Next Article in Journal
Racial and Ethnic Disparities in NAFLD: Harnessing Epigenetic and Gut Microbiota Pathways for Targeted Therapeutic Approaches
Previous Article in Journal
Biological Activities of Essential Oils and Hydrolates from Different Parts of Croatian Sea Fennel (Crithmum maritimum L.)
Previous Article in Special Issue
A 30-Min Exposure on Permethrin and Deltamethrin Modifies Ion Transport Pathways in the Skin
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Senior–Loken Syndrome: Ocular Perspectives on Genetics, Pathogenesis, and Management

1
Center for Drug Evaluation, National Medical Products Administration, Beijing 100076, China
2
Xiangya School of Medicine, Central South University, Changsha 410013, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomolecules 2025, 15(5), 667; https://doi.org/10.3390/biom15050667
Submission received: 20 March 2025 / Revised: 2 May 2025 / Accepted: 3 May 2025 / Published: 5 May 2025
(This article belongs to the Special Issue Molecular Aspects of Diseases Origin and Development)

Abstract

:
Senior–Loken syndrome (SLSN) is a group of rare autosomal recessive disorders caused by dysfunction of the primary cilium, primarily affecting the kidneys (typically leading to nephronophthisis) and eyes (typically leading to retinal degeneration). Moreover, patients with SLSN may experience additional multisystemic symptoms, such as developmental delay, intellectual disability, ataxia, and nystagmus. To date, eight genes have been demonstrated to cause SLSN, all encoding for proteins involved in the structure and functions of the primary cilium. This places SLSN within an expanding category of diseases known as “ciliopathies”. Due to the genetic heterogeneity and significant phenotypic overlap with other ciliopathies, establishing a definitive diagnosis during the initial consultation remains a challenge for clinicians. Furthermore, current research on SLSN-related ciliopathies predominantly focuses on renal involvement, while the ocular manifestations remain insufficiently explored and lack a comprehensive review. Therefore, with the goal of offering practical guidance for clinical practice, this review aims to provide a comprehensive overview of the clinical features, along with an ocular perspective on the molecular mechanisms, genetic underpinnings, and advances in the treatment of SLSN.

1. Introduction

Senior–Loken syndrome (SLSN) is a rare autosomal recessive disorder characterized by retinal degeneration and nephronophthisis (NPHP), affecting approximately 1 in every 1 million individuals [1,2]. The pathogenetic basis of SLSN involves the primary cilium, a highly conserved structure in cellular development. In retinal photoreceptors, it serves as a specialized structure bridging the inner and outer segments, facilitating protein and vesicle trafficking critical for photoreceptor maintenance and visual function [3,4]. With advancements in molecular biology, eight genes—NPHP1, NPHP3, NPHP4, IQCB1, CEP290, SDCCAG8, WDR19, and TRAF3IP1—have been identified as pathogenic for SLSN, and their individual functions and overall roles in the primary cilium are gradually being understood. Furthermore, the genotype–phenotype correlation introduces further complexity to the concept of ciliopathies, driving ongoing genetic investigations. Based on clinical manifestations and molecular evidence, an increasing number of studies have concentrated on improving the prognosis of SLSN patients, particularly regarding their ocular phenotypes. Therefore, this review aims to examine the clinical characteristics and pathogenetic mechanisms of SLSN, explore the genotypic–phenotypic correlation of SLSN to improve early diagnosis, and summarize current advancements in treatment to inform future research directions.

2. Clinical Features of SLSN

2.1. Retina

The most common ocular manifestations of SLSN are Leber’s congenital amaurosis (LCA) and retinitis pigmentosa, both of which are characterized by progressive retinal degeneration that can ultimately result in complete vision loss [5]. Retinal degeneration can be divided into two clinical subgroups [6]. Early-onset SLSN typically manifests as LCA, exhibiting nystagmus, amaurotic pupils, and/or blindness in children before 2 years of age. A full-field electroretinogram often reveals markedly reduced or absent responses. The fundoscopic examination may appear normal in the early stages but progressively reveals abnormalities such as optic disc pallor, vascular attenuation, and peripheral pigmentary retinopathy over time [5,7,8]. In contrast, delayed-onset SLSN typically presents initially with night blindness and progressively advances to complete vision loss, similar to the clinical progression seen in retinitis pigmentosa. In these patients, electroretinography shows reduced amplitudes of the a- and b-waves, along with prolonged implicit times in rod- and cone-isolated responses [9,10]. The fundoscopic examination may reveal intraretinal pigmentation, often described as bone-spicule deposits, reflecting the migration of retinal pigment epithelium (RPE) cells following photoreceptor degeneration [9]. It is essential to note that the ocular phenotypes associated with SLSN exhibit considerable heterogeneity. While certain hallmark features mentioned above are commonly observed, the individual clinical presentations and examination findings largely depend on the underlying gene mutation and its specific location [5,8].

2.2. Kidney

NPHP is a chronic tubulointerstitial nephropathy with progressive renal failure, which is a leading cause of end-stage renal disease (ESRD) in children and adolescents. Patients with NPHP may present with increased urine output, nocturia, excessive thirst, weakness, and severe fatigue. NPHP can be classified into three clinical subgroups: infantile, juvenile, and adult forms, with a median age of ESRD at 1, 13, and >15 years, respectively [11]. Generally, juvenile and adult forms share similar clinicopathological characteristics. Classic ultrasound findings include normal or reduced renal size, loss of corticomedullary differentiation, and corticomedullary cysts [12]. Renal histology reveals a characteristic triad of tubular atrophy, diffuse interstitial fibrosis, and tubular basement membrane anomalies [13]. Some patients have kidney tissue biopsies showing some glomeruli with complete sclerosis, while others demonstrate compensatory hypertrophy, a manifestation of moderate chronic kidney disease [14]. Notably, infantile NPHP differs from typical NPHP, as renal ultrasound imaging may reveal kidney enlargement [15].

2.3. Others

Although SLSN is a ciliopathy characterized by retinal and renal dysplasia, it is widely pleiotropic, and clinicians should not overlook other accompanying symptoms. For example, developmental delay, hypertension, and premature ovarian failure are common in patients with SLSN, while abnormal bone density, ataxia, cone-shaped epiphyses, and congenital liver fibrosis occur occasionally [16]. In addition, other symptoms may vary across different patients. Su Ann Tay’s case report presents the first reported case of an SLSN patient with concomitant intracranial hypertension [17]. The heterogeneity of clinical symptoms may reflect the significant role of the primary cilium during individual development.
Currently, there is no cure for SLSN, but the presence of characteristic clinical complications, such as retinal degeneration and renal dysfunction, still enables clinicians to provide timely symptomatic and supportive interventions to improve patients’ quality of life.

3. Primary Cilium

3.1. General Structure and Function

The primary cilium is a microtubule-based, immotile organelle that protrudes as a single unit from the basal body, which is derived from the centrosomal mother centriole in most human cell types. Key structural components of the primary cilium consist of an axoneme, basal body, and transition zone (TZ).
The axoneme of a primary cilium is composed of nine outer doublet microtubules, which are enveloped by the ciliary membrane. The ciliary membrane is contiguous with the plasma membrane and is enriched with specific signaling receptors and ion channels [18]. This structural specialization allows the cilium to detect changes in the extracellular environment and transmit signaling information to the cell, thereby regulating a wide range of cellular, developmental, and physiological processes [19]. Anchored to the ciliary root, the basal body is a modified centriole containing a ring of triplet microtubules and specialized structures named transition fibers at its distal end [20,21,22], which serve as an interaction platform and regulate critical aspects of the ciliary biogenesis and function [23,24,25]. Located between the basal body and the axoneme, the TZ is a crucial component of the primary cilium, containing specialized gating structures such as Y-links. Together with the transition fibers of the basal body, the TZ serves as a gating and docking station for proteins entering and exiting the primary cilium [26]. In addition to gating by the TZ and basal body transition fibers, the composition and function of the cilium are also regulated by active transport mechanisms (Figure 1).
Crucially, numerous genes mutated in ciliopathies, such as Joubert syndrome (JS), Meckel–Gruber syndrome (MKS), and NPHP encode protein components of the TZ or basal body transition fibers. This highlights the physiological significance of these structures [27].

3.2. Primary Cilia in the Eye

The capacity to generate cilia is present in nearly every human cell type, but not all cell types within a specific tissue or organ will produce cilia. Moreover, cilia formed in different cells may exhibit specificity, with their ciliary protein composition varying according to cell type and physiological state. While current research mainly focuses on cilia in photoreceptor cells, there are numerous other types of ciliated cells in the vertebrate eyes, encompassing the cornea, ciliary body, iris, lens, trabecular meshwork, and the RPE [28]. Although the biological role of these primary cilia remains incompletely understood, growing evidence suggests that cilia contribute to organogenetic tissue repair and vision [29,30,31].

3.3. Primary Cilium in Retinal Photoreceptors

Photoreceptors are specialized neurons in the retina that detect light and convert it into electrical signals. They have four distinct compartments: the outer segment (OS), which contains the light-sensitive molecules called rhodopsin in rods and opsin in cones, the inner segment (IS), which houses the metabolic machinery, the nucleus, which contains the genetic material, and a short axon that extends to second-order neurons [32]. To facilitate efficient phototransduction, primary cilia in the OS are specialized into photosensitive cilia. Both cone and rod photoreceptor outer segments contain hundreds of membrane discs that surround the axoneme and form coin-like stacks [33]. The IS houses mitochondria, rough endoplasmic reticulum, the Golgi apparatus, and microtubules, all of which play crucial roles in normal cellular functions [34]. The axoneme within the OS is securely anchored to the basal body located at the apex of the IS by a specialized TZ known as the connecting cilium, which is the sole direct link between the IS and OS of photoreceptors, facilitating the exchange of visual signaling molecules between the two compartments [3]. The outer segment’s apical surface is encased by the RPE (Figure 2). Within this environment, the apical membrane discs of the OS undergo both phagocytosis by the RPE and regeneration from the base. Furthermore, the RPE plays a crucial role in recycling retinol after phototransduction. PR cells exhibit a high degree of specialization and function in coordinated synchrony within the phototransduction cascade. As a result, any alterations, whether direct or indirect, caused by dysfunctional PRs that compromise the structural or functional integrity of this system can lead to progressive retinal degeneration. These changes may arise from variations in proteins found in the OS, IS, cilia, synapses, and mitochondria [35].

4. Pathogenesis Mechanisms

4.1. The SLSN Genes

SLSN displays both clinical and genetic heterogeneity. Since Senior and Loken first described the features of SLSN with nephronophthisis and Leber congenital amaurosis in 1961 [36,37], the Online Mendelian Inheritance in Man (OMIM) database has cataloged eight types of SLSN mutations in specific genes [38]. Table 1 provides a summary of the genes implicated in SLSN, along with their chromosomal locations and the corresponding encoded proteins [39,40].

4.1.1. NPHP1

NPHP1, mapped to chromosome 3q12, encodes a 4.5-kb transcript and an 83-kDa protein, and was the first SLSN gene to be localized [65]. It encodes NPHP1, a cytoplasmic protein associated with cell–cell junctions, focal adhesions, and cilia [66]. Indeed, NPHP1 plays a crucial role in organizing epithelial cells, as demonstrated by the disruption of apicobasal polarity upon NPHP1 knockdown in Madin–Darby canine kidney cells and its interaction with BCAR1 in IMCD3 cells [67,68]. Besides its roles in adhesion and epithelial morphogenesis, the NPHP1 product primarily regulates the transport of proteins bound to the OS/primary cilium, as well as overseeing a specific subset of IFT particles and their associated cargos destined for the OS, which is required for retinal development [69]. However, the precise molecular function of the NPHP1 protein is largely unknown, and the molecular defects underlying the pathogenesis of the disease remain unclear. The primary structure of NPHP1 supports its role as an adaptor protein, featuring a Src homology 3 (SH3) domain surrounded by two E-rich domains. These two domains interact with Crk-associated substrates and appear to play roles in regulating cell division and mediating cell–cell and cell–matrix adhesion signaling [65].

4.1.2. NPHP3

The genomic region of the NPHP3 gene, extending to 6.5 kb, which encodes a 1330-amino acid protein, has been proven to be expressed in the retina and contributes to the SLSN development [70]. This gene encodes a protein (NPHP3) that contains an N-terminal coiled–coil domain, a tubulin–tyrosine ligase domain, and a tetratricopeptide repeat domain [70]. Some studies have shown that the N-terminal myristoylation of the NPHP3 mediates cilia targeting [71,72]. Furthermore, Bergmann et al. demonstrated that the NPHP3 protein is essential for proper ciliary development and function. It inhibits Disheveled-1-induced canonical Wnt-signaling activity and is involved in controlling non-canonical Wnt signaling, which regulates planar cell polarity. Additionally, it acts as a molecular switch between various Wnt-signaling pathways [46].

4.1.3. NPHP4

Mutations in the NPHP4 gene, located on chromosome 1p36 and encoding a 1250-amino acid protein called NPHP4, can also cause SLSN [73]. NPHP4 interacts with retinitis GTPase regulator (RPGR) and RPGR-interacting protein 1 (RPGRIP1), potentially establishing a connection to the development of retinal degeneration in patients with an NPHP4 mutation. Studies have shown that mutations in NPHP4 disrupt the interaction and cause NPHP or LCA [74,75]. Interestingly, as an important ciliary protein, it does not appear to be strictly required for ciliogenesis in human cells but is essential for building functional cilia and involved in the organization of the subapical actin network in multiciliated Xenopus laevis epithelial cells [68,76]. In addition to its role in the visual cycle, the NPHP4 protein plays a crucial role in signal transduction. The conserved Hippo-signaling pathway regulates organ size in mammals and has an essential role in tumor suppression and the control of cell proliferation. Moreover, it functions as a negative regulator of the Hippo pathway by associating with LATS1 (large tumor suppressor 1), modifying LATS1-dependent phosphorylation, and leading to derepression of these protooncogenic transcriptional regulators [77]. In collaboration with NPHP2, the NPHP4 protein may downregulate the canonical Wnt pathway and promote the Wnt–PCP pathway by regulating the expression and subcellular localization of disheveled proteins. In addition, it stabilizes JADE1 protein levels and promotes its translocation to the nucleus, leading to cooperative inhibition of canonical Wnt signaling [78,79].

4.1.4. IQCB1/NPHP5

IQCB1, also called NPHP5, spans 6.6 kb on human chromosome 3, consists of 15 exons, and encodes a 69 kD protein called nephrocystin-5/IQ calmodulin-binding motif-containing protein 1(IQCB1/NPHP5). The encoded protein possesses a central coiled-coil region and two calmodulin-binding IQ domains. It localizes to the primary cilia of renal epithelial cells and the connecting cilium of photoreceptor cells [2]. Especially, unlike patients with NPHP1-4 mutation who have a 10–33% chance of developing retinal degeneration, 100% of patients with IQCB1 mutation are thought to develop SLSN [80]. Moreover, in contrast to other SLSN genes, which may affect multiple organs, NPHP5 mutations only associate with the retinal–renal phenotype and are the most frequent cause of SLSN, making it the classic SLSN gene [80,81]. The severity and high prevalence of eye complications in patients with IQCB1 mutations may be attributed to the critical role of the encoded protein in ciliary function. Apart from IQ domains and CC domains, NPHP5 has a CEP290/NPHP6-binding site and Bardet–Biedl syndrome (BBS) interaction sites [81]. Through its interaction with CEP290, the IQCB1 protein is involved in ciliogenesis and functions in early-stage cilia formation [82]. Proper assembly of BBSome plays a crucial role in ciliary transport and formation in both photoreceptor cilia and non-photoreceptor primary cilia [83]. The IQCB1 protein regulates the BBSome complex integrity, specifically ensuring the presence of BBS2 and BBS5 within the complex and their ciliary targeting of selected BBSome cargos [83]. Knockdown of IQCB1 in zebrafish can ultimately cause the mislocalization of opsins and rhodopsin to the IS, as well as OS dysplasia in mice, suggesting its role in the specific transport of proteins to the OS and IS development [81,84].

4.1.5. CEP290/NPHP6

CEP290, also known as NPHP6, is situated on chromosome 12q21.32 and encodes a 290 kDa centrosomal protein (CEP290/NPHP6) [85]. This protein is characterized by 13 putative coiled-coil domains, a region exhibiting homology to SMC chromosome segregation ATPases, six KID motifs, three tropomyosin homology domains, and an ATP/GTP binding site motif A. It specifically localizes to the basal bodies of photoreceptors, facilitating the linage of the cilium and significantly contributing to its formation, stability, and transport function [5]. Due to its important role in ciliogenesis, CEP290 is a major focus among the pathogenic genes associated with SLSN. It is required for both early and late steps in cilia formation through its interaction with the IQCB1 protein [86]. Furthermore, CEP290 plays a significant role in the early stages of ciliogenesis, involving the disappearance of centriolar satellites and the transition of primary ciliary vesicles to capped ciliary vesicles where it is essential for the centrosomal recruitment of RAB8A and the targeting of centriole satellite proteins, such as PCM1, to the centrosome [87,88]. In cooperation with IQCB1, the IQCB1 protein also contributes to the assembly of BBS complexes [89].

4.1.6. SDCCAG8/SLSN7/NPHP10

SDCCAG8, also known as SLSN7 or NPHP10, produces a 3.3 kb transcript that encodes an 88 kDa protein called SDCCAG8. This protein comprises an N-terminal globular domain, a nuclear localization signal, and eight potential coiled-coil domains [90]. The carboxyl-terminal region of SDCCAG8 contains a crucial functional module essential for cilia formation, organ development, and homeostasis [91]. Previous studies have focused on its role in BBS syndrome [57,92], but its significance in SLSN should not be ignored. During ciliogenesis, it interacts with RABEP2 and mediates its centrosomal localization. Furthermore, SDCCAG8 is required for the proper activation of the Hedgehog signaling pathway, which depends on the presence of intact primary cilia [93].

4.1.7. WDR19

WDR19 contains 37 exons and encodes three different types of domains in the IFT144 protein: six WD40 domains, which are short structural motifs of 40 amino acids ending in the tryptophan–aspartic acid dipeptide, two transmembrane domains, and six tetratricopeptide repeat domains. The common function of all WD40-containing proteins is the coordination of the assembly of multi-protein complexes, while tetratricopeptide repeat domains mediate protein–protein interactions within these complexes [94]. The IFT144 protein is a component of the IFT-A complex, which plays a crucial role in the retrograde transport of cargo within the cilium [60]. This protein is primarily expressed in the retina, particularly in photoreceptors, but also in various other tissues. In this context, WDR19 acts as a component of the IFT-A complex, which is essential for retrograde ciliary transport and the entry of G protein-coupled receptors into the cilium. This involvement in retrograde transport is critical for maintaining the cilia function and assembly [95].

4.1.8. TRAF3IP1

The TRAF3IP1 gene encodes the homolog of the intraflagellar transport protein IFT54, a subunit of the IFT-B complex that facilitates anterograde transport within cilia [62]. This 625-amino acid protein contains a KKE motif and a C-terminal coiled-coil domain [96]. Mutations in the N-terminal region of TRAF3IP1 impair its ability to bind to MAP4 [62]. Notably, MAP4 is also localized within primary cilia, where it functions as a negative regulator of ciliary length, counteracting the activity of septins [97].

4.2. The SLSN Interactome

The SLSN interactome encompasses several ciliary protein modules that converge at TZ, a critical gatekeeper for ciliary compartmentalization and signaling. Among these, the NPHP1-4-8 and NPHP5-6 complexes are two canonical modules essential for TZ integrity and are widely expressed in cilia-dependent tissues, such as retinal photoreceptors, kidney tubular epithelial cells, and cerebral cortical neurons. Dysfunction of these modules results in a spectrum of ciliopathies, including SLSN. Elucidating their molecular mechanisms will enhance our understanding of the retinal pathogenesis of SLSN and provide a basis for the development of targeted therapies.

4.2.1. NPHP1-4-8 Complex

Functioning as a classical SLSN-related module, the NPHP1, NPHP4, and NPHP8 protein share many common characteristics:
  • Containing multiple C2 domains in their core regions. These C2 domains play a central role in the membrane association at the ciliary transitional zone and serve as the protein–protein interaction modules connecting different transition zone proteins [98,99].
  • Co-dependent localization. For example, the loss of NPHP-4 leads to the delocalization of NPHP-1 [100], and a similar situation occurs between NPHP8 and NPHP1 [101,102], as well as between NPHP4 and NPHP8 [51].
  • Elegant connections with microtubules. NPHP1, NPHP4, and NPHP8 interact with β-, α-, and γ-tubulin, respectively, which happen to be the three main components of microtubules [103,104].
These characteristics enable the NPHP1-4-8 complex to locate proximally to the microtubule sides in the TZ [27], where it participates in both its composition and function. Since the connecting cilium (CC) in photoreceptors is the equivalent of the TZ, which is the only direct link between the ISs and Oss [105], alterations in NPHP1-4-8 complex may be a core mechanism underlying the ocular phenotypes of SLSN. Structurally, defects in the NPHP1-4-8 complex can damage the Y-link structure at the TZ [41], thus impairing its gating function [106]. Moreover, the NPHP1-4-8 complex seems to regulate the cargo transport in the CC, since the defects in each protein of the complex can cause the sluggish movement of the IFT [27,41,69,107], which may indicate that the complex mediates the interaction between TZ and IFT particles [41,69,108]. The RPGR provides evidence for this explanation. It extensively interacts with the NPHP1-4-8 complex and acts as a guanine nucleotide exchange factor in modulating the rhodopsin trafficking in photoreceptors [109,110,111]. The defects of NPHP1-4-8 can impair its interactions with RPGR, leading to the mislocalization of rhodopsin [75,112] and ultimately resulting in the dysfunction of photoreceptors, which is the underlying cause of ocular phenotypes in SLSN.
Notably, NPHP8 seems to act as a central scaffold at the TZ, functionally linking the NPHP and other modules [101,102,106]. Hence, the mutations in NPHP8 tend to cause more severe clinical phenotypes, making it a secondary pathogenic gene in SLSN. Rather, the mutations in NPHP1 and NPHP4 cause only mild defects in ciliogenesis and lead to less severe clinical phenotypes [27,41]. This theory is supported by a large-sample clinical study [113].
Moreover, NPHP1, NPHP4, and NPHP8 have also been found to accumulate at cell–cell contacts and participate in the process of cell polarity, particularly during the development of polarized epithelial monolayers [67,68], which may represent an additional mechanism independent of ciliopathies. However, most research only focuses on the NPHP1-4 complex, demonstrating its ability to regulate epithelial morphogenesis by interacting with polarity proteins such as the PALS1/PATJ complex and p130Cas [67,114]. Fortunately, recent studies have shown that NPHP8 is required for stabilizing epidermal adhesion and participating in the cell polarity process [115,116]. This may indicate the combination of NPHP1-4-8 complex outside the primary cilium.

4.2.2. NPHP5-6 Complex

Functional studies have demonstrated that NPHP5 interacts strongly with NPHP6, binding to its N-terminal region via its C-terminal domain [82,117]. In both mammalian cells and IMCD3 cells, they have been proven to be located in centrosomes. Additionally, studies have concluded that NPHP6 binds NPHP5 and facilitates its localization to centrosomes [68]. However, subsequent research using a large panel of mutants revealed that the NPHP6-binding domain in NPHP5 is distinct from its localization domain [82]. These findings show that while some mutants disrupting the NPHP6-binding domain impair cilia formation, they do not directly contribute to mislocalization [82]. While earlier studies proposed that NPHP5 forms a complex with NPHP6 and calmodulin, recent research demonstrates that a drug named Eupatilin can disrupt the CaM–NPHP5 interaction, promoting NPHP5 centrosomal localization in NPHP6-null cells [118]. This suggests that while NPHP6 influences NPHP5 localization, the mechanism is indirect.
NPHP5 and 6 form a complex to play a role in ciliogenesis and protein trafficking [82,89]. For example, the complex can regulate BBSome integrity and ciliary trafficking. Both of them can independently bind to BBSome, and the deletion of NPHP5 leads to a loss of BBS2 and BBS5, while the deletion of NPHP6 causes the dissociation of BBS8 from BBSome [89,119]. Moreover, the complex functions in ciliogenesis by copurifying with ciliogenesis-required proteins such as Sec3, Sec8, and Sec10 [120].
Findings from recent pharmacological rescue research indicate that NPHP5 and NPHP6 coordinately influence a common pathway converging on ciliogenesis and protein trafficking. Drugs such as cytochalasin D, which rescue the NPHP5-deletion phenotype, exhibit a similar effect on the NPHP6-deficient phenotype [82]. However, there is recent research showing that Eupatilin can only have a rescuing effect by restoring NPHP5 levels in NPHP6-null cells [89,118]. BBSome integrity can be rescued by NPHP5 overexpression in NPHP6-null cells [121]. Moreover, the ciliary gating can be controlled by Rpgrip1l via maintaining appropriate levels of CEP290 at the TZ [122]. As a result, there is still a significant difference between NPHP5 and NPHP6, which is unclear. Figuring out the proteins “downstream” and their binding mechanisms with the complex of NPHP5 and NPHP6 may be a thoughtful way for dissecting their biological roles and functions [82].

5. Genetic Basis

5.1. Shared Phenotypes Among Ciliopathies

Although the clinical phenotypes of SLSN are mainly concentrated in the eyes and kidneys, it is clinically heterogeneous with variable multiorgan involvement, which shares extensive overlap with other ciliopathies, such as Joubert syndrome, Meckel syndrome, Bardet–Biedl syndrome, oral–facial–digital syndrome, Alström Syndrome, and Usher syndrome. Additional shared phenotypes include CNS (intellectual disability, developmental delay, ataxia), skeletal(polydactyly), ear (hearing loss), liver (liver fibrosis), endocrine (diabetes, hyperinsulinemia), and obesity [6,35,123,124,125,126,127] (Figure 3).
The remarkable phenotypic overlap among these ciliopathies is rooted in the shared molecular biology foundation. Firstly, the primary cilium exists in the development process of most human cells, which is highly conserved and essential for regulating signaling pathways during development and adult homeostasis, so the defects of the primary cilium can cause multiple organic dysfunctions [148,149]. Additionally, ciliopathy proteins function as complexes and interact modularly in the biogenesis of the primary cilium [6,150,151,152], so mutations in genes encoding SLSN proteins can disrupt their interaction with other ciliopathy protein modules, eventually causing similar multi-organ phenotypes in analogy with other ciliopathies.
Recently, some research has proposed a new perspective that the phenotypic spectrum in these ciliopathies is more of a continuum rather than distinct syndromes, emphasizing the importance of considering the broader context, instead of sticking to a specific syndromic diagnosis based on the presence of certain phenotypes [133,153].

5.2. Genotype–Phenotype Correlations

In some ciliopathies caused by mutations in certain genes, significant gene–phenotype correlations have been observed. For example, mutations in CEP290 are always associated with retinal dystrophy, while mutations in WDR19 are often linked to skeletal anomalies [58,154,155,156,157]. However, considering its extensive phenotypic overlap with other ciliopathies and remarkable clinical heterogeneity, it is obvious that SLSN, like most ciliopathies, extends beyond the simple one-gene-to-one-phenotype relationship. To explain these complex correlations, different genetic mechanisms have been proposed, some of which have been identified in SLSN.

5.2.1. Locus Heterogeneity

Mutations in different genes can cause similar phenotypes, and this potential is amplified in ciliopathies due to the modular expression of ciliopathy genes and related proteins [158]. However, similar phenotypes do not necessarily indicate similar severity, and the latter is determined by specific genes [159]. For example, NPHP1, NPHP4, and NPHP8 are all found to localize to the TZ as a complex [160], and mutations in genes encoding any of them can lead to NPHP and potential extrarenal organ manifestations, but variants of NPHP1 seem to link with the lowest extrarenal performance rate and the mildest disease phenotype [113].

5.2.2. Allelic Heterogeneity

Different mutations in the same gene can cause distinct phenotypes. If two truncating mutations act during development, they may affect morphogenesis and cause a severe, early-onset phenotype, characterized by organ dysplasia or malformation; whereas two missense mutations act during tissue maintenance and repair in adult tissue, they may proceed slowly and finally cause organ degeneration [6]. This probably explains why individuals with SLSN have significant differences in the onset of the eye and kidney syndromes [64]. The correlations between hypomorphic mutations and mild phenotypes have been identified in several studies [153,161,162]. One striking example is a patient with compound heterozygous variants p.D1781E and p.T412M in CEP290, who only suffered mild RP and retained good vision at the age of 55 years [162]. However, the association between the nature of mutations and phenotypes is still challenged in some cases. For example, patients with two truncating mutations in CEP290 suffer from diseases ranging from mild SLSN, JS to lethal MKS [163,164,165]. This may imply that the mutation domain within the protein is of the same importance [113,166], although there may exist other mechanisms such as genetic modifiers and oligogenic inheritance.

5.2.3. Genetic Modifiers and Epistatic Effects

Genetic modifiers and epistatic effects describe the effects of one gene/allele on the phenotypic outcome of another gene/allele [167], which is another explanation for the genotype–phenotype complexity. A case in point is the allele A229T of RPGRIP1L, which has been found to be enriched in SLSN patients, can intensify the development of retinal degeneration by decreasing the stability of RPGRIP1L-RPGR complexity in the context of mutations in specific ciliopathy proteins [168]. Also, mutations in AHI1 (especially its variant p.R830W) seem to play a similar role in SLSN [169]. With the development and application of next-generation sequencing, more modifiers and epistatic effects have been reported in ciliopathies [53,170,171], contributing to a better understanding of the impact of genetic background. However, although genetic modifiers are collectively common, they may be individually rare and heterogeneous. Hence, they are unlikely to be discovered when studied in isolation [172], which provides research directions for some multicenter, large-sample cohort studies.

5.2.4. Oligogenic Inheritance

Different from genetic modifiers, oligogenic inheritance requires the co-inheritance of mutations in two non-alleles on separate genes, which together are both necessary and sufficient to cause the phenotype [159,167,173]. “True oligogenic inheritance” was confirmed in BBS [174,175], and some evidence was found in NPHP [176]. However, in most studies, it is difficult to distinguish whether a phenotype derives from genetic modifiers or oligogenic inheritance, and sometimes the borderline between them is blurred [170,177], considering the unpredictable polymorphic nature of the human genome and limited genetic analyses [172,173]. In fact, both mechanisms support the hypothesis of the mutational load, in which the expressivity and penetrance of ciliopathies in an individual are determined by their own genetic background, with potential additional variants in other ciliary genes [178]. The co-occurrence of distinct ciliopathy phenotypes within a single family provides an elegant case [176,179].
Although it seems that ciliopathies mostly obey classic Mendelian inheritance [180], the potential effects of variants in genetic backgrounds should never be underestimated [181]. In this context, NGS may be an effective method for improving diagnostic accuracy, identifying pathogenic mechanisms, and eventually providing guidance for clinical therapy.

6. Treatments

6.1. Gene Therapy

At present, the main methods of gene therapy for SLSN can be divided into gene substitution and gene editing. For gene substitution, the most widely used and efficient delivery vector is the recombinant adeno-associated virus (AAV) [182,183]. AAV-mediated NPHP5 gene augmentation has been confirmed to be capable of achieving therapeutic effects in a dog ciliopathy model of Leber congenital amaurosis, which provides a possible way for the treatment of SLSN in humans [184]. Yet, it cannot deliver genes of large sizes owing to its limited packaging capacity. Consequently, it is an alternative choice to deliver only a partial gene encoding a specific functional domain impacted by mutation(s) rather than a full-length gene. Research has also proved that a CEP290 fragment representing a part of the most important SLSN-causing genes can reconstitute CEP290 function and result in cone preservation and delayed rod death [185]. This finding suggests that the approach of delivering partial gene fragments holds implementation potential. However, genetic heterogeneity precludes gene therapy based on the delivery of the same gene to all SLSN patients. Hence, any future gene therapy would need to be personalized based on which gene is mutated in each patient [186,187,188].
Moreover, SLSN caused by mutations in CEP290 can be potentially treated by gene editing using CRISPR/Cas9, which offers a viable solution to the limited packaging capacity of AAV [189]. CRISPR/Cas9 stands as the most representative method in current gene-editing therapies. Its high-precision editing has transformed ocular gene-therapy paradigms, allowing for the development of disease models and targeted correction of pathogenic mutations to drive therapeutic progress [187]. EDIT-101 is a CRISPR/Cas9-based gene-editing therapy designed for CEP290-associated inherited retinal diseases. Preclinical research confirmed EDIT-101’s mechanistic feasibility in vitro, demonstrating targeted nuclease activity in human cells and retinal tissues, while in vivo models including mice and non-human primates showed effective, long-lasting gene editing and therapeutic efficacy [188]. Building on these preclinical milestones, a Phase 1–2 clinical trial subsequently evaluated EDIT-101 in 14 patients with CEP290-related IRDs, revealing favorable safety profiles and meaningful improvements in visual function [186]. Significantly, these outcomes have established a robust foundation for advancing subsequent CRISPR-based translational research in this field.
In addition, RNA-based therapies, such as RNA interference and antisense oligonucleotides, can be considered to overcome the limitations of gene delivery, which have been shown to be more economical and efficient than DNA-based therapies [190,191,192]. Specifically, for SLSN, antisense oligonucleotide technology has already been tested in a humanized mouse model of CEP290-LCA. The results demonstrated that it can reduce the expression of erroneous mutations, thereby increasing cilia length and improving their functionality [193,194].

6.2. Drug Discovery

To date, SLSN has been linked to mutations in multiple disease-causing genes, with novel pathogenic variants continuously being identified [15]. Consequently, as previously discussed, tailoring gene therapy to individual mutations remains challenging and less feasible due to genetic heterogeneity. SLSN is a condition characterized by retinitis pigmentosa and NPHP [64,134,160], making symptom-targeted therapies a promising alternative approach.
For the purpose of slowing retinal degeneration and maintaining patients’ vision, neuroprotective agents are an excellent choice, which includes neurotrophic factors, anti-apoptotic agents, and antioxidants [195]. Supplements of antioxidants such as docosahexaenoic acid and vitamin A have shown the potential in preventing photoreceptor apoptosis [196,197,198]. Moreover, the ciliary neurotrophic factor, a type of neurotrophic factor, is one of the most effective cytokines in slowing retinal degeneration. It prevents protein degradation to protect the retinal cells from death [199]. And tauroursodeoxycholic acid has also shown potential to be applied in the treatment of retinitis pigmentosa as an anti-apoptotic agent [200]. In addition to therapies targeting ocular symptoms, antibiotic administration can be used to reduce inflammation and ease symptoms.
Beyond drugs focusing on the clinical manifestations of SLSN, pharmacological modulators that act on ciliary-signaling pathways and enhance ciliary stability offer a complementary approach as well, particularly to address the limitations of gene therapy. Treatment with the small molecules eupatilin and fasudil reduces the accumulation of CEP290 and IFT88 along cilia, improving rhodopsin trafficking to photoreceptor OSs and reducing its mislocalization in the outer nuclear layer [201]. These effects suggest that both drugs can target ciliary protein homeostasis, thereby holding promise for treating SLSN and other ciliopathies driven by ciliary dysfunction [118,202,203]. Previous research has reported that the microtubule-targeting agent MI-181 (mitotic inhibitor-181) is a potent modulator of cilia length and biogenesis [204]. Although its direct efficacy in SLSN remains unproven, MI-181’s ability to restore ciliation at low concentrations without toxicity suggests the potential for treating ciliopathies.

6.3. Other Discoveries

Recently, optogenetics has shown the potential for the treatment of retinal diseases, involving the introduction of optic proteins into the degenerated retina to restore its function and confer photosensitivity to residual retinal cells [205]. Up to now, it has been proven to successfully restore vision in mice [206]. Thanks to the continuous developments, such as photosensitive proteins with near-infrared activation wavelengths, it may hold promise for treating SLSN, which is manifested as treatable retinal degeneration [207].
Moreover, when organ function is partly or completely lost, partial or complete organ replacement is a reasonable choice. In the case of retinal diseases, before the complete loss of rod and cone photoreceptors, cell transplantation offers a promising approach. In this method, normal cells are transplanted to replace functionally impaired cells [208]. For example, the results of implanting RPE monolayers differentiated from human embryonic stem cells into the sub-retinal space of patients with AMD have been shown to improve vision [209]. Stem cell therapy is an evolving technique, and further exploration is expected to lead to a promising prospect for SLSN treatment. In cases of severe photoreceptor loss or even complete degeneration, artificial retina implantation can be an alternative strategy. Although it can simulate the function of the natural human retinas, there still remain problems like cytotoxicity and the disruption of the blood–retinal barrier [210,211]. As a result, there is still a long way to go for the safety assurance and promotion of retinal transplantation.
As we already know, phenotypic heterogeneity in SLSN arises from mutations in different genes, which suggests that precision medicine targeting specific families and individuals will become the future direction of SLSN treatment development [64]. Complementing this approach, cilia-targeted therapies offer a promising strategy to address shared pathogenic mechanisms across different SLSN genotypes. These interventions broadly encompass three categories: (1) restoring normal ciliary function, as demonstrated by ectopic expression of arl3, arl13b, and unc119b to partially rescue photoreceptor OS abnormalities in CEP290-mutant zebrafish; (2) compensating for disrupted ciliary signaling, such as rebalancing aberrant Wnt/β–catenin pathways—implicated in renal cystogenesis in CEP290/NPHP4 mutation models—via targeting GSK3β or the Wnt co-receptor LRP6; and (3) correcting or bypassing genetic defects underlying ciliary dysfunction, including the aforementioned gene substitution or editing strategies [186,212,213]. However, most of these approaches remain in preclinical stages or are primarily focused on renal phenotypes, with few clinical trials targeting ocular manifestations, highlighting their untapped potential for further translational development.
In conclusion, regardless of the treatment modality, standardized and precise medical treatment based on individual mutations and phenotypes should be conducted. At the same time, early diagnosis and regular testing are also indispensable parts of treatment.

7. Animal Models

Similar to the advances in molecular diagnosis and gene therapy, animal models have significantly contributed to the exploration and research on SLSN, especially to the research on mechanisms. For example, some researchers have created a knock-in mouse model carrying truncating mutations of the mouse SDCCAG8 gene by using homology-directed recombination technology, which corresponds to SLSN-causing mutations in humans, respectively. Mouse embryonic fibroblasts from the knock-in mice exhibited impaired biogenesis and structural defects in cilia, which ultimately caused major phototransduction protein mislocalization outside the OS [214]. Moreover, a germline NPHP5-knockout mouse model was generated by placing a β-Geo gene trap in intron 4, removing all known functional domains. This model revealed that NPHP5/IQCB1 is necessary for mouse photoreceptor OS formation in mice [81]. Additionally, the function of CEP290 has been explored by using a mouse model of LCA with a disrupted microtubule-binding domain, which caused significant deficits in cilia formation and retinal degeneration [215]. In addition to the exploration of pathogenic mechanisms for commonly studied genes, there are mouse models for other SLSN-associated genes, revealing differences in gene expression that likely contribute to phenotypic diversity and offering new insights into the disease’s pathogenesis and individual variability [46,68,107,216,217].
Constantly, researchers are developing new mouse models, such as a mouse model deficient in TMEM218, whose major observed phenotypes are similar to those observed in SLSN. As a result, this mouse model may provide a promising prospect for elucidating the pathogenesis of diseases like SLSN, as well as exploiting new therapeutic strategies [218].
Actually, abundant research on pathological mechanisms aims at paving the way for the development of treatments, and currently, some studies using mouse models are directly exploring therapeutic techniques. For instance, by generating the Nphp5–/–; Nrl–/– double-knockout mouse model, rat cones persistently express cone pigments in the ISs without obvious degeneration, which is extremely important for prolonging the duration of gene delivery and expression and provides hope for human SLSN gene therapy [219]. Similarly, the CEP290 mouse model mentioned above provides a reasonable way for solving problems of the limited capacity of delivery carriers in gene therapy by delivering only the functional domain impacted by mutations [215].
Apart from the most researched mouse models, there are also other animal models used for SLSN relative exploration. Naturally occurring SLSN-causing mutations in genes such as CEP290 and ICQB1 have been identified in retinal degeneration cat models [220,221]. Zebrafish models with gene knockdown are also commonly used in the exploration of the pathogenic mechanism of SLSN [71,85]. Furthermore, a naturally occurring NPHP5 mutation in dogs causes disease similar to SLSN in humans, and, thus, its gene therapy could simulate and predict the efficacy of human SLSN gene treatment [184].

8. Discussion

As one of the ciliopathies mainly characterized by retinal degeneration and glomerulonephritis, SLSN was first discovered in the 1960s. However, its pathogenic mechanism is not completely clear now. With respect to the ocular phenotype of SLSN, current research mainly focuses on the connecting cilia of photoreceptors, especially the NPHP1-4-8 complex and the NPHP5-6 complex. The destruction of these structures will lead to the disruption of the corresponding functions of CC, thus triggering retinal degeneration. However, certain details remain unclear. For example, why does the degeneration of NPHP5-6, unlike the NPHP1-4-8 complex, cause a 100% incidence of retinal degeneration [80]? Moreover, other mechanisms may be involved in photoreceptor degeneration. The latest research shows that RPE, an epithelial cell that maintains the physiological cycle of photoreceptors, also has many primary cilia during development, and the impact of RPE primary cilia degeneration may occur before retinal photoreceptor defects. The role of this degeneration in SLSN may be one of the directions for future research [222].
Interestingly, with the discovery and definition of more and more ciliopathies, it is clear that they share extensive phenotypic overlaps. These connections can be explained by the molecular mechanisms and genetic basis of these disorders. As previously discussed, there exist interactions and connections between the NPHP1-4-8 complex and the NPHP5-6 complex. In fact, as a part of the TZ, they also mediate interactions with MKS, JBS proteins, and BBSome complexes. Whether these proteins that cause ciliopathies all perform biological functions as a whole remains to be verified. The genetic mutation load found in large cohorts provides strong support for the hypothesis that the severity of an individual’s overall ciliopathy phenotype is related to the population of mutations in his or her ciliopathy-associated proteins [178].
The above phenomena increasingly support the idea that ciliopathies are essentially a unified continuum of phenotypes, which increasingly emphasizes the overall concept of ciliopathies and weakens the fragmented definition of ciliopathies by phenotype [131]. This should inspire clinicians to focus on molecular and genetic diagnosis, especially the judgment of mutation load, in order to provide individualized and precise treatment.
In fact, there is no complete and targeted fundamental solution for the treatment of fibrotic disorders, and most treatments are based on supportive treatment of relevant fibrotic symptoms, and the same applies to the treatment of SLSN. For SLSN, the current treatment still focuses on the treatment of the eyes, with dialysis transplantation being the main treatment for the kidneys. There are also drugs that have therapeutic effects on the kidneys, but their effectiveness in treating eye lesions in SLSN needs to be studied and explored [160]. In clinical research, the study of these drugs on the eyes and kidneys is often fragmented, lacking an integrated approach, which is undoubtedly detrimental to the treatment of SLSN [160]. In addition, when developing eye medications, attention should also be paid to whether these agents have nephrotoxicity, which is particularly important for the treatment of SLSN that may already lead to renal impairment.
Numerous new treatments have been developed for retinitis pigmentosa in terms of supportive ocular therapy, including optogenetic therapy. However, there have been no reports on its treatment for SLSN, highlighting the need for further experimental investigations to assess its effectiveness [206,207].
Gene therapy can solve the problem of symptomatic supportive therapy, especially for the delivery of certain gene fragments, making it a promising area of research. However, there are still significant challenges in the clinical application of gene therapy, such as personalized treatment for patients with multiple modified genes and mutations, which requires considerable progress before it can become a routine therapeutic option [192].
Moreover, current animal model research remains largely unsystematic. While numerous models exist, their characterization is often fragmented. These models typically focus on isolated phenotypes or pathways rather than comprehensive disease modeling, which complicates their translational utility. A critical limitation is the uncertainty regarding functional differences in orthologous proteins between humans and model organisms, casting doubt on the relevance of animal studies for understanding human SLSN pathogenesis and guiding therapeutic development.

9. Conclusions

As a ciliopathy, the ocular mechanisms and genetic features of SLSN have yet to be thoroughly elucidated through systematic exploration. Current research has predominantly concentrated on the primary cilium structure in retinitis pigmentosa. Alterations in SLSN-associated proteins disrupt the biogenesis and structural assembly of the primary cilium in these tissues, thereby impairing signaling and molecular trafficking. Due to the intricate protein interactions, SLSN gene mutations often exert widespread effects, with clinical manifestations that overlap considerably with other ciliopathies. Additionally, despite recent advances in gene therapy and optogenetics, major challenges remain in translating these approaches into effective clinical interventions for SLSN. A deeper understanding of its ocular pathophysiology and genotype–phenotype correlations is crucial to refining therapeutic strategies and ultimately improving patient outcomes.

Author Contributions

Writing—original draft preparation, C.L. (Chen Li), C.L. (Chenyang Leng), Y.Z., and W.L.; writing—review and editing, C.L. (Chen Li) and C.L. (Chenyang Leng); visualization, C.L. (Chen Li) and Y.Z.; supervision, D.Z.; project administration, D.Z. funding acquisition, D.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Natural Science Foundation of China [No. 82170907].

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
SLSNSenior–Loken syndrome
LCALeber’s congenital amaurosis
NPHPNephronophthisis
ESRDend-stage renal disease
TZtransition zone
RPEretinal pigment epithelium
OSouter segment
ISinner segment
OMIMOnline Mendelian Inheritance
MSSMainzer–Saldino syndrome
SRTDShort-rib thoracic dysplasia
ATDAsphyxiating thoracic dystrophy
SRPSShort-rib polydactyly syndrome
IFTIntraflagellar ciliary transport
RPGRRetinitis GTPase Regulator
RPGRIP1RPGR-Interacting Protein 1
IQCB1IQ calmodulin-binding motif-containing protein 1
JSJoubert Syndrome
MKSMeckel syndrome
BBSBardet–Biedl syndrome
OFDOral–facial–digital syndrome
ALMSAlström Syndrome
USHUsher syndrome
CNSCentral Nervous System
MTSMolar Tooth Sign
DPMDuctal plate malformation
AAVAdeno-associated virus

References

  1. Hemachandar, R. Senior- Loken Syndrome—A Ciliopathy. J. Clin. Diagn. Res. 2014, 8, MD04–MD05. [Google Scholar] [CrossRef]
  2. Otto, E.A.; Loeys, B.; Khanna, H.; Hellemans, J.; Sudbrak, R.; Fan, S.; Muerb, U.; O’Toole, J.F.; Helou, J.; Attanasio, M.; et al. Nephrocystin-5, a Ciliary IQ Domain Protein, Is Mutated in Senior-Loken Syndrome and Interacts with RPGR and Calmodulin. Nat. Genet. 2005, 37, 282–288. [Google Scholar] [CrossRef] [PubMed]
  3. Baehr, W.; Hanke-Gogokhia, C.; Sharif, A.; Reed, M.; Dahl, T.; Frederick, J.M.; Ying, G. Insights into Photoreceptor Ciliogenesis Revealed by Animal Models. Prog. Retin. Eye Res. 2019, 71, 26–56. [Google Scholar] [CrossRef]
  4. Ran, J.; Zhou, J. Targeting the Photoreceptor Cilium for the Treatment of Retinal Diseases. Acta Pharmacol. Sin. 2020, 41, 1410–1415. [Google Scholar] [CrossRef]
  5. Huang, C.-H.; Yang, C.-M.; Yang, C.-H.; Hou, Y.-C.; Chen, T.-C. Leber’s Congenital Amaurosis: Current Concepts of Genotype-Phenotype Correlations. Genes 2021, 12, 1261. [Google Scholar] [CrossRef]
  6. Hildebrandt, F.; Zhou, W. Nephronophthisis-Associated Ciliopathies. J. Am. Soc. Nephrol. 2007, 18, 1855–1871. [Google Scholar] [CrossRef]
  7. Kumaran, N.; Moore, A.T.; Weleber, R.G.; Michaelides, M. Leber Congenital Amaurosis/Early-Onset Severe Retinal Dystrophy: Clinical Features, Molecular Genetics and Therapeutic Interventions. Br. J. Ophthalmol. 2017, 101, 1147–1154. [Google Scholar] [CrossRef]
  8. Georgiou, M.; Robson, A.G.; Fujinami, K.; de Guimarães, T.A.C.; Fujinami-Yokokawa, Y.; Daich Varela, M.; Pontikos, N.; Kalitzeos, A.; Mahroo, O.A.; Webster, A.R.; et al. Phenotyping and Genotyping Inherited Retinal Diseases: Molecular Genetics, Clinical and Imaging Features, and Therapeutics of Macular Dystrophies, Cone and Cone-Rod Dystrophies, Rod-Cone Dystrophies, Leber Congenital Amaurosis, and Cone Dysfunction Syndromes. Prog. Retin. Eye Res. 2024, 100, 101244. [Google Scholar] [CrossRef]
  9. Hartong, D.T.; Berson, E.L.; Dryja, T.P. Retinitis Pigmentosa. Lancet 2006, 368, 1795–1809. [Google Scholar] [CrossRef]
  10. Liu, W.; Liu, S.; Li, P.; Yao, K. Retinitis Pigmentosa: Progress in Molecular Pathology and Biotherapeutical Strategies. Int. J. Mol. Sci. 2022, 23, 4883. [Google Scholar] [CrossRef]
  11. Stokman, M.F.; Saunier, S.; Benmerah, A. Renal Ciliopathies: Sorting Out Therapeutic Approaches for Nephronophthisis. Front. Cell Dev. Biol. 2021, 9, 653138. [Google Scholar] [CrossRef] [PubMed]
  12. Simms, R.J.; Eley, L.; Sayer, J.A. Nephronophthisis. Eur. J. Hum. Genet. 2009, 17, 406–416. [Google Scholar] [CrossRef]
  13. Salomon, R.; Saunier, S.; Niaudet, P. Nephronophthisis. Pediatr. Nephrol. 2008, 24, 2333. [Google Scholar] [CrossRef] [PubMed]
  14. Othman, M.; Rashed, A.; Bakr, A. The Senior-Loken Syndrome: Two Cases from the State of Qatar. J. Clin. Diagn. Res. 2012, 6, 1411–1413. [Google Scholar] [CrossRef]
  15. Liu, L.; Zhou, Y.; Liu, Y.; Ding, J.; Xie, Y.; Li, N. A Novel Pathogenic Variant of CEP164 in an Infant with Senior-Loken Syndrome. Pediatr. Investig. 2023, 7, 140–143. [Google Scholar] [CrossRef]
  16. Mainzer, F.; Saldino, R.M.; Ozonoff, M.B.; Minagi, H. Familial Nephropathy Associatdd with Retinitis Pigmentosa, Cerebellar Ataxia and Skeletal Abnormalities. Am. J. Med. 1970, 49, 556–562. [Google Scholar] [CrossRef]
  17. Tay, S.A.; Vincent, A.L. Senior-Løken Syndrome and Intracranial Hypertension. Ophthalmic Genet. 2020, 41, 354–357. [Google Scholar] [CrossRef]
  18. Ishikawa, H.; Marshall, W.F. Ciliogenesis: Building the Cell’s Antenna. Nat. Rev. Mol. Cell Biol. 2011, 12, 222–234. [Google Scholar] [CrossRef]
  19. Anvarian, Z.; Mykytyn, K.; Mukhopadhyay, S.; Pedersen, L.B.; Christensen, S.T. Cellular Signalling by Primary Cilia in Development, Organ Function and Disease. Nat. Rev. Nephrol. 2019, 15, 199–219. [Google Scholar] [CrossRef]
  20. Reddy Palicharla, V.; Mukhopadhyay, S. Molecular and Structural Perspectives on Protein Trafficking to the Primary Cilium Membrane. Biochem. Soc. Trans. 2024, 52, 1473–1487. [Google Scholar] [CrossRef]
  21. Bai, Y.; Wei, C.; Li, P.; Sun, X.; Cai, G.; Chen, X.; Hong, Q. Primary Cilium in Kidney Development, Function and Disease. Front. Endocrinol. 2022, 13, 952055. [Google Scholar] [CrossRef] [PubMed]
  22. Yang, T.T.; Chong, W.M.; Wang, W.-J.; Mazo, G.; Tanos, B.; Chen, Z.; Tran, T.M.N.; Chen, Y.-D.; Weng, R.R.; Huang, C.-E.; et al. Super-Resolution Architecture of Mammalian Centriole Distal Appendages Reveals Distinct Blade and Matrix Functional Components. Nat. Commun. 2018, 9, 2023. [Google Scholar] [CrossRef]
  23. Marshall, W.F. Basal Bodies Platforms for Building Cilia. Curr. Top. Dev. Biol. 2008, 85, 1–22. [Google Scholar] [CrossRef]
  24. Sorokin, S. Centrioles and the Formation of Rudimentary Cilia by Fibroblasts and Smooth Muscle Cells. J. Cell Biol. 1962, 15, 363–377. [Google Scholar] [CrossRef]
  25. Sorokin, S.P. Reconstructions of Centriole Formation and Ciliogenesis in Mammalian Lungs. J. Cell Sci. 1968, 3, 207–230. [Google Scholar] [CrossRef]
  26. Wang, L.; Wen, X.; Wang, Z.; Lin, Z.; Li, C.; Zhou, H.; Yu, H.; Li, Y.; Cheng, Y.; Chen, Y.; et al. Ciliary Transition Zone Proteins Coordinate Ciliary Protein Composition and Ectosome Shedding. Nat. Commun. 2022, 13, 3997. [Google Scholar] [CrossRef]
  27. Garcia-Gonzalo, F.R.; Reiter, J.F. Open Sesame: How Transition Fibers and the Transition Zone Control Ciliary Composition. Cold Spring Harb. Perspect. Biol. 2017, 9, a028134. [Google Scholar] [CrossRef]
  28. May-Simera, H.; Nagel-Wolfrum, K.; Wolfrum, U. Cilia—The Sensory Antennae in the Eye. Prog. Retin. Eye Res. 2017, 60, 144–180. [Google Scholar] [CrossRef]
  29. Blitzer, A.L.; Panagis, L.; Gusella, G.L.; Danias, J.; Mlodzik, M.; Iomini, C. Primary Cilia Dynamics Instruct Tissue Patterning and Repair of Corneal Endothelium. Proc. Natl. Acad. Sci. USA 2011, 108, 2819–2824. [Google Scholar] [CrossRef]
  30. Chrystal, P.W.; Lambacher, N.J.; Doucette, L.P.; Bellingham, J.; Schiff, E.R.; Noel, N.C.L.; Li, C.; Tsiropoulou, S.; Casey, G.A.; Zhai, Y.; et al. The Inner Junction Protein CFAP20 Functions in Motile and Non-Motile Cilia and Is Critical for Vision. Nat. Commun. 2022, 13, 6595. [Google Scholar] [CrossRef]
  31. Sun, C.; Zhou, J.; Meng, X. Primary Cilia in Retinal Pigment Epithelium Development and Diseases. J. Cell. Mol. Med. 2021, 25, 9084–9088. [Google Scholar] [CrossRef] [PubMed]
  32. Liu, X.; Pacwa, A.; Bresciani, G.; Swierczynska, M.; Dorecka, M.; Smedowski, A. Retinal Primary Cilia and Their Dysfunction in Retinal Neurodegenerative Diseases: Beyond Ciliopathies. Mol. Med. 2024, 30, 109. [Google Scholar] [CrossRef] [PubMed]
  33. Chen, H.Y.; Kelley, R.A.; Li, T.; Swaroop, A. Primary Cilia Biogenesis and Associated Retinal Ciliopathies. Semin. Cell Dev. Biol. 2021, 110, 70–88. [Google Scholar] [CrossRef] [PubMed]
  34. Ghossoub, R.; Molla-Herman, A.; Bastin, P.; Benmerah, A. The Ciliary Pocket: A Once-Forgotten Membrane Domain at the Base of Cilia. Biol. Cell 2011, 103, 131–144. [Google Scholar] [CrossRef]
  35. Chandra, B.; Tung, M.L.; Hsu, Y.; Scheetz, T.; Sheffield, V.C. Retinal Ciliopathies through the Lens of Bardet-Biedl Syndrome: Past, Present and Future. Prog. Retin. Eye Res. 2022, 89, 101035. [Google Scholar] [CrossRef]
  36. Løken, A.C.; Hanssen, O.; Halvorsen, S.; Jølster, N.J. Hereditary Renal Dysplasia and Blindness. Acta Paediatr. 1961, 50, 177–184. [Google Scholar] [CrossRef]
  37. Senior, B.; Friedmann, A.I.; Braudo, J.L. Juvenile Familial Nephropathy with Tapetoretinal Degeneration. A New Oculorenal Dystrophy. Am. J. Ophthalmol. 1961, 52, 625–633. [Google Scholar] [CrossRef]
  38. Home—OMIM. Available online: https://www.omim.org/ (accessed on 26 April 2025).
  39. UniProt. Available online: https://www.uniprot.org (accessed on 26 April 2025).
  40. Gene. Available online: https://www.ncbi.nlm.nih.gov/datasets/gene/ (accessed on 26 April 2025).
  41. Jauregui, A.R.; Nguyen, K.C.Q.; Hall, D.H.; Barr, M.M. The Caenorhabditis Elegans Nephrocystins Act as Global Modifiers of Cilium Structure. J. Cell Biol. 2008, 180, 973–988. [Google Scholar] [CrossRef]
  42. Caridi, G.; Dagnino, M.; Rossi, A.; Valente, E.M.; Bertini, E.; Fazzi, E.; Emma, F.; Murer, L.; Verrina, E.; Ghiggeri, G.M. Nephronophthisis Type 1 Deletion Syndrome with Neurological Symptoms: Prevalence and Significance of the Association. Kidney Int. 2006, 70, 1342–1347. [Google Scholar] [CrossRef]
  43. Lindstrand, A.; Davis, E.E.; Carvalho, C.M.B.; Pehlivan, D.; Willer, J.R.; Tsai, I.-C.; Ramanathan, S.; Zuppan, C.; Sabo, A.; Muzny, D.; et al. Recurrent CNVs and SNVs at the NPHP1 Locus Contribute Pathogenic Alleles to Bardet-Biedl Syndrome. Am. J. Hum. Genet. 2014, 94, 745–754. [Google Scholar] [CrossRef]
  44. Yahalom, C.; Volovelsky, O.; Macarov, M.; Altalbishi, A.; Alsweiti, Y.; Schneider, N.; Hanany, M.; Khan, M.I.; Cremers, F.P.M.; Anteby, I.; et al. SENIOR-LØKEN SYNDROME: A Case Series and Review of the Renoretinal Phenotype and Advances of Molecular Diagnosis. Retina 2021, 41, 2179–2187. [Google Scholar] [CrossRef] [PubMed]
  45. Roig, J. NEK8, a NIMA-Family Protein Kinase at the Core of the Ciliary INV Complex. Cell Commun. Signal. 2025, 23, 170. [Google Scholar] [CrossRef] [PubMed]
  46. Bergmann, C.; Fliegauf, M.; Brüchle, N.O.; Frank, V.; Olbrich, H.; Kirschner, J.; Schermer, B.; Schmedding, I.; Kispert, A.; Kränzlin, B.; et al. Loss of Nephrocystin-3 Function Can Cause Embryonic Lethality, Meckel-Gruber-like Syndrome, Situs Inversus, and Renal-Hepatic-Pancreatic Dysplasia. Am. J. Hum. Genet. 2008, 82, 959–970. [Google Scholar] [CrossRef] [PubMed]
  47. Yue, Z.; Lin, H.; Li, M.; Wang, H.; Liu, T.; Hu, M.; Chen, H.; Tong, H.; Sun, L. Clinical and Pathological Features and Varied Mutational Spectra of Pathogenic Genes in 55 Chinese Patients with Nephronophthisis. Clin. Chim. Acta 2020, 506, 136–144. [Google Scholar] [CrossRef]
  48. Omran, H.; Sasmaz, G.; Häffner, K.; Volz, A.; Olbrich, H.; Melkaoui, R.; Otto, E.; Wienker, T.F.; Korinthenberg, R.; Brandis, M.; et al. Identification of a Gene Locus for Senior-Løken Syndrome in the Region of the Nephronophthisis Type 3 Gene. J. Am. Soc. Nephrol. 2002, 13, 75–79. [Google Scholar] [CrossRef]
  49. Tang, X.; Liu, C.; Liu, X.; Chen, J.; Fan, X.; Liu, J.; Ma, D.; Cao, G.; Chen, Z.; Xu, D.; et al. Phenotype and Genotype Spectra of a Chinese Cohort with Nephronophthisis-Related Ciliopathy. J. Med. Genet. 2022, 59, 147–154. [Google Scholar] [CrossRef]
  50. Penchev, V.; Boueva, A.; Kamenarova, K.; Roussinov, D.; Tzveova, R.; Ivanova, M.; Dimitrova, V.; Kremensky, I.; Mitev, V.; Kaneva, R.; et al. A Familial Case of Severe Infantile Nephronophthisis Explained by Oligogenic Inheritance. Eur. J. Med. Genet. 2017, 60, 321–325. [Google Scholar] [CrossRef]
  51. Arts, H.H.; Doherty, D.; van Beersum, S.E.C.; Parisi, M.A.; Letteboer, S.J.F.; Gorden, N.T.; Peters, T.A.; Märker, T.; Voesenek, K.; Kartono, A.; et al. Mutations in the Gene Encoding the Basal Body Protein RPGRIP1L, a Nephrocystin-4 Interactor, Cause Joubert Syndrome. Nat. Genet. 2007, 39, 882–888. [Google Scholar] [CrossRef]
  52. Kruczek, K.; Qu, Z.; Welby, E.; Shimada, H.; Hiriyanna, S.; English, M.A.; Zein, W.M.; Brooks, B.P.; Swaroop, A. In Vitro Modeling and Rescue of Ciliopathy Associated with IQCB1/NPHP5 Mutations Using Patient-Derived Cells. Stem Cell Rep. 2022, 17, 2172–2186. [Google Scholar] [CrossRef]
  53. Tory, K.; Lacoste, T.; Burglen, L.; Morinière, V.; Boddaert, N.; Macher, M.-A.; Llanas, B.; Nivet, H.; Bensman, A.; Niaudet, P.; et al. High NPHP1 and NPHP6 Mutation Rate in Patients with Joubert Syndrome and Nephronophthisis: Potential Epistatic Effect of NPHP6 and AHI1 Mutations in Patients with NPHP1 Mutations. J. Am. Soc. Nephrol. 2007, 18, 1566–1575. [Google Scholar] [CrossRef]
  54. Coppieters, F.; Lefever, S.; Leroy, B.P.; De Baere, E. CEP290, a Gene with Many Faces: Mutation Overview and Presentation of CEP290base. Hum. Mutat. 2010, 31, 1097–1108. [Google Scholar] [CrossRef] [PubMed]
  55. Parfitt, D.A.; Lane, A.; Ramsden, C.M.; Carr, A.-J.F.; Munro, P.M.; Jovanovic, K.; Schwarz, N.; Kanuga, N.; Muthiah, M.N.; Hull, S.; et al. Identification and Correction of Mechanisms Underlying Inherited Blindness in Human iPSC-Derived Optic Cups. Cell Stem Cell 2016, 18, 769–781. [Google Scholar] [CrossRef] [PubMed]
  56. Airik, R.; Slaats, G.G.; Guo, Z.; Weiss, A.-C.; Khan, N.; Ghosh, A.; Hurd, T.W.; Bekker-Jensen, S.; Schrøder, J.M.; Elledge, S.J.; et al. Renal-Retinal Ciliopathy Gene Sdccag8 Regulates DNA Damage Response Signaling. J. Am. Soc. Nephrol. 2014, 25, 2573–2583. [Google Scholar] [CrossRef] [PubMed]
  57. Insolera, R.; Shao, W.; Airik, R.; Hildebrandt, F.; Shi, S.-H. SDCCAG8 Regulates Pericentriolar Material Recruitment and Neuronal Migration in the Developing Cortex. Neuron 2014, 83, 805–822. [Google Scholar] [CrossRef]
  58. Bredrup, C.; Saunier, S.; Oud, M.M.; Fiskerstrand, T.; Hoischen, A.; Brackman, D.; Leh, S.M.; Midtbø, M.; Filhol, E.; Bole-Feysot, C.; et al. Ciliopathies with Skeletal Anomalies and Renal Insufficiency Due to Mutations in the IFT-A Gene WDR19. Am. J. Hum. Genet. 2011, 89, 634–643. [Google Scholar] [CrossRef]
  59. Tang, X.; Yi, S.; Qin, Z.; Yi, S.; Chen, J.; Yang, Q.; Li, S.; Luo, J. Compound Heterozygous WDR19 Variants Associated with Nephronophthisis, Caroli Disease, Refractory Epilepsy and Congenital Bilateral Central Blindness: Case Report. Heliyon 2024, 10, e23257. [Google Scholar] [CrossRef]
  60. Coussa, R.G.; Otto, E.A.; Gee, H.Y.; Arthurs, P.; Ren, H.; Lopez, I.; Keser, V.; Fu, Q.; Faingold, R.; Khan, A.; et al. WDR19: An Ancient, Retrograde, Intraflagellar Ciliary Protein Is Mutated in Autosomal Recessive Retinitis Pigmentosa and in Senior-Loken Syndrome. Clin. Genet. 2013, 84, 150–159. [Google Scholar] [CrossRef]
  61. Liu, L.; Huang, Y.; Fang, F.; Zhou, H.; Liu, X. A Case Report of Intrahepatic Bile Duct Dilatation Caused by WDR19 Gene Mutation and Presented as Caroli Syndrome. Transl. Pediatr. 2024, 13, 682–689. [Google Scholar] [CrossRef]
  62. Bizet, A.A.; Becker-Heck, A.; Ryan, R.; Weber, K.; Filhol, E.; Krug, P.; Halbritter, J.; Delous, M.; Lasbennes, M.-C.; Linghu, B.; et al. Mutations in TRAF3IP1/IFT54 Reveal a New Role for IFT Proteins in Microtubule Stabilization. Nat. Commun. 2015, 6, 8666. [Google Scholar] [CrossRef]
  63. Zhang, W.; Taylor, S.P.; Ennis, H.A.; Forlenza, K.N.; Duran, I.; Li, B.; Sanchez, J.A.O.; Nevarez, L.; Nickerson, D.A.; Bamshad, M.; et al. Expanding the Genetic Architecture and Phenotypic Spectrum in the Skeletal Ciliopathies. Hum. Mutat. 2018, 39, 152–166. [Google Scholar] [CrossRef]
  64. Wang, J.; Li, S.; Jiang, Y.; Wang, Y.; Ouyang, J.; Yi, Z.; Sun, W.; Jia, X.; Xiao, X.; Wang, P.; et al. Pathogenic Variants in CEP290 or IQCB1 Cause Earlier-Onset Retinopathy in Senior-Loken Syndrome Compared to Those in INVS, NPHP3, or NPHP4. Am. J. Ophthalmol. 2023, 252, 188–204. [Google Scholar] [CrossRef] [PubMed]
  65. Hildebrandt, F.; Otto, E.; Rensing, C.; Nothwang, H.G.; Vollmer, M.; Adolphs, J.; Hanusch, H.; Brandis, M. A Novel Gene Encoding an SH3 Domain Protein Is Mutated in Nephronophthisis Type 1. Nat. Genet. 1997, 17, 149–153. [Google Scholar] [CrossRef] [PubMed]
  66. Lienkamp, S.; Ganner, A.; Walz, G. Inversin, Wnt Signaling and Primary Cilia. Differentiation 2012, 83, S49–S55. [Google Scholar] [CrossRef] [PubMed]
  67. Delous, M.; Hellman, N.E.; Gaudé, H.-M.; Silbermann, F.; Le Bivic, A.; Salomon, R.; Antignac, C.; Saunier, S. Nephrocystin-1 and Nephrocystin-4 Are Required for Epithelial Morphogenesis and Associate with PALS1/PATJ and Par6. Hum. Mol. Genet. 2009, 18, 4711–4723. [Google Scholar] [CrossRef]
  68. Sang, L.; Miller, J.J.; Corbit, K.C.; Giles, R.H.; Brauer, M.J.; Otto, E.A.; Baye, L.M.; Wen, X.; Scales, S.J.; Kwong, M.; et al. Mapping the NPHP-JBTS-MKS Protein Network Reveals Ciliopathy Disease Genes and Pathways. Cell 2011, 145, 513–528. [Google Scholar] [CrossRef]
  69. Jiang, S.-T.; Chiou, Y.-Y.; Wang, E.; Chien, Y.-L.; Ho, H.-H.; Tsai, F.-J.; Lin, C.-Y.; Tsai, S.-P.; Li, H. Essential Role of Nephrocystin in Photoreceptor Intraflagellar Transport in Mouse. Hum. Mol. Genet. 2009, 18, 1566–1577. [Google Scholar] [CrossRef]
  70. Olbrich, H.; Fliegauf, M.; Hoefele, J.; Kispert, A.; Otto, E.; Volz, A.; Wolf, M.T.; Sasmaz, G.; Trauer, U.; Reinhardt, R.; et al. Mutations in a Novel Gene, NPHP3, Cause Adolescent Nephronophthisis, Tapeto-Retinal Degeneration and Hepatic Fibrosis. Nat. Genet. 2003, 34, 455–459. [Google Scholar] [CrossRef]
  71. Wright, K.J.; Baye, L.M.; Olivier-Mason, A.; Mukhopadhyay, S.; Sang, L.; Kwong, M.; Wang, W.; Pretorius, P.R.; Sheffield, V.C.; Sengupta, P.; et al. An ARL3-UNC119-RP2 GTPase Cycle Targets Myristoylated NPHP3 to the Primary Cilium. Genes. Dev. 2011, 25, 2347–2360. [Google Scholar] [CrossRef]
  72. Nakata, K.; Shiba, D.; Kobayashi, D.; Yokoyama, T. Targeting of Nphp3 to the Primary Cilia Is Controlled by an N-Terminal Myristoylation Site and Coiled-Coil Domains. Cytoskeleton 2012, 69, 221–234. [Google Scholar] [CrossRef]
  73. Schuermann, M.J.; Otto, E.; Becker, A.; Saar, K.; Rüschendorf, F.; Polak, B.C.; Ala-Mello, S.; Hoefele, J.; Wiedensohler, A.; Haller, M.; et al. Mapping of Gene Loci for Nephronophthisis Type 4 and Senior-Løken Syndrome, to Chromosome 1p36. Am. J. Hum. Genet. 2002, 70, 1240–1246. [Google Scholar] [CrossRef]
  74. Murga-Zamalloa, C.A.; Swaroop, A.; Khanna, H. RPGR-Containing Protein Complexes in Syndromic and Non-Syndromic Retinal Degeneration Due to Ciliary Dysfunction. J. Genet. 2009, 88, 399–407. [Google Scholar] [CrossRef] [PubMed]
  75. Patil, H.; Tserentsoodol, N.; Saha, A.; Hao, Y.; Webb, M.; Ferreira, P.A. Selective Loss of RPGRIP1-Dependent Ciliary Targeting of NPHP4, RPGR and SDCCAG8 Underlies the Degeneration of Photoreceptor Neurons. Cell Death Dis. 2012, 3, e355. [Google Scholar] [CrossRef] [PubMed]
  76. Yasunaga, T.; Hoff, S.; Schell, C.; Helmstädter, M.; Kretz, O.; Kuechlin, S.; Yakulov, T.A.; Engel, C.; Müller, B.; Bensch, R.; et al. The Polarity Protein Inturned Links NPHP4 to Daam1 to Control the Subapical Actin Network in Multiciliated Cells. J. Cell Biol. 2015, 211, 963–973. [Google Scholar] [CrossRef]
  77. Habbig, S.; Bartram, M.P.; Müller, R.U.; Schwarz, R.; Andriopoulos, N.; Chen, S.; Sägmüller, J.G.; Hoehne, M.; Burst, V.; Liebau, M.C.; et al. NPHP4, a Cilia-Associated Protein, Negatively Regulates the Hippo Pathway. J. Cell Biol. 2011, 193, 633–642. [Google Scholar] [CrossRef]
  78. Burcklé, C.; Gaudé, H.-M.; Vesque, C.; Silbermann, F.; Salomon, R.; Jeanpierre, C.; Antignac, C.; Saunier, S.; Schneider-Maunoury, S. Control of the Wnt Pathways by Nephrocystin-4 Is Required for Morphogenesis of the Zebrafish Pronephros. Hum. Mol. Genet. 2011, 20, 2611–2627. [Google Scholar] [CrossRef]
  79. Borgal, L.; Habbig, S.; Hatzold, J.; Liebau, M.C.; Dafinger, C.; Sacarea, I.; Hammerschmidt, M.; Benzing, T.; Schermer, B. The Ciliary Protein Nephrocystin-4 Translocates the Canonical Wnt Regulator Jade-1 to the Nucleus to Negatively Regulate β-Catenin Signaling. J. Biol. Chem. 2012, 287, 25370–25380. [Google Scholar] [CrossRef]
  80. Hildebrandt, F.; Attanasio, M.; Otto, E. Nephronophthisis: Disease Mechanisms of a Ciliopathy. J. Am. Soc. Nephrol. 2009, 20, 23–35. [Google Scholar] [CrossRef]
  81. Ronquillo, C.C.; Hanke-Gogokhia, C.; Revelo, M.P.; Frederick, J.M.; Jiang, L.; Baehr, W. Ciliopathy-Associated IQCB1/NPHP5 Protein Is Required for Mouse Photoreceptor Outer Segment Formation. FASEB J. 2016, 30, 3400–3412. [Google Scholar] [CrossRef]
  82. Barbelanne, M.; Song, J.; Ahmadzai, M.; Tsang, W.Y. Pathogenic NPHP5 Mutations Impair Protein Interaction with Cep290, a Prerequisite for Ciliogenesis. Hum. Mol. Genet. 2013, 22, 2482–2494. [Google Scholar] [CrossRef]
  83. Hsu, Y.; Seo, S.; Sheffield, V.C. Photoreceptor Cilia, in Contrast to Primary Cilia, Grant Entry to a Partially Assembled BBSome. Hum. Mol. Genet. 2021, 30, 87–102. [Google Scholar] [CrossRef]
  84. Zhao, C.; Malicki, J. Nephrocystins and MKS Proteins Interact with IFT Particle and Facilitate Transport of Selected Ciliary Cargos. EMBO J. 2011, 30, 2532–2544. [Google Scholar] [CrossRef] [PubMed]
  85. Sayer, J.A.; Otto, E.A.; O’Toole, J.F.; Nurnberg, G.; Kennedy, M.A.; Becker, C.; Hennies, H.C.; Helou, J.; Attanasio, M.; Fausett, B.V.; et al. The Centrosomal Protein Nephrocystin-6 Is Mutated in Joubert Syndrome and Activates Transcription Factor ATF4. Nat. Genet. 2006, 38, 674–681. [Google Scholar] [CrossRef]
  86. Tsang, W.Y.; Bossard, C.; Khanna, H.; Peränen, J.; Swaroop, A.; Malhotra, V.; Dynlacht, B.D. CP110 Suppresses Primary Cilia Formation through Its Interaction with CEP290, a Protein Deficient in Human Ciliary Disease. Dev. Cell 2008, 15, 187–197. [Google Scholar] [CrossRef]
  87. Kobayashi, T.; Kim, S.; Lin, Y.-C.; Inoue, T.; Dynlacht, B.D. The CP110-Interacting Proteins Talpid3 and Cep290 Play Overlapping and Distinct Roles in Cilia Assembly. J. Cell Biol. 2014, 204, 215–229. [Google Scholar] [CrossRef]
  88. Frank, V.; den Hollander, A.I.; Brüchle, N.O.; Zonneveld, M.N.; Nürnberg, G.; Becker, C.; Du Bois, G.; Kendziorra, H.; Roosing, S.; Senderek, J.; et al. Mutations of the CEP290 Gene Encoding a Centrosomal Protein Cause Meckel-Gruber Syndrome. Hum. Mutat. 2008, 29, 45–52. [Google Scholar] [CrossRef]
  89. Barbelanne, M.; Hossain, D.; Chan, D.P.; Peränen, J.; Tsang, W.Y. Nephrocystin Proteins NPHP5 and Cep290 Regulate BBSome Integrity, Ciliary Trafficking and Cargo Delivery. Hum. Mol. Genet. 2015, 24, 2185–2200. [Google Scholar] [CrossRef]
  90. Otto, E.A.; Hurd, T.W.; Airik, R.; Chaki, M.; Zhou, W.; Stoetzel, C.; Patil, S.B.; Levy, S.; Ghosh, A.K.; Murga-Zamalloa, C.A.; et al. Candidate Exome Capture Identifies Mutation of SDCCAG8 as the Cause of a Retinal-Renal Ciliopathy. Nat. Genet. 2010, 42, 840–850. [Google Scholar] [CrossRef]
  91. Tsutsumi, R.; Chaya, T.; Tsujii, T.; Furukawa, T. The Carboxyl-Terminal Region of SDCCAG8 Comprises a Functional Module Essential for Cilia Formation as Well as Organ Development and Homeostasis. J. Biol. Chem. 2022, 298, 101686. [Google Scholar] [CrossRef]
  92. Schaefer, E.; Zaloszyc, A.; Lauer, J.; Durand, M.; Stutzmann, F.; Perdomo-Trujillo, Y.; Redin, C.; Bennouna Greene, V.; Toutain, A.; Perrin, L.; et al. Mutations in SDCCAG8/NPHP10 Cause Bardet-Biedl Syndrome and Are Associated with Penetrant Renal Disease and Absent Polydactyly. Mol. Syndromol. 2011, 1, 273–281. [Google Scholar] [CrossRef]
  93. Airik, R.; Schueler, M.; Airik, M.; Cho, J.; Ulanowicz, K.A.; Porath, J.D.; Hurd, T.W.; Bekker-Jensen, S.; Schrøder, J.M.; Andersen, J.S.; et al. SDCCAG8 Interacts with RAB Effector Proteins RABEP2 and ERC1 and Is Required for Hedgehog Signaling. PLoS ONE 2016, 11, e0156081. [Google Scholar] [CrossRef] [PubMed]
  94. Lin, B.; White, J.T.; Utleg, A.G.; Wang, S.; Ferguson, C.; True, L.D.; Vessella, R.; Hood, L.; Nelson, P.S. Isolation and Characterization of Human and Mouse WDR19,a Novel WD-Repeat Protein Exhibiting Androgen-Regulated Expression in Prostate Epithelium. Genomics 2003, 82, 331–342. [Google Scholar] [CrossRef] [PubMed]
  95. Mukhopadhyay, S.; Wen, X.; Chih, B.; Nelson, C.D.; Lane, W.S.; Scales, S.J.; Jackson, P.K. TULP3 Bridges the IFT-A Complex and Membrane Phosphoinositides to Promote Trafficking of G Protein-Coupled Receptors into Primary Cilia. Genes. Dev. 2010, 24, 2180–2193. [Google Scholar] [CrossRef] [PubMed]
  96. Ling, L.; Goeddel, D.V. MIP-T3, a Novel Protein Linking Tumor Necrosis Factor Receptor-Associated Factor 3 to the Microtubule Network. J. Biol. Chem. 2000, 275, 23852–23860. [Google Scholar] [CrossRef]
  97. Ghossoub, R.; Hu, Q.; Failler, M.; Rouyez, M.-C.; Spitzbarth, B.; Mostowy, S.; Wolfrum, U.; Saunier, S.; Cossart, P.; Jamesnelson, W.; et al. Septins 2, 7 and 9 and MAP4 Colocalize along the Axoneme in the Primary Cilium and Control Ciliary Length. J. Cell Sci. 2013, 126, 2583–2594. [Google Scholar] [CrossRef]
  98. Zhang, D.; Aravind, L. Novel Transglutaminase-like Peptidase and C2 Domains Elucidate the Structure, Biogenesis and Evolution of the Ciliary Compartment. Cell Cycle 2012, 11, 3861–3875. [Google Scholar] [CrossRef]
  99. Remans, K.; Bürger, M.; Vetter, I.R.; Wittinghofer, A. C2 Domains as Protein-Protein Interaction Modules in the Ciliary Transition Zone. Cell Rep. 2014, 8, 1–9. [Google Scholar] [CrossRef]
  100. Winkelbauer, M.E.; Schafer, J.C.; Haycraft, C.J.; Swoboda, P.; Yoder, B.K. The C. Elegans Homologs of Nephrocystin-1 and Nephrocystin-4 Are Cilia Transition Zone Proteins Involved in Chemosensory Perception. J. Cell Sci. 2005, 118, 5575–5587. [Google Scholar] [CrossRef]
  101. Shi, X.; Garcia, G.; Van De Weghe, J.C.; McGorty, R.; Pazour, G.J.; Doherty, D.; Huang, B.; Reiter, J.F. Super-Resolution Microscopy Reveals That Disruption of Ciliary Transition-Zone Architecture Causes Joubert Syndrome. Nat. Cell Biol. 2017, 19, 1178–1188. [Google Scholar] [CrossRef]
  102. Williams, C.L.; Li, C.; Kida, K.; Inglis, P.N.; Mohan, S.; Semenec, L.; Bialas, N.J.; Stupay, R.M.; Chen, N.; Blacque, O.E.; et al. MKS and NPHP Modules Cooperate to Establish Basal Body/Transition Zone Membrane Associations and Ciliary Gate Function during Ciliogenesis. J. Cell Biol. 2011, 192, 1023–1041. [Google Scholar] [CrossRef]
  103. Adamiok-Ostrowska, A.; Piekiełko-Witkowska, A. Ciliary Genes in Renal Cystic Diseases. Cells 2020, 9, 907. [Google Scholar] [CrossRef]
  104. Mollet, G.; Silbermann, F.; Delous, M.; Salomon, R.; Antignac, C.; Saunier, S. Characterization of the Nephrocystin/Nephrocystin-4 Complex and Subcellular Localization of Nephrocystin-4 to Primary Cilia and Centrosomes. Hum. Mol. Genet. 2005, 14, 645–656. [Google Scholar] [CrossRef] [PubMed]
  105. Frederick, J.M.; Hanke-Gogokhia, C.; Ying, G.; Baehr, W. Diffuse or Hitch a Ride: How Photoreceptor Lipidated Proteins Get from Here to There. Biol. Chem. 2020, 401, 573–584. [Google Scholar] [CrossRef]
  106. Park, K.; Leroux, M.R. Composition, Organization and Mechanisms of the Transition Zone, a Gate for the Cilium. EMBO Rep. 2022, 23, e55420. [Google Scholar] [CrossRef]
  107. Won, J.; de Evsikova, C.M.; Smith, R.S.; Hicks, W.L.; Edwards, M.M.; Longo-Guess, C.; Li, T.; Naggert, J.K.; Nishina, P.M. NPHP4 Is Necessary for Normal Photoreceptor Ribbon Synapse Maintenance and Outer Segment Formation, and for Sperm Development. Hum. Mol. Genet. 2011, 20, 482–496. [Google Scholar] [CrossRef]
  108. Khanna, H. Photoreceptor Sensory Cilium: Traversing the Ciliary Gate. Cells 2015, 4, 674–686. [Google Scholar] [CrossRef]
  109. Vössing, C.; Atigbire, P.; Eilers, J.; Markus, F.; Stieger, K.; Song, F.; Neidhardt, J. The Major Ciliary Isoforms of RPGR Build Different Interaction Complexes with INPP5E and RPGRIP1L. Int. J. Mol. Sci. 2021, 22, 3583. [Google Scholar] [CrossRef]
  110. Murga-Zamalloa, C.A.; Desai, N.J.; Hildebrandt, F.; Khanna, H. Interaction of Ciliary Disease Protein Retinitis Pigmentosa GTPase Regulator with Nephronophthisis-Associated Proteins in Mammalian Retinas. Mol. Vis. 2010, 16, 1373–1381. [Google Scholar]
  111. Anand, M.; Khanna, H. Ciliary Transition Zone (TZ) Proteins RPGR and CEP290: Role in Photoreceptor Cilia and Degenerative Diseases. Expert. Opin. Ther. Targets 2012, 16, 541–551. [Google Scholar] [CrossRef]
  112. Raghupathy, R.K.; Zhang, X.; Liu, F.; Alhasani, R.H.; Biswas, L.; Akhtar, S.; Pan, L.; Moens, C.B.; Li, W.; Liu, M.; et al. Rpgrip1 Is Required for Rod Outer Segment Development and Ciliary Protein Trafficking in Zebrafish. Sci. Rep. 2017, 7, 16881. [Google Scholar] [CrossRef]
  113. Chaki, M.; Hoefele, J.; Allen, S.J.; Ramaswami, G.; Janssen, S.; Bergmann, C.; Heckenlively, J.R.; Otto, E.A.; Hildebrandt, F. Genotype-Phenotype Correlation in 440 Patients with NPHP-Related Ciliopathies. Kidney Int. 2011, 80, 1239–1245. [Google Scholar] [CrossRef]
  114. Liebau, M.C.; Höpker, K.; Müller, R.U.; Schmedding, I.; Zank, S.; Schairer, B.; Fabretti, F.; Höhne, M.; Bartram, M.P.; Dafinger, C.; et al. Nephrocystin-4 Regulates Pyk2-Induced Tyrosine Phosphorylation of Nephrocystin-1 to Control Targeting to Monocilia. J. Biol. Chem. 2011, 286, 14237–14245. [Google Scholar] [CrossRef] [PubMed]
  115. Choi, Y.J.; Laclef, C.; Yang, N.; Andreu-Cervera, A.; Lewis, J.; Mao, X.; Li, L.; Snedecor, E.R.; Takemaru, K.-I.; Qin, C.; et al. RPGRIP1L Is Required for Stabilizing Epidermal Keratinocyte Adhesion through Regulating Desmoglein Endocytosis. PLoS Genet. 2019, 15, e1007914. [Google Scholar] [CrossRef] [PubMed]
  116. Mahuzier, A.; Gaudé, H.-M.; Grampa, V.; Anselme, I.; Silbermann, F.; Leroux-Berger, M.; Delacour, D.; Ezan, J.; Montcouquiol, M.; Saunier, S.; et al. Dishevelled Stabilization by the Ciliopathy Protein Rpgrip1l Is Essential for Planar Cell Polarity. J. Cell Biol. 2012, 198, 927–940. [Google Scholar] [CrossRef]
  117. Schäfer, T.; Pütz, M.; Lienkamp, S.; Ganner, A.; Bergbreiter, A.; Ramachandran, H.; Gieloff, V.; Gerner, M.; Mattonet, C.; Czarnecki, P.G.; et al. Genetic and Physical Interaction between the NPHP5 and NPHP6 Gene Products. Hum. Mol. Genet. 2008, 17, 3655–3662. [Google Scholar] [CrossRef]
  118. Kim, Y.J.; Kim, S.; Jung, Y.; Jung, E.; Kwon, H.J.; Kim, J. Eupatilin Rescues Ciliary Transition Zone Defects to Ameliorate Ciliopathy-Related Phenotypes. J. Clin. Investig. 2018, 128, 3642–3648. [Google Scholar] [CrossRef]
  119. Tian, X.; Zhao, H.; Zhou, J. Organization, Functions, and Mechanisms of the BBSome in Development, Ciliopathies, and Beyond. eLife 2023, 12, e87623. [Google Scholar] [CrossRef]
  120. Zuo, X.; Guo, W.; Lipschutz, J.H. The Exocyst Protein Sec10 Is Necessary for Primary Ciliogenesis and Cystogenesis in Vitro. Mol. Biol. Cell 2009, 20, 2522–2529. [Google Scholar] [CrossRef]
  121. Shimada, H.; Lu, Q.; Insinna-Kettenhofen, C.; Nagashima, K.; English, M.A.; Semler, E.M.; Mahgerefteh, J.; Cideciyan, A.V.; Li, T.; Brooks, B.P.; et al. In Vitro Modeling Using Ciliopathy-Patient-Derived Cells Reveals Distinct Cilia Dysfunctions Caused by CEP290 Mutations. Cell Rep. 2017, 20, 384–396. [Google Scholar] [CrossRef]
  122. Wiegering, A.; Dildrop, R.; Vesque, C.; Khanna, H.; Schneider-Maunoury, S.; Gerhardt, C. Rpgrip1l Controls Ciliary Gating by Ensuring the Proper Amount of Cep290 at the Vertebrate Transition Zone. Mol. Biol. Cell 2021, 32, 675–689. [Google Scholar] [CrossRef]
  123. Casteels, I.; Spileers, W.; Demaerel, P.; Casaer, P.; De Cock, P.; Dralands, L.; Missotten, L. Leber Congenital Amaurosis--Differential Diagnosis, Ophthalmological and Neuroradiological Report of 18 Patients. Neuropediatrics 1996, 27, 189–193. [Google Scholar] [CrossRef]
  124. Hildebrandt, F.; Waldherr, R.; Kutt, R.; Brandis, M. The Nephronophthisis Complex: Clinical and Genetic Aspects. Clin. Investig. 1992, 70, 802–808. [Google Scholar] [CrossRef] [PubMed]
  125. Katagiri, S.; Hayashi, T.; Yoshitake, K.; Murai, N.; Matsui, Z.; Kubo, H.; Satoh, H.; Matsufuji, S.; Takamura, T.; Yokoo, T.; et al. Compound Heterozygous Splice Site Variants in the SCLT1 Gene Highlight an Additional Candidate Locus for Senior-Løken Syndrome. Sci. Rep. 2018, 8, 16733. [Google Scholar] [CrossRef]
  126. Kuzinska, M.Z.; Lin, S.Y.-Y.; Klämbt, V.; Bufler, P.; Rezvani, M. Ciliopathy Organoid Models: A Comprehensive Review. Am. J. Physiol. Cell Physiol. 2024, 327, C1604–C1625. [Google Scholar] [CrossRef]
  127. Alzarka, B.; Charnaya, O.; Gunay-Aygun, M. Diseases of the Primary Cilia: A Clinical Characteristics Review. Pediatr. Nephrol. 2025, 40, 611–627. [Google Scholar] [CrossRef]
  128. Romani, M.; Micalizzi, A.; Valente, E.M. Joubert Syndrome: Congenital Cerebellar Ataxia with the Molar Tooth. Lancet Neurol. 2013, 12, 894–905. [Google Scholar] [CrossRef]
  129. Bergmann, C. Educational Paper: Ciliopathies. Eur. J. Pediatr. 2012, 171, 1285–1300. [Google Scholar] [CrossRef]
  130. Garg, S.; Rakesh, K.; Khattri, S.; Mishra, P.; Tikka, S.K. Bipolar Disorder with Intellectual Disability in Joubert Syndrome: A Case Report. Psychiatry Clin. Neurosci. 2019, 73, 761–762. [Google Scholar] [CrossRef]
  131. Paprocka, J.; Jamroz, E. Joubert Syndrome and Related Disorders. Neurol. Neurochir. Pol. 2012, 46, 379–383. [Google Scholar] [CrossRef]
  132. Parisi, M.; Glass, I. Joubert Syndrome. In GeneReviews®; Adam, M.P., Mirzaa, G.M., Pagon, R.A., Wallace, S.E., Bean, L.J., Gripp, K.W., Amemiya, A., Eds.; University of Washington: Seattle, WA, USA, 1993. [Google Scholar]
  133. Van De Weghe, J.C.; Gomez, A.; Doherty, D. The Joubert-Meckel-Nephronophthisis Spectrum of Ciliopathies. Annu. Rev. Genom. Hum. Genet. 2022, 23, 301–329. [Google Scholar] [CrossRef]
  134. Chen, C.-P. Meckel Syndrome: Genetics, Perinatal Findings, and Differential Diagnosis. Taiwan J. Obstet. Gynecol. 2007, 46, 9–14. [Google Scholar] [CrossRef]
  135. Forsyth, R.; Gunay-Aygun, M. Bardet-Biedl Syndrome Overview. In GeneReviews®; Adam, M.P., Mirzaa, G.M., Pagon, R.A., Wallace, S.E., Bean, L.J., Gripp, K.W., Amemiya, A., Eds.; University of Washington: Seattle, WA, USA, 1993. [Google Scholar]
  136. Forsythe, E.; Beales, P.L. Bardet-Biedl Syndrome. Eur. J. Hum. Genet. 2013, 21, 8–13. [Google Scholar] [CrossRef] [PubMed]
  137. Gurrieri, F.; Franco, B.; Toriello, H.; Neri, G. Oral-Facial-Digital Syndromes: Review and Diagnostic Guidelines. Am. J. Med. Genet. A 2007, 143A, 3314–3323. [Google Scholar] [CrossRef] [PubMed]
  138. Bruel, A.-L.; Levy, J.; Elenga, N.; Defo, A.; Favre, A.; Lucron, H.; Capri, Y.; Perrin, L.; Passemard, S.; Vial, Y.; et al. INTU-Related Oral-Facial-Digital Syndrome Type VI: A Confirmatory Report. Clin. Genet. 2018, 93, 1205–1209. [Google Scholar] [CrossRef]
  139. Franco, B.; Bruel, A.-L.; Thauvin-Robinet, C. Oral-Facial-Digital Syndrome Type I. In GeneReviews®; Adam, M.P., Mirzaa, G.M., Pagon, R.A., Wallace, S.E., Bean, L.J., Gripp, K.W., Amemiya, A., Eds.; University of Washington: Seattle, WA, USA, 1993. [Google Scholar]
  140. Tsang, S.H.; Aycinena, A.R.P.; Sharma, T. Ciliopathy: Alström Syndrome. In Atlas of Inherited Retinal Diseases; Tsang, S.H., Sharma, T., Eds.; Advances in Experimental Medicine and Biology; Springer International Publishing: Cham, Switzerland, 2018; pp. 179–180. ISBN 978-3-319-95046-4. [Google Scholar]
  141. Dassie, F.; Favaretto, F.; Bettini, S.; Parolin, M.; Valenti, M.; Reschke, F.; Danne, T.; Vettor, R.; Milan, G.; Maffei, P. Alström Syndrome: An Ultra-Rare Monogenic Disorder as a Model for Insulin Resistance, Type 2 Diabetes Mellitus and Obesity. Endocrine 2021, 71, 618–625. [Google Scholar] [CrossRef]
  142. Dassie, F.; Lorusso, R.; Benavides-Varela, S.; Milan, G.; Favaretto, F.; Callus, E.; Cagnin, S.; Reggiani, F.; Minervini, G.; Tosatto, S.; et al. Neurocognitive Assessment and DNA Sequencing Expand the Phenotype and Genotype Spectrum of Alström Syndrome. Am. J. Med. Genet. Part A 2021, 185, 732–742. [Google Scholar] [CrossRef]
  143. Marshall, J.D.; Bronson, R.T.; Collin, G.B.; Nordstrom, A.D.; Maffei, P.; Paisey, R.B.; Carey, C.; MacDermott, S.; Russell-Eggitt, I.; Shea, S.E.; et al. New Alström Syndrome Phenotypes Based on the Evaluation of 182 Cases. Arch. Intern. Med. 2005, 165, 675–683. [Google Scholar] [CrossRef]
  144. Kanna, R.M.; Gradil, D.; Boszczyk, B.M. Management of Cervical Myelopathy Due to Ossification of Posterior Longitudinal Ligament in a Patient with Alström Syndrome. Eur. Spine J. 2012, 21, 2418–2424. [Google Scholar] [CrossRef]
  145. Fuster-García, C.; García-Bohórquez, B.; Rodríguez-Muñoz, A.; Aller, E.; Jaijo, T.; Millán, J.M.; García-García, G. Usher Syndrome: Genetics of a Human Ciliopathy. Int. J. Mol. Sci. 2021, 22, 6723. [Google Scholar] [CrossRef]
  146. Dammeyer, J. Children with Usher Syndrome: Mental and Behavioral Disorders. Behav. Brain Funct. 2012, 8, 16. [Google Scholar] [CrossRef]
  147. Wang, H.; Huo, L.; Wang, Y.; Sun, W.; Gu, W. Usher Syndrome Type 2A Complicated with Glycogen Storage Disease Type 3 Due to Paternal Uniparental Isodisomy of Chromosome 1 in a Sporadic Patient. Mol. Genet. Genom. Med. 2021, 9, e1779. [Google Scholar] [CrossRef]
  148. Mill, P.; Christensen, S.T.; Pedersen, L.B. Primary Cilia as Dynamic and Diverse Signalling Hubs in Development and Disease. Nat. Rev. Genet. 2023, 24, 421–441. [Google Scholar] [CrossRef] [PubMed]
  149. Zheng, N.-X.; Miao, Y.-T.; Zhang, X.; Huang, M.-Z.; Jahangir, M.; Luo, S.; Lang, B. Primary Cilia-Associated Protein IFT172 in Ciliopathies. Front. Cell Dev. Biol. 2023, 11, 1074880. [Google Scholar] [CrossRef] [PubMed]
  150. Hoffmann, F.; Bolz, S.; Junger, K.; Klose, F.; Schubert, T.; Woerz, F.; Boldt, K.; Ueffing, M.; Beyer, T. TTC30A and TTC30B Redundancy Protects IFT Complex B Integrity and Its Pivotal Role in Ciliogenesis. Genes 2022, 13, 1191. [Google Scholar] [CrossRef] [PubMed]
  151. Cilleros-Rodriguez, D.; Martin-Morales, R.; Barbeito, P.; Deb Roy, A.; Loukil, A.; Sierra-Rodero, B.; Herranz, G.; Pampliega, O.; Redrejo-Rodriguez, M.; Goetz, S.C.; et al. Multiple Ciliary Localization Signals Control INPP5E Ciliary Targeting. eLife 2022, 11, e78383. [Google Scholar] [CrossRef]
  152. He, R.; Chen, G.; Li, Z.; Li, J. Structure of the N-Terminal Coiled-Coil Domains of the Ciliary Protein Rpgrip1l. iScience 2023, 26, 106249. [Google Scholar] [CrossRef]
  153. Mitchison, H.M.; Valente, E.M. Motile and Non-Motile Cilia in Human Pathology: From Function to Phenotypes. J. Pathol. 2017, 241, 294–309. [Google Scholar] [CrossRef]
  154. Bachmann-Gagescu, R.; Dempsey, J.C.; Phelps, I.G.; O’Roak, B.J.; Knutzen, D.M.; Rue, T.C.; Ishak, G.E.; Isabella, C.R.; Gorden, N.; Adkins, J.; et al. Joubert Syndrome: A Model for Untangling Recessive Disorders with Extreme Genetic Heterogeneity. J. Med. Genet. 2015, 52, 514–522. [Google Scholar] [CrossRef]
  155. Walczak-Sztulpa, J.; Posmyk, R.; Bukowska-Olech, E.M.; Wawrocka, A.; Jamsheer, A.; Oud, M.M.; Schmidts, M.; Arts, H.H.; Latos-Bielenska, A.; Wasilewska, A. Compound Heterozygous IFT140 Variants in Two Polish Families with Sensenbrenner Syndrome and Early Onset End-Stage Renal Disease. Orphanet J. Rare Dis. 2020, 15, 36. [Google Scholar] [CrossRef]
  156. Ashe, A.; Butterfield, N.C.; Town, L.; Courtney, A.D.; Cooper, A.N.; Ferguson, C.; Barry, R.; Olsson, F.; Liem, K.F.; Parton, R.G.; et al. Mutations in Mouse Ift144 Model the Craniofacial, Limb and Rib Defects in Skeletal Ciliopathies. Hum. Mol. Genet. 2012, 21, 1808–1823. [Google Scholar] [CrossRef]
  157. Ishida, Y.; Kobayashi, T.; Chiba, S.; Katoh, Y.; Nakayama, K. Molecular Basis of Ciliary Defects Caused by Compound Heterozygous IFT144/WDR19 Mutations Found in Cranioectodermal Dysplasia. Hum. Mol. Genet. 2021, 30, 213–225. [Google Scholar] [CrossRef]
  158. Keith, B.P.; Robertson, D.L.; Hentges, K.E. Locus Heterogeneity Disease Genes Encode Proteins with High Interconnectivity in the Human Protein Interaction Network. Front. Genet. 2014, 5, 434. [Google Scholar] [CrossRef] [PubMed]
  159. Hildebrandt, F.; Benzing, T.; Katsanis, N. Ciliopathies. N. Engl. J. Med. 2011, 364, 1533–1543. [Google Scholar] [CrossRef]
  160. McConnachie, D.J.; Stow, J.L.; Mallett, A.J. Ciliopathies and the Kidney: A Review. Am. J. Kidney Dis. 2021, 77, 410–419. [Google Scholar] [CrossRef] [PubMed]
  161. Littink, K.W.; Pott, J.-W.R.; Collin, R.W.J.; Kroes, H.Y.; Verheij, J.B.G.M.; Blokland, E.A.W.; de Castro Miró, M.; Hoyng, C.B.; Klaver, C.C.W.; Koenekoop, R.K.; et al. A Novel Nonsense Mutation in CEP290 Induces Exon Skipping and Leads to a Relatively Mild Retinal Phenotype. Investig. Ophthalmol. Vis. Sci. 2010, 51, 3646–3652. [Google Scholar] [CrossRef]
  162. Zhu, T.; Shen, Y.; Sun, Z.; Han, X.; Wei, X.; Li, W.; Lu, C.; Cheng, T.; Zou, X.; Li, H.; et al. Clinical and Molecular Features of a Chinese Cohort With Syndromic and Nonsyndromic Retinal Dystrophies Related to the CEP290 Gene. Am. J. Ophthalmol. 2023, 248, 96–106. [Google Scholar] [CrossRef]
  163. Baala, L.; Audollent, S.; Martinovic, J.; Ozilou, C.; Babron, M.-C.; Sivanandamoorthy, S.; Saunier, S.; Salomon, R.; Gonzales, M.; Rattenberry, E.; et al. Pleiotropic Effects of CEP290 (NPHP6) Mutations Extend to Meckel Syndrome. Am. J. Hum. Genet. 2007, 81, 170–179. [Google Scholar] [CrossRef]
  164. Helou, J.; Otto, E.A.; Attanasio, M.; Allen, S.J.; Parisi, M.A.; Glass, I.; Utsch, B.; Hashmi, S.; Fazzi, E.; Omran, H.; et al. Mutation Analysis of NPHP6/CEP290 in Patients with Joubert Syndrome and Senior-Løken Syndrome. J. Med. Genet. 2007, 44, 657–663. [Google Scholar] [CrossRef]
  165. Williamson, M.; Traboulsi, E.; DeBenedictis, M. Investigation of CEP290 Genotype-Phenotype Correlations in a Patient with Retinitis Pigmentosa, Infertility, End-Stage Renal Disease, and a Novel Mutation. Ophthalmic Genet. 2020, 41, 171–174. [Google Scholar] [CrossRef]
  166. Reiter, J.F.; Leroux, M.R. Genes and Molecular Pathways Underpinning Ciliopathies. Nat. Rev. Mol. Cell Biol. 2017, 18, 533–547. [Google Scholar] [CrossRef]
  167. Kousi, M.; Katsanis, N. Genetic Modifiers and Oligogenic Inheritance. Cold Spring Harb. Perspect. Med. 2015, 5, a017145. [Google Scholar] [CrossRef]
  168. Khanna, H.; Davis, E.E.; Murga-Zamalloa, C.A.; Estrada-Cuzcano, A.; Lopez, I.; den Hollander, A.I.; Zonneveld, M.N.; Othman, M.I.; Waseem, N.; Chakarova, C.F.; et al. A Common Allele in RPGRIP1L Is a Modifier of Retinal Degeneration in Ciliopathies. Nat. Genet. 2009, 41, 739–745. [Google Scholar] [CrossRef] [PubMed]
  169. Louie, C.M.; Caridi, G.; Lopes, V.S.; Brancati, F.; Kispert, A.; Lancaster, M.A.; Schlossman, A.M.; Otto, E.A.; Leitges, M.; Groene, H.-J.; et al. AHI1 Is Required for Outer Segment Development and Is a Modifier for Retinal Degeneration in Nephronophthisis. Nat. Genet. 2010, 42, 175–180. [Google Scholar] [CrossRef]
  170. Perea-Romero, I.; Solarat, C.; Blanco-Kelly, F.; Sanchez-Navarro, I.; Bea-Mascato, B.; Martin-Salazar, E.; Lorda-Sanchez, I.; Swafiri, S.T.; Avila-Fernandez, A.; Martin-Merida, I.; et al. Allelic Overload and Its Clinical Modifier Effect in Bardet-Biedl Syndrome. NPJ Genom. Med. 2022, 7, 41. [Google Scholar] [CrossRef]
  171. Ramsbottom, S.A.; Thelwall, P.E.; Wood, K.M.; Clowry, G.J.; Devlin, L.A.; Silbermann, F.; Spiewak, H.L.; Shril, S.; Molinari, E.; Hildebrandt, F.; et al. Mouse Genetics Reveals Barttin as a Genetic Modifier of Joubert Syndrome. Proc. Natl. Acad. Sci. USA 2020, 117, 1113–1118. [Google Scholar] [CrossRef]
  172. Leggatt, G.P.; Seaby, E.G.; Veighey, K.; Gast, C.; Gilbert, R.D.; Ennis, S. A Role for Genetic Modifiers in Tubulointerstitial Kidney Diseases. Genes 2023, 14, 1582. [Google Scholar] [CrossRef]
  173. Deltas, C. Digenic Inheritance and Genetic Modifiers. Clin. Genet. 2018, 93, 429–438. [Google Scholar] [CrossRef]
  174. Beales, P.L.; Badano, J.L.; Ross, A.J.; Ansley, S.J.; Hoskins, B.E.; Kirsten, B.; Mein, C.A.; Froguel, P.; Scambler, P.J.; Lewis, R.A.; et al. Genetic Interaction of BBS1 Mutations with Alleles at Other BBS Loci Can Result in Non-Mendelian Bardet-Biedl Syndrome. Am. J. Hum. Genet. 2003, 72, 1187–1199. [Google Scholar] [CrossRef]
  175. Katsanis, N.; Ansley, S.J.; Badano, J.L.; Eichers, E.R.; Lewis, R.A.; Hoskins, B.E.; Scambler, P.J.; Davidson, W.S.; Beales, P.L.; Lupski, J.R. Triallelic Inheritance in Bardet-Biedl Syndrome, a Mendelian Recessive Disorder. Science 2001, 293, 2256–2259. [Google Scholar] [CrossRef]
  176. Hoefele, J.; Wolf, M.T.F.; O’Toole, J.F.; Otto, E.A.; Schultheiss, U.; Dêschenes, G.; Attanasio, M.; Utsch, B.; Antignac, C.; Hildebrandt, F. Evidence of Oligogenic Inheritance in Nephronophthisis. J. Am. Soc. Nephrol. 2007, 18, 2789–2795. [Google Scholar] [CrossRef]
  177. Manara, E.; Paolacci, S.; D’Esposito, F.; Abeshi, A.; Ziccardi, L.; Falsini, B.; Colombo, L.; Iarossi, G.; Pilotta, A.; Boccone, L.; et al. Mutation Profile of BBS Genes in Patients with Bardet-Biedl Syndrome: An Italian Study. Ital. J. Pediatr. 2019, 45, 72. [Google Scholar] [CrossRef]
  178. Pollara, L.; Sottile, V.; Valente, E.M. Patient-Derived Cellular Models of Primary Ciliopathies. J. Med. Genet. 2022, 59, 517–527. [Google Scholar] [CrossRef] [PubMed]
  179. Zaki, M.S.; Sattar, S.; Massoudi, R.A.; Gleeson, J.G. Co-Occurrence of Distinct Ciliopathy Diseases in Single Families Suggests Genetic Modifiers. Am. J. Med. Genet. A 2011, 155, 3042–3049. [Google Scholar] [CrossRef]
  180. Shaheen, R.; Szymanska, K.; Basu, B.; Patel, N.; Ewida, N.; Faqeih, E.; Al Hashem, A.; Derar, N.; Alsharif, H.; Aldahmesh, M.A.; et al. Characterizing the Morbid Genome of Ciliopathies. Genome Biol. 2016, 17, 242. [Google Scholar] [CrossRef] [PubMed]
  181. Zaghloul, N.A.; Katsanis, N. Functional Modules, Mutational Load and Human Genetic Disease. Trends Genet. 2010, 26, 168–176. [Google Scholar] [CrossRef]
  182. Fahim, A. Retinitis Pigmentosa: Recent Advances and Future Directions in Diagnosis and Management. Curr. Opin. Pediatr. 2018, 30, 725–733. [Google Scholar] [CrossRef]
  183. Robert, M.-A.; Chahal, P.S.; Audy, A.; Kamen, A.; Gilbert, R.; Gaillet, B. Manufacturing of Recombinant Adeno-Associated Viruses Using Mammalian Expression Platforms. Biotechnol. J. 2017, 12, 1600193. [Google Scholar] [CrossRef]
  184. Aguirre, G.D.; Cideciyan, A.V.; Dufour, V.L.; Ripolles-García, A.; Sudharsan, R.; Swider, M.; Nikonov, R.; Iwabe, S.; Boye, S.L.; Hauswirth, W.W.; et al. Gene Therapy Reforms Photoreceptor Structure and Restores Vision in NPHP5-Associated Leber Congenital Amaurosis. Mol. Ther. 2021, 29, 2456–2468. [Google Scholar] [CrossRef]
  185. Mookherjee, S.; Chen, H.Y.; Isgrig, K.; Yu, W.; Hiriyanna, S.; Levron, R.; Li, T.; Colosi, P.; Chien, W.; Swaroop, A.; et al. A CEP290 C-Terminal Domain Complements the Mutant CEP290 of Rd16 Mice In Trans and Rescues Retinal Degeneration. Cell Rep. 2018, 25, 611–623.e6. [Google Scholar] [CrossRef]
  186. Pierce, E.A.; Aleman, T.S.; Jayasundera, K.T.; Ashimatey, B.S.; Kim, K.; Rashid, A.; Jaskolka, M.C.; Myers, R.L.; Lam, B.L.; Bailey, S.T.; et al. Gene Editing for CEP290-Associated Retinal Degeneration. N Engl J Med 2024, 390, 1972–1984. [Google Scholar] [CrossRef]
  187. Zhao, Q.; Wei, L.; Chen, Y. From Bench to Bedside: Developing CRISPR/Cas-Based Therapy for Ocular Diseases. Pharmacol. Res. 2025, 213, 107638. [Google Scholar] [CrossRef]
  188. Maeder, M.L.; Stefanidakis, M.; Wilson, C.J.; Baral, R.; Barrera, L.A.; Bounoutas, G.S.; Bumcrot, D.; Chao, H.; Ciulla, D.M.; DaSilva, J.A.; et al. Development of a Gene-Editing Approach to Restore Vision Loss in Leber Congenital Amaurosis Type 10. Nat. Med. 2019, 25, 229–233. [Google Scholar] [CrossRef] [PubMed]
  189. Ruan, G.-X.; Barry, E.; Yu, D.; Lukason, M.; Cheng, S.H.; Scaria, A. CRISPR/Cas9-Mediated Genome Editing as a Therapeutic Approach for Leber Congenital Amaurosis 10. Mol Ther 2017, 25, 331–341. [Google Scholar] [CrossRef]
  190. Adams, D.; Gonzalez-Duarte, A.; O’Riordan, W.D.; Yang, C.-C.; Ueda, M.; Kristen, A.V.; Tournev, I.; Schmidt, H.H.; Coelho, T.; Berk, J.L.; et al. Patisiran, an RNAi Therapeutic, for Hereditary Transthyretin Amyloidosis. N. Engl. J. Med. 2018, 379, 11–21. [Google Scholar] [CrossRef]
  191. Rossor, A.M.; Reilly, M.M.; Sleigh, J.N. Antisense Oligonucleotides and Other Genetic Therapies Made Simple. Pract. Neurol. 2018, 18, 126–131. [Google Scholar] [CrossRef]
  192. Weng, Y.; Li, C.; Yang, T.; Hu, B.; Zhang, M.; Guo, S.; Xiao, H.; Liang, X.-J.; Huang, Y. The Challenge and Prospect of mRNA Therapeutics Landscape. Biotechnol. Adv. 2020, 40, 107534. [Google Scholar] [CrossRef]
  193. Garanto, A.; Chung, D.C.; Duijkers, L.; Corral-Serrano, J.C.; Messchaert, M.; Xiao, R.; Bennett, J.; Vandenberghe, L.H.; Collin, R.W.J. In Vitro and in Vivo Rescue of Aberrant Splicing in CEP290-Associated LCA by Antisense Oligonucleotide Delivery. Hum. Mol. Genet. 2016, 25, 2552–2563. [Google Scholar] [CrossRef]
  194. Parfitt, D.A.; Lane, A.; Ramsden, C.; Jovanovic, K.; Coffey, P.J.; Hardcastle, A.J.; Cheetham, M.E. Using Induced Pluripotent Stem Cells to Understand Retinal Ciliopathy Disease Mechanisms and Develop Therapies. Biochem. Soc. Trans. 2016, 44, 1245–1251. [Google Scholar] [CrossRef]
  195. Mathur, P.; Yang, J. Usher Syndrome: Hearing Loss, Retinal Degeneration and Associated Abnormalities. Biochim. Biophys. Acta 2015, 1852, 406–420. [Google Scholar] [CrossRef]
  196. Johansson, I.; Monsen, V.T.; Pettersen, K.; Mildenberger, J.; Misund, K.; Kaarniranta, K.; Schønberg, S.; Bjørkøy, G. The Marine N-3 PUFA DHA Evokes Cytoprotection against Oxidative Stress and Protein Misfolding by Inducing Autophagy and NFE2L2 in Human Retinal Pigment Epithelial Cells. Autophagy 2015, 11, 1636–1651. [Google Scholar] [CrossRef]
  197. Choi, E.H.; Daruwalla, A.; Suh, S.; Leinonen, H.; Palczewski, K. Retinoids in the Visual Cycle: Role of the Retinal G Protein-Coupled Receptor. J. Lipid Res. 2021, 62, 100040. [Google Scholar] [CrossRef]
  198. Guadagni, V.; Novelli, E.; Piano, I.; Gargini, C.; Strettoi, E. Pharmacological Approaches to Retinitis Pigmentosa: A Laboratory Perspective. Prog. Retin. Eye Res. 2015, 48, 62–81. [Google Scholar] [CrossRef] [PubMed]
  199. Lipinski, D.M.; Barnard, A.R.; Singh, M.S.; Martin, C.; Lee, E.J.; Davies, W.I.L.; MacLaren, R.E. CNTF Gene Therapy Confers Lifelong Neuroprotection in a Mouse Model of Human Retinitis Pigmentosa. Mol. Ther. 2015, 23, 1308–1319. [Google Scholar] [CrossRef]
  200. Dias, M.F.; Joo, K.; Kemp, J.A.; Fialho, S.L.; da Silva Cunha, A.; Woo, S.J.; Kwon, Y.J. Molecular Genetics and Emerging Therapies for Retinitis Pigmentosa: Basic Research and Clinical Perspectives. Prog. Retin. Eye Res. 2018, 63, 107–131. [Google Scholar] [CrossRef]
  201. Athanasiou, D.; Afanasyeva, T.A.V.; Chai, N.; Ziaka, K.; Jovanovic, K.; Guarascio, R.; Boldt, K.; Corral-Serrano, J.C.; Kanuga, N.; Roepman, R.; et al. Small Molecule Treatment Alleviates Photoreceptor Cilia Defects in LCA5-Deficient Human Retinal Organoids. Acta Neuropathol. Commun. 2025, 13, 26. [Google Scholar] [CrossRef]
  202. Smith, C.E.L.; Streets, A.J.; Lake, A.V.R.; Natarajan, S.; Best, S.K.; Szymanska, K.; Karwatka, M.; Stevenson, T.; Trowbridge, R.; Grant, G.; et al. Drug and siRNA Screens Identify ROCK2 as a Therapeutic Target for Ciliopathies. Commun. Med. 2025, 5, 129. [Google Scholar] [CrossRef]
  203. Howden, S.E.; Thomson, J.A.; Little, M.H. Simultaneous Reprogramming and Gene Editing of Human Fibroblasts. Nat. Protoc. 2018, 13, 875–898. [Google Scholar] [CrossRef]
  204. Gholkar, A.A.; Gimeno, T.V.; Edgemon, J.E.; Sim, M.S.; Torres, J.Z. MI-181 Modulates Cilia Length and Restores Cilia Length in Cells with Defective Shortened Cilia. ACS Chem. Biol. 2024, 19, 1733–1742. [Google Scholar] [CrossRef]
  205. Stern, J.H.; Tian, Y.; Funderburgh, J.; Pellegrini, G.; Zhang, K.; Goldberg, J.L.; Ali, R.R.; Young, M.; Xie, Y.; Temple, S. Regenerating Eye Tissues to Preserve and Restore Vision. Cell Stem Cell 2018, 22, 834–849. [Google Scholar] [CrossRef]
  206. Garita-Hernandez, M.; Lampič, M.; Chaffiol, A.; Guibbal, L.; Routet, F.; Santos-Ferreira, T.; Gasparini, S.; Borsch, O.; Gagliardi, G.; Reichman, S.; et al. Restoration of Visual Function by Transplantation of Optogenetically Engineered Photoreceptors. Nat. Commun. 2019, 10, 4524. [Google Scholar] [CrossRef]
  207. Pansare, V.; Hejazi, S.; Faenza, W.; Prud’homme, R.K. Review of Long-Wavelength Optical and NIR Imaging Materials: Contrast Agents, Fluorophores and Multifunctional Nano Carriers. Chem. Mater. 2012, 24, 812–827. [Google Scholar] [CrossRef]
  208. Jones, M.K.; Lu, B.; Girman, S.; Wang, S. Cell-Based Therapeutic Strategies for Replacement and Preservation in Retinal Degenerative Diseases. Prog. Retin. Eye Res. 2017, 58, 1–27. [Google Scholar] [CrossRef] [PubMed]
  209. da Cruz, L.; Fynes, K.; Georgiadis, O.; Kerby, J.; Luo, Y.H.; Ahmado, A.; Vernon, A.; Daniels, J.T.; Nommiste, B.; Hasan, S.M.; et al. Phase 1 Clinical Study of an Embryonic Stem Cell-Derived Retinal Pigment Epithelium Patch in Age-Related Macular Degeneration. Nat. Biotechnol. 2018, 36, 328–337. [Google Scholar] [CrossRef] [PubMed]
  210. Yue, L.; Weiland, J.D.; Roska, B.; Humayun, M.S. Retinal Stimulation Strategies to Restore Vision: Fundamentals and Systems. Prog. Retin. Eye Res. 2016, 53, 21–47. [Google Scholar] [CrossRef] [PubMed]
  211. Winter, J.O.; Cogan, S.F.; Rizzo, J.F. Retinal Prostheses: Current Challenges and Future Outlook. J. Biomater. Sci. Polym. Ed. 2007, 18, 1031–1055. [Google Scholar] [CrossRef]
  212. Cardenas-Rodriguez, M.; Austin-Tse, C.; Bergboer, J.G.M.; Molinari, E.; Sugano, Y.; Bachmann-Gagescu, R.; Sayer, J.A.; Drummond, I.A. Genetic Compensation for Cilia Defects in Cep290 Mutants by Upregulation of Cilia-Associated Small GTPases. J. Cell Sci. 2021, 134, jcs258568. [Google Scholar] [CrossRef]
  213. Zhang, K.; Da Silva, F.; Seidl, C.; Wilsch-Bräuninger, M.; Herbst, J.; Huttner, W.B.; Niehrs, C. Primary Cilia Are WNT-Transducing Organelles Whose Biogenesis Is Controlled by a WNT-PP1 Axis. Dev. Cell 2023, 58, 139–154.e8. [Google Scholar] [CrossRef]
  214. Ren, Z.-L.; Zhang, H.-B.; Li, L.; Yang, Z.-L.; Jiang, L. Characterization of Two Novel Knock-in Mouse Models of Syndromic Retinal Ciliopathy Carrying Hypomorphic Sdccag8 Mutations. Zool. Res. 2022, 43, 442–456. [Google Scholar] [CrossRef]
  215. Drivas, T.G.; Holzbaur, E.L.F.; Bennett, J. Disruption of CEP290 Microtubule/Membrane-Binding Domains Causes Retinal Degeneration. J. Clin. Investig. 2013, 123, 4525–4539. [Google Scholar] [CrossRef]
  216. Jiang, S.-T.; Chiou, Y.-Y.; Wang, E.; Lin, H.-K.; Lee, S.-P.; Lu, H.-Y.; Wang, C.-K.L.; Tang, M.-J.; Li, H. Targeted Disruption of Nphp1 Causes Male Infertility Due to Defects in the Later Steps of Sperm Morphogenesis in Mice. Hum. Mol. Genet. 2008, 17, 3368–3379. [Google Scholar] [CrossRef]
  217. O’Toole, J.F.; Otto, E.A.; Frishberg, Y.; Hildebrandt, F. Retinitis Pigmentosa and Renal Failure in a Patient with Mutations in INVS. Nephrol. Dial. Transplant. 2006, 21, 1989–1991. [Google Scholar] [CrossRef]
  218. Vogel, P.; Gelfman, C.M.; Issa, T.; Payne, B.J.; Hansen, G.M.; Read, R.W.; Jones, C.; Pitcher, M.R.; Ding, Z.-M.; DaCosta, C.M.; et al. Nephronophthisis and Retinal Degeneration in Tmem218-/- Mice: A Novel Mouse Model for Senior-Løken Syndrome? Vet. Pathol. 2015, 52, 580–595. [Google Scholar] [CrossRef] [PubMed]
  219. Hanke-Gogokhia, C.; Chiodo, V.A.; Hauswirth, W.W.; Frederick, J.M.; Baehr, W. Rescue of Cone Function in Cone-Only Nphp5 Knockout Mouse Model with Leber Congenital Amaurosis Phenotype. Mol. Vis. 2018, 24, 834–846. [Google Scholar] [PubMed]
  220. Oh, A.; Pearce, J.W.; Gandolfi, B.; Creighton, E.K.; Suedmeyer, W.K.; Selig, M.; Bosiack, A.P.; Castaner, L.J.; Whiting, R.E.H.; Belknap, E.B.; et al. Early-Onset Progressive Retinal Atrophy Associated with an IQCB1 Variant in African Black-Footed Cats (Felis Nigripes). Sci. Rep. 2017, 7, 43918. [Google Scholar] [CrossRef] [PubMed]
  221. Narfström, K. Hereditary Progressive Retinal Atrophy in the Abyssinian Cat. J. Hered. 1983, 74, 273–276. [Google Scholar] [CrossRef]
  222. May-Simera, H.L.; Wan, Q.; Jha, B.S.; Hartford, J.; Khristov, V.; Dejene, R.; Chang, J.; Patnaik, S.; Lu, Q.; Banerjee, P.; et al. Primary Cilium-Mediated Retinal Pigment Epithelium Maturation Is Disrupted in Ciliopathy Patient Cells. Cell Rep. 2018, 22, 189–205. [Google Scholar] [CrossRef]
Figure 1. The schematic diagram of primary cilium structure and function. Abbreviations: TZ, transition zone; TF, transition fibers.
Figure 1. The schematic diagram of primary cilium structure and function. Abbreviations: TZ, transition zone; TF, transition fibers.
Biomolecules 15 00667 g001
Figure 2. Schematic representation of the structure and function of primary cilium in photoreceptors.
Figure 2. Schematic representation of the structure and function of primary cilium in photoreceptors.
Biomolecules 15 00667 g002
Figure 3. Clinical overlap and distinctions between SLSN and other ciliopathies. The typicality and frequency of features in different syndromes are reflected through the gradation of three colors (the darkest means major/obligate features, the medium means minor/common features, and the lightest means possibly involved features), while some features not yet reported are left blank. Abbreviations: SLSN: Senior–Loken syndrome; JS: Joubert syndrome: MKS: Meckel syndrome; BBS: Bardet-Biedl syndrome; OFD: Oral-facial-digital syndrome; ALMS: Alström syndrome; USH: Usher syndrome; CNS: central nervous system; MTS: molar tooth sign; NPHP: nephronophthisis; DPM: ductal plate malformation. abn.: abnormalities. Reference: SLSN: [6,35,123,124,125]; JS: [35,128,129,130,131,132,133]; MKS: [128,129,132,134]; BBS: [35,128,129,135,136]; OFD: [137,138,139]; ALMS: [140,141,142,143,144]; USH: [145,146,147]; Other information comes from the Human Phenotype Ontology (HPO) database.
Figure 3. Clinical overlap and distinctions between SLSN and other ciliopathies. The typicality and frequency of features in different syndromes are reflected through the gradation of three colors (the darkest means major/obligate features, the medium means minor/common features, and the lightest means possibly involved features), while some features not yet reported are left blank. Abbreviations: SLSN: Senior–Loken syndrome; JS: Joubert syndrome: MKS: Meckel syndrome; BBS: Bardet-Biedl syndrome; OFD: Oral-facial-digital syndrome; ALMS: Alström syndrome; USH: Usher syndrome; CNS: central nervous system; MTS: molar tooth sign; NPHP: nephronophthisis; DPM: ductal plate malformation. abn.: abnormalities. Reference: SLSN: [6,35,123,124,125]; JS: [35,128,129,130,131,132,133]; MKS: [128,129,132,134]; BBS: [35,128,129,135,136]; OFD: [137,138,139]; ALMS: [140,141,142,143,144]; USH: [145,146,147]; Other information comes from the Human Phenotype Ontology (HPO) database.
Biomolecules 15 00667 g003
Table 1. Summary of the identified SLSN genes.
Table 1. Summary of the identified SLSN genes.
GeneMIM
Number
Chromosome LocationProteinProtein FunctionLocalization in CiliumPhenotype
NPHP1266900,
607100
2q13Nephrocystin-1Cell-cell and cell-matrix signalingTransition zone [41]NPHP, SLSN, JS, BBS [42,43,44]
NPHP3606995,
606995
3q22Nephrocystin-3Wnt signalingInversin compartments [45]NPHP, SLSN, MKS, BBS, JS, MSS, COACH, SRTD [46,47,48,49]
NPHP4606996,
607215
1p36.31Nephrocystin-4Wnt signalingBasal body and transition zone [41]NPHP, SLSN [50,51]
IQCB1609254,
609237
3q13.33IQ calmodulin-binding motif-containing protein-1Ciliogenesis, regulation of BBSome complex integrityBasal body [27]LCA, NPHP, SLSN, JS [2,52,53]
CEP290610189,
610142
12q21.32Centrosomal protein of 290 kDaCiliogenesis, regulation of BBSome complex integrityTransition zone [27]LCA, NPHP, SLSN, JS, MKS, BBS [54,55]
SDCCAG8613615,
613524
1q43-44Serologically defined colon cancer antigen 8Ciliogenesis, Hedgehog signalingBasal body [56]NPHP, SLSN, BBS [17,57]
WDR19616307,
608151
4p14WD repeat-containing protein 19Ciliary transport, assemblyIFT-A complex [58]NPHP, SLSN, ATD, Sensenbrenner syndrome, Caroli syndrome [58,59,60,61]
TRAF3IP1616629,
607380
2q37.3TRAF-3 interacting protein 1Cilia importIFT-B complex [62]NPHP, SLSN, SRPS, ATD [63,64]
Abbreviations: LCA: Leber congenital amaurosis; SLSN: Senior–Loken syndrome; JS: Joubert syndrome: MKS: Meckel syndrome; MSS: Mainzer-Saldino syndrome; BBS: Bardet-Biedl syndrome; NPHP: nephronophthisis; SRTD: short-rib thoracic dysplasia; ATD: asphyxiating thoracic dystrophy; SRPS: short-rib polydactyly syndrome.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zhou, D.; Zeng, Y.; Luo, W.; Leng, C.; Li, C. Senior–Loken Syndrome: Ocular Perspectives on Genetics, Pathogenesis, and Management. Biomolecules 2025, 15, 667. https://doi.org/10.3390/biom15050667

AMA Style

Zhou D, Zeng Y, Luo W, Leng C, Li C. Senior–Loken Syndrome: Ocular Perspectives on Genetics, Pathogenesis, and Management. Biomolecules. 2025; 15(5):667. https://doi.org/10.3390/biom15050667

Chicago/Turabian Style

Zhou, Di, Yi Zeng, Weihan Luo, Chenyang Leng, and Chen Li. 2025. "Senior–Loken Syndrome: Ocular Perspectives on Genetics, Pathogenesis, and Management" Biomolecules 15, no. 5: 667. https://doi.org/10.3390/biom15050667

APA Style

Zhou, D., Zeng, Y., Luo, W., Leng, C., & Li, C. (2025). Senior–Loken Syndrome: Ocular Perspectives on Genetics, Pathogenesis, and Management. Biomolecules, 15(5), 667. https://doi.org/10.3390/biom15050667

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop