Map-ping of gene loci for nephronophthisis type4 and Senior-Loken syndrome, to chromo-some 1p36. Am J Hum Genet 70: 1240–1246, 2002

62. Wolf MT, Lee J, Panther F, Otto EA, GuanKL, Hildebrandt F: Expression and pheno-type analysis of the nephrocystin-1 andnephrocystin-4 homologs in Caenorhabdi-tis elegans. J Am Soc Nephrol 16: 676–687, 2005

63. Badano JL, Teslovich TM, Katsanis N: Thecentrosome in human genetic disease. NatRev Genet 6: 194–205, 2005

64. Barr MM, DeModena J, Braun D, NguyenCQ, Hall DH, Sternberg PW: The Caeno-rhabditis elegans autosomal dominantpolycystic kidney disease gene homologslov-1 and pkd-2 act in the same pathway.Curr Biol 11: 1341–1346, 2001

65. Bae YK, Qin H, Knobel KM, Hu J, Rosen-baum JL, Barr MM: General and cell-typespecific mechanisms target TRPP2/PKD-2to cilia. Development 133: 3859–3870,2006

66. Jauregui AR, Barr MM: Functional charac-terization of the C. elegans nephrocystinsNPHP-1 and NPHP-4 and their role in ciliaand male sensory behaviors. Exp Cell Res305: 333–342, 2005

67. Bae YK, Lyman-Gingerich J, Barr MM, Kno-bel KM: Identification of genes involved inthe ciliary trafficking of C. elegans PKD-2.Dev Dyn 237: 2021–2029, 2008

68. Jauregui AR, Nguyen KC, Hall DH, BarrMM: The Caenorhabditis elegans nephro-cystins act as global modifiers of ciliumstructure. J Cell Biol 180: 973–988, 2008

69. Winkelbauer ME, Schafer JC, Haycraft CJ,Swoboda P, Yoder BK: The C. elegans ho-mologs of nephrocystin-1 and nephrocys-tin-4 are cilia transition zone proteins in-volved in chemosensory perception. J CellSci 118: 5575–5587, 2005

70. Mykytyn K, Sheffield VC: Establishing aconnection between cilia and Bardet-Biedlsyndrome. Trends Mol Med 10: 106–109,2004

71. Efimenko E, Bubb K, Mak HY, Holzman T,Leroux MR, Ruvkun G, Thomas JH, SwobodaP: Analysis of xbx genes in C. elegans. De-velopment 132: 1923–1934, 2005

72. Baala L, Audollent S, Martinovic J, OzilouC, Babron MC, Sivanandamoorthy S,Saunier S, Salomon R, Gonzales M, Ratten-berry E, Esculpavit C, Toutain A, MoraineC, Parent P, Marcorelles P, Dauge MC,Roume J, Le Merrer M, Meiner V, Meir K,Menez F, Beaufrere AM, Francannet C,Tantau J, Sinico M, Dumez Y, MacDonaldF, Munnich A, Lyonnet S, Gubler MC, Ge-nin E, Johnson CA, Vekemans M, Encha-Razavi F, Attie-Bitach T: Pleiotropic effects

of CEP290 (NPHP6) mutations extend toMeckel syndrome. Am J Hum Genet 81:170–179, 2007

73. Otto EA, Helou J, Allen SJ, O’Toole JF,Wise EL, Ashraf S, Attanasio M, Zhou W,Wolf MT, Hildebrandt F: Mutation analysisin nephronophthisis using a combined ap-proach of homozygosity mapping, CEL Iendonuclease cleavage, and direct se-quencing. Hum Mutat 29: 418–426, 2008

74. Sohara E, Luo Y, Zhang J, Manning DK,Beier DR, Zhou J: Nek8 regulates the ex-pression and localization of polycystin-1and polycystin-2. J Am Soc Nephrol 19:469–476, 2008

75. Liu S, Lu W, Obara T, Kuida S, Lehoczky J,Dewar K, Drummond IA, Beier DR: A de-fect in a novel Nek-family kinase causescystic kidney disease in the mouse and inzebrafish. Development 129: 5839–5846,2002

76. Bhunia AK, Piontek K, Boletta A, Liu L, QianF, Xu PN, Germino FJ, Germino GG: PKD1induces p21(waf1) and regulation of thecell cycle via direct activation of the JAK-STAT signaling pathway in a process re-quiring PKD2. Cell 109: 157–168, 2002

77. Bukanov NO, Smith LA, Klinger KW, Led-better SR, Ibraghimov-Beskrovnaya O:Long-lasting arrest of murine polycystickidney disease with CDK inhibitor roscovi-tine. Nature 444: 949–952, 2006

78. Senior B, Friedmann AI, Braudo JL: Juve-nile familial nephropathy with tapetoretinaldegeneration: a new oculorenal dystrophy.Am J Ophthalmol 52: 625–633, 1961

79. Loken AC, Hanssen O, Halvorsen S, JolsterNJ: Hereditary renal dysplasia and blind-ness. Acta Paediatr 50: 177–184, 1961

80. Saraiva JM, Baraitser M: Joubert syn-drome: A review. Am J Med Genet 43:726–731, 1992

81. Valente EM, Marsh SE, Castori M, Dixon-Salazar T, Bertini E, Al-Gazali L, Messer J,Barbot C, Woods CG, Boltshauser E, Al-Tawari AA, Salpietro CD, Kayserili H, Sz-triha L, Gribaa M, Koenig M, DallapiccolaB, Gleeson JG: Distinguishing the four ge-netic causes of Jouberts syndrome-relateddisorders. Ann Neurol 57: 513–519, 2005

82. Saunier S, Morin G, Calado J, Bennessy F,Silbermann F, Antignac C: Large deletionsof the NPH1 region in Cogan syndrome(CS) associated with familial juvenilenephronophthisis (NPH). Am J Hum Genet61: A346, 1997

83. Boichis H, Passwell J, David R, Miller H:Congenital hepatic fibrosis and nephron-ophthisis: A family study. Q J Med 42: 221–233, 1973

84. Mainzer F, Saldino RM, Ozonoff MB, Mi-nagi H: Familial nephropathy associatedwith retinitis pigmentosa, cerebellar ataxiaand skeletal abnormalities. Am J Med 49:556–562, 1970

85. O’Toole JF, Otto E, Frishberg YF: Retinitis

BRIEF REVIEWwww.jasn.org

J Am Soc Nephrol 20: 23–35, 2009 Mechanisms of Nephronophthisis 33

Page 12: Nephronophthisis: Disease Mechanisms of a Ciliopathy

pigmentosa and renal failure in a patientwith mutations in inversin [Abstract]. J AmSoc Nephrol 15: 215A, 2004

86. Pazour GJ, Dickert BL, Vucica Y, Seeley ES,Rosenbaum JL, Witman GB, Cole DG:Chlamydomonas IFT88 and its mouse ho-mologue, polycystic kidney disease genetg737, are required for assembly of ciliaand flagella. J Cell Biol 151: 709–718, 2000

87. Chang B, Khanna H, Hawes N, Jimeno D,He S, Lillo C, Parapuram SK, Cheng H,Scott A, Hurd RE, Sayer JA, Otto EA,Attanasio M, O’Toole JF, Jin G, Shou C,Hildebrandt F, Williams DS, HeckenlivelyJR, Swaroop A: In-frame deletion in a novelcentrosomal/ciliary protein CEP290/NPHP6perturbs its interaction with RPGR and re-sults in early-onset retinal degeneration inthe rd16 mouse. Hum Mol Genet 15: 1847–1857, 2006

88. Joubert M, Eisenring JJ, Robb JP, Ander-mann F: Familial agenesis of the cerebellarvermis: A syndrome of episodic hyperpnea,abnormal eye movements, ataxia, and re-tardation. Neurology 19: 813–825, 1969

89. Parisi MA, Dobyns WB: Human malforma-tions of the midbrain and hindbrain: Re-view and proposed classification scheme.Mol Genet Metab 80: 36–53, 2003

90. Gleeson JG, Keeler LC, Parisi MA, MarshSE, Chance PF, Glass IA, Graham JM Jr,Maria BL, Barkovich AJ, Dobyns WB: Molartooth sign of the midbrain-hindbrain junc-tion: Occurrence in multiple distinct syn-dromes. Am J Med Genet 125A: 125–134,discussion 117, 2004

91. Castori M, Valente EM, Donati MA, Salvi S,Fazzi E, Procopio E, Galluccio T, Emma F,Dallapiccola B, Bertini E: NPHP1 gene de-letion is a rare cause of Joubert syndromerelated disorders. J Med Genet 42: e9,2005

92. Betz R, Rensing C, Otto E, Mincheva A,Zehnder D, Lichter P, Hildebrandt F: Chil-dren with ocular motor apraxia type Cogancarry deletions in the gene (NPHP1) forjuvenile nephronophthisis. J Pediatr 136:828–831, 2000

93. Utsch B, Sayer JA, Attanasio M, Pereira RR,Eccles M, Hennies HC, Otto EA, Hilde-brandt F: Identification of the first AHI1gene mutations in nephronophthisis-asso-ciated Joubert syndrome. Pediatr Nephrol21: 32–35, 2006

94. Parisi MA, Doherty D, Eckert ML, ShawDW, Ozyurek H, Aysun S, Giray O, Al SwaidA, Al Shahwan S, Dohayan N, Bakhsh E,Indridason OS, Dobyns WB, Bennett CL,Chance PF, Glass IA: AHI1 mutations causeboth retinal dystrophy and renal cystic dis-ease in Joubert syndrome. J Med Genet43: 334–339, 2006

95. Saar K, Al-Gazali L, Sztriha L, RueschendorfF, Nur EK, Reis A, Bayoumi R: Homozygos-ity mapping in families with Joubert syn-drome identifies a locus on chromosome

9q34.3 and evidence for genetic heteroge-neity. Am J Hum Genet 65: 1666–1671,1999

96. Keeler LC, Marsh SE, Leeflang EP, WoodsCG, Sztriha L, Al-Gazali L, Gururaj A, Glee-son JG: Linkage analysis in families withJoubert syndrome plus oculo-renal involve-ment identifies the CORS2 locus on chro-mosome 11p12–q13.3. Am J Hum Genet73: 656–662, 2003

97. Delaney V, Mullaney J, Bourke E: Juvenilenephronophthisis, congenital hepatic fibro-sis and retinal hypoplasia in twins. Q J Med47: 281–290, 1978

98. Proesmans W, Van Damme B, Macken J:Nephronophthisis and tapetoretinal de-generation associated with liver fibrosis.Clin Nephrol 3: 160–164, 1975

99. Rayfield EJ, McDonald FD: Red and blondehair in renal medullary cystic disease. ArchIntern Med 130: 72–75, 1972

100. Kumada S, Hayashi M, Arima K, NakayamaH, Sugai K, Sasaki M, Kurata K, Nagata M:Renal disease in Arima syndrome isnephronophthisis as in other Joubert-re-lated cerebello-oculo-renal syndromes.Am J Med Genet A 131: 71–76, 2004

101. Satran D, Pierpont ME, Dobyns WB: Cer-ebello-oculo-renal syndromes includingArima, Senior-Loken and COACH syn-dromes: More than just variants of Joubertsyndrome. Am J Med Genet 86: 459–469,1999

102. Chance PF, Cavalier L, Satran D, PellegrinoJE, Koenig M, Dobyns WB: Clinical noso-logic and genetic aspects of Joubert andrelated syndromes. J Child Neurol 14:660–666, discussion 669–672, 1999

103. Kyttala M, Tallila J, Salonen R, Kopra O,Kohlschmidt N, Paavola-Sakki P, PeltonenL, Kestila M: MKS1, encoding a componentof the flagellar apparatus basal body pro-teome, is mutated in Meckel syndrome.Nat Genet 38: 155–157, 2006

104. Smith UM, Consugar M, Tee LJ, McKeeBM, Maina EN, Whelan S, Morgan NV,Goranson E, Gissen P, Lilliquist S, AligianisIA, Ward CJ, Pasha S, Punyashthiti R, MalikSharif S, Batman PA, Bennett CP, WoodsCG, McKeown C, Bucourt M, Miller CA,Cox P, Algazali L, Trembath RC, Torres VE,Attie-Bitach T, Kelly DA, Maher ER, Gat-tone VH 2nd, Harris PC, Johnson CA: Thetransmembrane protein meckelin (MKS3) ismutated in Meckel-Gruber syndrome andthe wpk rat. Nat Genet 38: 191–196, 2006

105. Roume J, Genin E, Cormier-Daire V, MaHW, Mehaye B, Attie T, Razavi-Encha F,Fallet-Bianco C, Buenerd A, Clerget-Dar-poux F, Munnich A, Le Merrer M: A genefor Meckel syndrome maps to chromo-some 11q13. Am J Hum Genet 63: 1095–1101, 1998

106. Khaddour R, Smith U, Baala L, Martinovic J,Clavering D, Shaffiq R, Ozilou C, CullinaneA, Kyttala M, Shalev S, Audollent S,

d’Humieres C, Kadhom N, Esculpavit C,Viot G, Boone C, Oien C, Encha-Razavi F,Batman PA, Bennett CP, Woods CG,Roume J, Lyonnet S, Genin E, Le Merrer M,Munnich A, Gubler MC, Cox P, MacdonaldF, Vekemans M, Johnson CA, Attie-BitachT: Spectrum of MKS1 and MKS3 mutationsin Meckel syndrome: A genotype-pheno-type correlation. Mutation in brief #960.[Online]. Hum Mutat 28: 523–524, 2007

107. McGrath J, Somlo S, Makova S, Tian X,Brueckner M: Two populations of nodemonocilia initiate left-right asymmetry inthe mouse. Cell 114: 61–73, 2003

108. Pennekamp P, Karcher C, Fischer A,Schweickert A, Skryabin B, Horst J, BlumM, Dworniczak B: The ion channel polycys-tin-2 is required for left-right axis determi-nation in mice. Curr Biol 12: 938–943, 2002

109. Donaldson MD, Warner AA, Trompeter RS,Haycock GB, Chantler C: Familial juvenilenephronophthisis, Jeune’s syndrome, andassociated disorders. Arch Intern Med 60:426–434, 1985

110. Jeune M, Beraud C, Carron R: Asphyxiatingthoracic dystrophy with familial characteris-tics [in French]. Arch Fr Pediatr 12: 886–891, 1955

111. Amirou M, Bourdat-Michel G, Pinel N, HuetG, Gaultier J, Cochat P: Successful renaltransplantation in Jeune syndrome type 2.Pediatr Nephrol 12: 293–294, 1998

112. Sarimurat N, Elcioglu N, Tekant GT, Elice-vik M, Yeker D: Jeune’s asphyxiating tho-racic dystrophy of the newborn. Eur J Pe-diatr Surg 8: 100–101, 1998

113. Moudgil A, Bagga A, Kamil ES, Rimoin DL,Lachman RS, Cohen AH, Jordan SC:Nephronophthisis associated with Ellis-vanCreveld syndrome. Pediatr Nephrol 12:20–22, 1998

114. Di Rocco M, Picco P, Arslanian A, RestagnoG, Perfumo F, Buoncompagni A, GattornoM, Borrone C: Retinitis pigmentosa, hy-popituitarism, nephronophthisis, and mildskeletal dysplasia (RHYNS): A new syn-drome? Am J Med Genet 73: 1–4, 1997

115. Tsimaratos M, Sarles J, Sigaudy S, Philip N:Renal and retinal involvement in theSensenbrenner syndrome. Am J MedGenet 77: 337, 1998

116. Costet C, Betis F, Berard E, Tsimaratos M,Sigaudy S, Antignac C, Gastaud P: Pig-mentosum retinis and tubulo-interstitialnephronophthisis in Sensenbrenner syn-drome: A case report [in French]. J Fr Oph-talmol 23: 158–160, 2000

117. Beales PL, Bland E, Tobin JL, Bacchelli C,Tuysuz B, Hill J, Rix S, Pearson CG, Kai M,Hartley J, Johnson C, Irving M, Elcioglu N,Winey M, Tada M, Scambler PJ: IFT80,which encodes a conserved intraflagellartransport protein, is mutated in Jeune as-phyxiating thoracic dystrophy. Nat Genet39: 727–729, 2007

118. Alton DJ, McDonald P: Urographic findings

BRIEF REVIEW www.jasn.org

34 Journal of the American Society of Nephrology J Am Soc Nephrol 20: 23–35, 2009

Page 13: Nephronophthisis: Disease Mechanisms of a Ciliopathy

in the Bardet-Biedl syndrome, formerly theLaurence-Moon-Biedl syndrome. Radiol-ogy 109: 659–663, 1973

119. Green JS, Parfrey PS, Harnett JD, Farid NR,Cramer BC, Johnson G, Heath O, McMana-mon PJ, O’Leary E, Pryse-Phillips W: Thecardinal manifestations of Bardet-Biedlsyndrome, a form of Laurence-Moon-Biedlsyndrome. N Engl J Med 321: 1002–1009,1989

120. Marshall JD, Bronson RT, Collin GB, Nord-strom AD, Maffei P, Paisey RB, Carey C,Macdermott S, Russell-Eggitt I, Shea SE,Davis J, Beck S, Shatirishvili G, Mihai CM,Hoeltzenbein M, Pozzan GB, Hopkinson I,Sicolo N, Naggert JK, Nishina PM: NewAlstrom syndrome phenotypes based onthe evaluation of 182 cases. Arch InternMed 165: 675–683, 2005

121. Andersen JS, Wilkinson CJ, Mayor T,Mortensen P, Nigg EA, Mann M: Proteomiccharacterization of the human centrosomeby protein correlation profiling. Nature426: 570–574, 2003

122. Hearn T, Spalluto C, Phillips VJ, RenforthGL, Copin N, Hanley NA, Wilson DI: Sub-cellular localization of ALMS1 supports in-volvement of centrosome and basal bodydysfunction in the pathogenesis of obesity,insulin resistance, and type 2 diabetes. Di-abetes 54: 1581–1587, 2005

123. Collin GB, Marshall JD, Ikeda A, So WV,Russell-Eggitt I, Maffei P, Beck S, BoerkoelCF, Sicolo N, Martin M, Nishina PM, Nag-gert JK: Mutations in ALMS1 cause obesity,type 2 diabetes and neurosensory degen-eration in Alstrom syndrome. Nat Genet31: 74–78, 2002

124. Hearn T, Renforth GL, Spalluto C, HanleyNA, Piper K, Brickwood S, White C, Con-nolly V, Taylor JF, Russell-Eggitt I, Bonneau

D, Walker M, Wilson DI: Mutation ofALMS1, a large gene with a tandem repeatencoding 47 amino acids, causes Alstromsyndrome. Nat Genet 31: 79–83, 2002

125. Alstrom CH, Hallgren B, Nilsson LB,Asander H: Retinal degeneration com-bined with obesity, diabetes mellitus andneurogenous deafness: A specific syn-drome (not hitherto described) distinctfrom the Laurence-Moon-Biedl syn-drome—A clinical endocrinological andgenetic examination based on a large ped-igree. Acta Psychiatr Neurol Scand34[Suppl 129]: 1–35, 1959

126. Goldstein JL, Fialkow PJ: The Alstrom syn-drome: Report of three cases with furtherdelineation of the clinical, pathophysiolog-ical, and genetic aspects of the disorder.Medicine (Baltimore) 52: 53–71, 1973

127. Fath MA, Mullins RF, Searby C, NishimuraDY, Wei J, Rahmouni K, Davis RE, Tayeh MK,Andrews M, Yang B, Sigmund CD, StoneEM, Sheffield VC: Mkks-null mice have a phe-notype resembling Bardet-Biedl Syndrome.Hum Mol Genet 14: 1109–1118, 2005

128. Torres VE, Wang X, Qian Q, Somlo S, Har-ris PC, Gattone VH 2nd: Effective treatmentof an orthologous model of autosomaldominant polycystic kidney disease. NatMed 10: 363–364, 2004

129. Kozminski KG, Johnson KA, Forscher P,Rosenbaum JL: A motility in the eukaryoticflagellum unrelated to flagellar beating. ProcNatl Acad Sci U S A 90: 5519–5523, 1993

130. Lin F, Hiesberger T, Cordes K, SinclairAM, Goldstein LS, Somlo S, Igarashi P:Kidney-specific inactivation of the KIF3Asubunit of kinesin-II inhibits renal cilio-genesis and produces polycystic kidneydisease. Proc Natl Acad Sci U S A 100:5286 –5291, 2003

131. Pazour GJ, Dickert BL, Witman GB: TheDHC1b (DHC2) isoform of cytoplasmic dy-nein is required for flagellar assembly.J Cell Biol 144: 473–481, 1999

132. Bisgrove BW, Yost HJ: The roles of cilia indevelopmental disorders and disease. De-velopment 133: 4131–4143, 2006

133. Nurnberger J, Bacallao RL, Phillips CL: In-versin forms a complex with catenins andN-cadherin in polarized epithelial cells. MolBiol Cell 13: 3096–3106, 2002

134. Morgan D, Eley L, Sayer J, Strachan T,Yates LM, Craighead AS, Goodship JA:Expression analyses and interactionwith the anaphase promoting complexprotein Apc2 suggest a role for inversin inprimary cilia and involvement in the cellcycle. Hum Mol Genet 11: 3345–3350,2002

135. Saadi-Kheddouci S, Berrebi D, RomagnoloB, Cluzeaud F, Peuchmaur M, Kahn A,Vandewalle A, Perret C: Early developmentof polycystic kidney disease in transgenicmice expressing an activated mutant of thebeta-catenin gene. Oncogene 20: 5972–5981, 2001

136. Huangfu D, Liu A, Rakeman AS, Murcia NS,Niswander L, Anderson KV: Hedgehog sig-nalling in the mouse requires intraflagellartransport proteins. Nature 426: 83–87,2003

137. Baala L, Romano S, Khaddour R, SaunierS, Smith UM, Audollent S, Ozilou C,Faivre L, Laurent N, Foliguet B, MunnichA, Lyonnet S, Salomon R, Encha-Razavi F,Gubler MC, Boddaert N, de Lonlay P,Johnson CA, Vekemans M, Antignac C,Attie-Bitach T: The Meckel-Gruber syn-drome gene, MKS3, is mutated in Jou-bert syndrome. Am J Hum Genet 80:186 –194, 2007

BRIEF REVIEWwww.jasn.org

J Am Soc Nephrol 20: 23–35, 2009 Mechanisms of Nephronophthisis 35