Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.881
Filter
Add more filters

Publication year range
1.
Genes Dev ; 35(21-22): 1445-1460, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34711653

ABSTRACT

Joubert syndrome (JS) is a recessive ciliopathy in which all affected individuals have congenital cerebellar vermis hypoplasia. Here, we report that CEP120, a JS-associated protein involved in centriole biogenesis and cilia assembly, regulates timely neuronal differentiation and the departure of granule neuron progenitors (GNPs) from their germinal zone during cerebellar development. Our results show that depletion of Cep120 perturbs GNP cell cycle progression, resulting in a delay of cell cycle exit in vivo. To dissect the potential mechanism, we investigated the association between CEP120 interactome and the JS database and identified KIAA0753 (a JS-associated protein) as a CEP120-interacting protein. Surprisingly, we found that CEP120 recruits KIAA0753 to centrioles, and that loss of this interaction induces accumulation of GNPs in the germinal zone and impairs neuronal differentiation. Importantly, the replenishment of wild-type CEP120 rescues the above defects, whereas expression of JS-associated CEP120 mutants, which hinder KIAA0753 recruitment, does not. Together, our data reveal a close interplay between CEP120 and KIAA0753 for the germinal zone exit and timely neuronal differentiation of GNPs during cerebellar development, and mutations in CEP120 and KIAA0753 may participate in the heterotopia and cerebellar hypoplasia observed in JS patients.


Subject(s)
Centrioles , Kidney Diseases, Cystic , Abnormalities, Multiple , Cell Cycle , Cell Cycle Proteins/metabolism , Centrioles/genetics , Centrioles/metabolism , Cerebellum/abnormalities , Cerebellum/metabolism , Eye Abnormalities , Humans , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/metabolism , Microtubule-Associated Proteins , Retina/abnormalities
2.
Cell ; 150(3): 533-48, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22863007

ABSTRACT

Nephronophthisis-related ciliopathies (NPHP-RC) are degenerative recessive diseases that affect kidney, retina, and brain. Genetic defects in NPHP gene products that localize to cilia and centrosomes defined them as "ciliopathies." However, disease mechanisms remain poorly understood. Here, we identify by whole-exome resequencing, mutations of MRE11, ZNF423, and CEP164 as causing NPHP-RC. All three genes function within the DNA damage response (DDR) pathway. We demonstrate that, upon induced DNA damage, the NPHP-RC proteins ZNF423, CEP164, and NPHP10 colocalize to nuclear foci positive for TIP60, known to activate ATM at sites of DNA damage. We show that knockdown of CEP164 or ZNF423 causes sensitivity to DNA damaging agents and that cep164 knockdown in zebrafish results in dysregulated DDR and an NPHP-RC phenotype. Our findings link degenerative diseases of the kidney and retina, disorders of increasing prevalence, to mechanisms of DDR.


Subject(s)
DNA Damage , DNA-Binding Proteins/metabolism , Exome , Kidney Diseases, Cystic/genetics , Microtubule Proteins/metabolism , Animals , Cilia/metabolism , Gene Knockdown Techniques , Genes, Recessive , Humans , MRE11 Homologue Protein , Mice , Proteins , Signal Transduction , Zebrafish/embryology , Zebrafish/metabolism
3.
Hum Mol Genet ; 33(16): 1442-1453, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-38751342

ABSTRACT

Primary cilia are antenna-like structures protruding from the surface of various eukaryotic cells, and have distinct protein compositions in their membranes. This distinct protein composition is maintained by the presence of the transition zone (TZ) at the ciliary base, which acts as a diffusion barrier between the ciliary and plasma membranes. Defects in cilia and the TZ are known to cause a group of disorders collectively called the ciliopathies, which demonstrate a broad spectrum of clinical features, such as perinatally lethal Meckel syndrome (MKS), relatively mild Joubert syndrome (JBTS), and nonsyndromic nephronophthisis (NPHP). Proteins constituting the TZ can be grouped into the MKS and NPHP modules. The MKS module is composed of several transmembrane proteins and three soluble proteins. TMEM218 was recently reported to be mutated in individuals diagnosed as MKS and JBTS. However, little is known about how TMEM218 mutations found in MKS and JBTS affect the functions of cilia. In this study, we found that ciliary membrane proteins were not localized to cilia in TMEM218-knockout cells, indicating impaired barrier function of the TZ. Furthermore, the exogenous expression of JBTS-associated TMEM218 variants but not MKS-associated variants in TMEM218-knockout cells restored the localization of ciliary membrane proteins. In particular, when expressed in TMEM218-knockout cells, the TMEM218(R115H) variant found in JBTS was able to restore the barrier function of cells, whereas the MKS variant TMEM218(R115C) could not. Thus, the severity of symptoms of MKS and JBTS individuals appears to correlate with the degree of their ciliary defects at the cellular level.


Subject(s)
Abnormalities, Multiple , Cilia , Ciliopathies , Encephalocele , Eye Abnormalities , Kidney Diseases, Cystic , Membrane Proteins , Mutation , Retina , Cilia/metabolism , Cilia/genetics , Cilia/pathology , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Ciliopathies/genetics , Ciliopathies/metabolism , Ciliopathies/pathology , Encephalocele/genetics , Encephalocele/metabolism , Encephalocele/pathology , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/metabolism , Kidney Diseases, Cystic/pathology , Abnormalities, Multiple/genetics , Abnormalities, Multiple/metabolism , Abnormalities, Multiple/pathology , Eye Abnormalities/genetics , Eye Abnormalities/pathology , Eye Abnormalities/metabolism , Retina/metabolism , Retina/abnormalities , Retina/pathology , Cerebellum/abnormalities , Cerebellum/metabolism , Cerebellum/pathology , Cerebellar Diseases/genetics , Cerebellar Diseases/metabolism , Cerebellar Diseases/pathology , Animals , Cell Membrane/metabolism , Mice , Ciliary Motility Disorders , Polycystic Kidney Diseases , Retinitis Pigmentosa
4.
J Cell Sci ; 137(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38841887

ABSTRACT

Centrosomal proteins play pivotal roles in orchestrating microtubule dynamics, and their dysregulation leads to disorders, including cancer and ciliopathies. Understanding the multifaceted roles of centrosomal proteins is vital to comprehend their involvement in disease development. Here, we report novel cellular functions of CEP41, a centrosomal and ciliary protein implicated in Joubert syndrome. We show that CEP41 is an essential microtubule-associated protein with microtubule-stabilizing activity. Purified CEP41 binds to preformed microtubules, promotes microtubule nucleation and suppresses microtubule disassembly. When overexpressed in cultured cells, CEP41 localizes to microtubules and promotes microtubule bundling. Conversely, shRNA-mediated knockdown of CEP41 disrupts the interphase microtubule network and delays microtubule reassembly, emphasizing its role in microtubule organization. Further, we demonstrate that the association of CEP41 with microtubules relies on its conserved rhodanese homology domain (RHOD) and the N-terminal region. Interestingly, a disease-causing mutation in the RHOD domain impairs CEP41-microtubule interaction. Moreover, depletion of CEP41 inhibits cell proliferation and disrupts cell cycle progression, suggesting its potential involvement in cell cycle regulation. These insights into the cellular functions of CEP41 hold promise for unraveling the impact of its mutations in ciliopathies.


Subject(s)
Cell Proliferation , Microtubules , Humans , Microtubules/metabolism , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Centrosome/metabolism , Retina/metabolism , Retina/pathology , Retina/abnormalities , Ciliopathies/metabolism , Ciliopathies/genetics , Ciliopathies/pathology , Cerebellum/metabolism , Cerebellum/abnormalities , Cerebellum/pathology , Kidney Diseases, Cystic/metabolism , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/pathology , Cilia/metabolism , Cilia/pathology , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Animals , Abnormalities, Multiple/metabolism , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Eye Abnormalities/metabolism , Eye Abnormalities/genetics , Eye Abnormalities/pathology , Protein Binding , Cell Cycle/genetics , HEK293 Cells
5.
Cell ; 145(4): 513-28, 2011 May 13.
Article in English | MEDLINE | ID: mdl-21565611

ABSTRACT

Nephronophthisis (NPHP), Joubert (JBTS), and Meckel-Gruber (MKS) syndromes are autosomal-recessive ciliopathies presenting with cystic kidneys, retinal degeneration, and cerebellar/neural tube malformation. Whether defects in kidney, retinal, or neural disease primarily involve ciliary, Hedgehog, or cell polarity pathways remains unclear. Using high-confidence proteomics, we identified 850 interactors copurifying with nine NPHP/JBTS/MKS proteins and discovered three connected modules: "NPHP1-4-8" functioning at the apical surface, "NPHP5-6" at centrosomes, and "MKS" linked to Hedgehog signaling. Assays for ciliogenesis and epithelial morphogenesis in 3D renal cultures link renal cystic disease to apical organization defects, whereas ciliary and Hedgehog pathway defects lead to retinal or neural deficits. Using 38 interactors as candidates, linkage and sequencing analysis of 250 patients identified ATXN10 and TCTN2 as new NPHP-JBTS genes, and our Tctn2 mouse knockout shows neural tube and Hedgehog signaling defects. Our study further illustrates the power of linking proteomic networks and human genetics to uncover critical disease pathways.


Subject(s)
Kidney Diseases, Cystic/genetics , Membrane Proteins/genetics , Signal Transduction , Animals , Ataxin-10 , Centrosome/metabolism , Cilia/metabolism , Ciliary Motility Disorders/genetics , Encephalocele/genetics , Hedgehog Proteins/metabolism , Humans , Kidney Diseases, Cystic/metabolism , Mice , NIH 3T3 Cells , Nerve Tissue Proteins/genetics , Polycystic Kidney Diseases/genetics , Retinitis Pigmentosa , Zebrafish
6.
Annu Rev Genomics Hum Genet ; 23: 301-329, 2022 08 31.
Article in English | MEDLINE | ID: mdl-35655331

ABSTRACT

The Joubert syndrome (JS), Meckel syndrome (MKS), and nephronophthisis (NPH) ciliopathy spectrum could be the poster child for advances and challenges in Mendelian human genetics over the past half century. Progress in understanding these conditions illustrates many core concepts of human genetics. The JS phenotype alone is caused by pathogenic variants in more than 40 genes; remarkably, all of the associated proteins function in and around the primary cilium. Primary cilia are near-ubiquitous, microtubule-based organelles that play crucial roles in development and homeostasis. Protruding from the cell, these cellular antennae sense diverse signals and mediate Hedgehog and other critical signaling pathways. Ciliary dysfunction causes many human conditions termed ciliopathies, which range from multiple congenital malformations to adult-onset single-organ failure. Research on the genetics of the JS-MKS-NPH spectrum has spurred extensive functional work exploring the broadly important role of primary cilia in health and disease. This functional work promises to illuminate the mechanisms underlying JS-MKS-NPH in humans, identify therapeutic targets across genetic causes, and generate future precision treatments.


Subject(s)
Abnormalities, Multiple , Ciliopathies , Eye Abnormalities , Polycystic Kidney Diseases , Abnormalities, Multiple/genetics , Abnormalities, Multiple/metabolism , Abnormalities, Multiple/pathology , Cerebellum/abnormalities , Cerebellum/metabolism , Cerebellum/pathology , Child , Cilia/genetics , Cilia/metabolism , Cilia/pathology , Ciliary Motility Disorders , Ciliopathies/genetics , Ciliopathies/metabolism , Ciliopathies/pathology , Encephalocele , Eye Abnormalities/genetics , Eye Abnormalities/metabolism , Eye Abnormalities/pathology , Hedgehog Proteins/metabolism , Humans , Kidney Diseases, Cystic , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/metabolism , Polycystic Kidney Diseases/pathology , Retina/abnormalities , Retina/metabolism , Retina/pathology , Retinitis Pigmentosa
7.
Proc Natl Acad Sci U S A ; 119(18): e2115960119, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35482924

ABSTRACT

Nephronophthisis (NPH) is an autosomal recessive tubulointerstitial nephropathy belonging to the ciliopathy disorders and known as the most common cause of hereditary end-stage renal disease in children. Yet, no curative treatment is available. The major gene, NPHP1, encodes a protein playing key functions at the primary cilium and cellular junctions. Using a medium-throughput drug-screen in NPHP1 knockdown cells, we identified 51 Food and Drug Administration-approved compounds by their ability to alleviate the cellular phenotypes associated with the loss of NPHP1; 11 compounds were further selected for their physicochemical properties. Among those compounds, prostaglandin E1 (PGE1) rescued ciliogenesis defects in immortalized patient NPHP1 urine-derived renal tubular cells, and improved ciliary and kidney phenotypes in our NPH zebrafish and Nphp1 knockout mouse models. Furthermore, Taprenepag, a nonprostanoid prostaglandin E2 receptor agonist, alleviated the severe retinopathy observed in Nphp1−/− mice. Finally, comparative transcriptomics allowed identification of key signaling pathways downstream PGE1, including cell cycle progression, extracellular matrix, adhesion, or actin cytoskeleton organization. In conclusion, using in vitro and in vivo models, we showed that prostaglandin E2 receptor agonists can ameliorate several of the pleotropic phenotypes caused by the absence of NPHP1; this opens their potential as a first therapeutic option for juvenile NPH-associated ciliopathies.


Subject(s)
Ciliopathies , Polycystic Kidney Diseases , Animals , Cilia/metabolism , Ciliopathies/drug therapy , Ciliopathies/genetics , Ciliopathies/metabolism , Female , Humans , Kidney Diseases, Cystic/congenital , Male , Mice , Polycystic Kidney Diseases/metabolism , Prostaglandins/metabolism , Receptors, Prostaglandin E/metabolism , Zebrafish
8.
Dev Biol ; 500: 1-9, 2023 08.
Article in English | MEDLINE | ID: mdl-37209936

ABSTRACT

ARL13B is a small GTPase enriched in cilia. Deletion of Arl13b in mouse kidney results in renal cysts and an associated absence of primary cilia. Similarly, ablation of cilia leads to kidney cysts. To investigate whether ARL13B functions from within cilia to direct kidney development, we examined kidneys of mice expressing an engineered cilia-excluded ARL13B variant, ARL13BV358A. These mice retained renal cilia and developed cystic kidneys. Because ARL13B functions as a guanine nucleotide exchange factor (GEF) for ARL3, we examined kidneys of mice expressing an ARL13B variant that lacks ARL3 GEF activity, ARL13BR79Q. We found normal kidney development with no evidence of cysts in these mice. Taken together, our results show that ARL13B functions within cilia to inhibit renal cystogenesis during mouse development, and that this function does not depend on its role as a GEF for ARL3.


Subject(s)
Kidney Diseases, Cystic , Kidney , Animals , Mice , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , Cilia/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Kidney/metabolism , Kidney Diseases, Cystic/genetics
9.
J Cell Physiol ; 239(4): e31189, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38219074

ABSTRACT

Joubert syndrome (JBTS) is a systematic developmental disorder mainly characterized by a pathognomonic mid-hindbrain malformation. All known JBTS-associated genes encode proteins involved in the function of antenna-like cellular organelle, primary cilium, which plays essential roles in cellular signal transduction and development. Here, we identified four unreported variants in ARL13B in two patients with the classical features of JBTS. ARL13B is a member of the Ras GTPase family and functions in ciliogenesis and cilia-related signaling. The two missense variants in ARL13B harbored the substitutions of amino acids at evolutionarily conserved positions. Using model cell lines, we found that the accumulations of the missense variants in cilia were impaired and the variants showed attenuated functions in ciliogenesis or the trafficking of INPP5E. Overall, these findings expanded the ARL13B pathogenetic variant spectrum of JBTS.


Subject(s)
ADP-Ribosylation Factors , Abnormalities, Multiple , Cerebellum , Eye Abnormalities , Kidney Diseases, Cystic , Retina , Humans , Abnormalities, Multiple/genetics , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , Cerebellum/abnormalities , Cilia/genetics , Eye Abnormalities/genetics , Eye Abnormalities/metabolism , Eye Abnormalities/pathology , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/metabolism , Kidney Diseases, Cystic/pathology , Phosphoric Monoester Hydrolases/metabolism , Retina/metabolism , Retina/abnormalities , Male , Female , Infant
10.
Hum Mol Genet ; 31(24): 4228-4240, 2022 12 16.
Article in English | MEDLINE | ID: mdl-35904445

ABSTRACT

Polycystic kidney disease (PKD) is a ciliopathy characterized by fluid-filled epithelial cysts in the kidney. Although it is well established that the primary cilium is essential for hedgehog (HH) signaling and HH signaling is abnormally activated in multiple PKD models, the mechanism and function of HH activation in PKD pathogenesis remain incompletely understood. Here we used a transgenic HH reporter mouse line to identify the target tissue of HH signaling in Arl13f/f;Ksp-Cre mutant kidney, in which the cilia biogenesis gene Arl13b is specifically deleted in epithelial cells of the distal nephron. In addition, we used a co-culture system to dissect cross-talk between epithelial and mesenchymal cells in the absence of expanding cysts. Finally, we treated Arl13bf/f;Ksp-Cre mice with the GLI inhibitor GANT61 and analyzed its impact on PKD progression in this model. We found that deletion of Arl13b in epithelial cells in the mouse kidney, in vivo, led to non-cell-autonomous activation of the HH pathway in the interstitium. In vitro, when co-cultured with mesenchymal cells, Arl13b-/- epithelial cells produced more sonic hedgehog in comparison to cells expressing Arl13b. Reciprocally, HH signaling was activated in mesenchymal cells co-cultured with Arl13b-/- epithelial cells. Finally, whole body inhibition of the HH pathway by GANT61 reduced the number of proliferating cells, inhibited cyst progression and fibrosis and preserved kidney function in Arl13bf/f;Ksp-Cre mice. Our results reveal non-cell-autonomous activation of HH signaling in the interstitium of the Arl13bf/f;Ksp-Cre kidney and suggest that abnormal activation of the HH pathway contributes to disease progression.


Subject(s)
Ciliopathies , Cysts , Kidney Diseases, Cystic , Mice , Animals , Hedgehog Proteins/metabolism , Kidney Diseases, Cystic/genetics , Disease Models, Animal , Mice, Transgenic , Ciliopathies/genetics , Disease Progression
11.
Hum Mol Genet ; 31(9): 1357-1369, 2022 05 04.
Article in English | MEDLINE | ID: mdl-34740236

ABSTRACT

Nephronophthisis-related ciliopathies (NPHP-RC) comprises a group of inherited kidney diseases, caused by mutations in genes encoding proteins localizing to primary cilia. NPHP-RC represents one of the most frequent monogenic causes of renal failure within the first three decades of life, but its molecular disease mechanisms remain unclear. Here, we identified biallelic ANKS6 mutations in two affected siblings with late-onset chronic kidney disease by whole-exome sequencing. We employed patient-derived fibroblasts generating an in vitro model to study the precise biological impact of distinct human ANKS6 mutations, completed by immunohistochemistry studies on renal biopsy samples. Functional studies using patient-derived cells showed an impaired integrity of the ciliary inversin compartment with reduced cilia length. Further analyses demonstrated that ANKS6 deficiency leads to a dysregulation of Hippo-signaling through nuclear yes-associated protein (YAP) imbalance and disrupted ciliary localization of YAP. In addition, an altered transcriptional activity of canonical Wnt target genes and altered expression of non-phosphorylated (active) ß-catenin and phosphorylated glycogen synthase kinase 3ß were observed. Upon ciliation, ANKS6 deficiency revealed a deranged subcellular localization and expression of components of the endocytic recycling compartment. Our results demonstrate that ANKS6 plays a key role in regulating the Hippo pathway, and ANKS6 deficiency is linked to dysregulation of signaling pathways. Our study provides molecular clues in understanding pathophysiological mechanisms of NPHP-RC and may offer new therapeutic targets.


Subject(s)
Ciliopathies , Kidney Diseases, Cystic , Polycystic Kidney Diseases , Renal Insufficiency, Chronic , Cilia/pathology , Ciliopathies/metabolism , Female , Humans , Kidney Diseases, Cystic/metabolism , Male , Mutation , Nuclear Proteins , Polycystic Kidney Diseases/genetics
12.
Hum Mol Genet ; 31(19): 3245-3265, 2022 09 29.
Article in English | MEDLINE | ID: mdl-35470378

ABSTRACT

Mutations in Talpid3, a basal body protein essential for the assembly of primary cilia, have been reported to be causative for Joubert Syndrome (JS). Herein, we report prominent developmental defects in the hippocampus of a conditional knockout mouse lacking the conserved exons 11 and 12 of Talpid3. At early postnatal stages, the Talpid3 mutants exhibit a reduction in proliferation in the dentate gyrus and a disrupted glial scaffold. The occurrence of mis-localized progenitors in the granule cell layer suggests a role for the disrupted glial scaffold in cell migration resulting in defective subpial neurogenic zone-to-hilar transition. Neurospheres derived from the hippocampus of Talpid3fl/flUbcCre mouse, in which Talpid3 was conditionally deleted, lacked primary cilia and were smaller in size. In addition, neurosphere cells showed a disrupted actin cytoskeleton and defective migration. Our findings suggest a link between the hippocampal defects and the learning/memory deficits seen in JS patients.


Subject(s)
Abnormalities, Multiple , Eye Abnormalities , Kidney Diseases, Cystic , Abnormalities, Multiple/genetics , Animals , Cerebellum/abnormalities , Dentate Gyrus , Eye Abnormalities/genetics , Hippocampus , Kidney Diseases, Cystic/genetics , Mice , Mice, Knockout , Neurogenesis/genetics , Retina/abnormalities
13.
Ann Hum Genet ; 88(1): 86-100, 2024 01.
Article in English | MEDLINE | ID: mdl-37921557

ABSTRACT

INTRODUCTION: Joubert syndrome (JS) arises from defects of primary cilia resulting in potential malformations of the brain, kidneys, eyes, liver, and limbs. Several of the 35+ genes associated with JS have recognized genotype/phenotype correlations, but most genes have not had enough reported individuals to draw meaningful conclusions. METHODS: A PubMed literature review identified 688 individuals with JS across 32 genes and 112 publications to bolster known genotype/phenotype relationships and identify new correlations. All included patients had the "molar tooth sign" and a confirmed genetic diagnosis. Individuals were categorized by age, ethnicity, sex and the presence of developmental disability/intellectual disability, hypotonia, abnormal eye movements, ataxia, visual impairment, renal impairment, polydactyly, and liver abnormalities. RESULTS: Most genes demonstrated unique phenotypic profiles. Grouping proteins based on physiologic interactions established stronger phenotypic relationships that reflect known ciliary pathophysiology. Age-stratified data demonstrated that end-organ disease is progressive in JS. Most genes demonstrated a significant skew towards having variants with either residual protein function or no residual protein function. CONCLUSION: This cohort demonstrates that clinically meaningful genotype/phenotype relationships exist within most JS-related genes and can be referenced to allow for more personalized clinical care.


Subject(s)
Abnormalities, Multiple , Eye Abnormalities , Kidney Diseases, Cystic , Humans , Abnormalities, Multiple/genetics , Cerebellum/abnormalities , Kidney Diseases, Cystic/genetics , Eye Abnormalities/genetics , Retina/abnormalities , Proteins/genetics , Biological Variation, Population
14.
Cell Tissue Res ; 396(2): 255-267, 2024 May.
Article in English | MEDLINE | ID: mdl-38502237

ABSTRACT

Joubert syndrome (JS) is a recessively inherited congenital ataxia characterized by hypotonia, psychomotor delay, abnormal ocular movements, intellectual disability, and a peculiar cerebellar and brainstem malformation, the "molar tooth sign." Over 40 causative genes have been reported, all encoding for proteins implicated in the structure or functioning of the primary cilium, a subcellular organelle widely present in embryonic and adult tissues. In this paper, we developed an in vitro neuronal differentiation model using patient-derived induced pluripotent stem cells (iPSCs), to evaluate possible neurodevelopmental defects in JS. To this end, iPSCs from four JS patients harboring mutations in distinct JS genes (AHI1, CPLANE1, TMEM67, and CC2D2A) were differentiated alongside healthy control cells to obtain mid-hindbrain precursors and cerebellar granule cells. Differentiation was monitored over 31 days through the detection of lineage-specific marker expression by qRT-PCR, immunofluorescence, and transcriptomics analysis. All JS patient-derived iPSCs, regardless of the mutant gene, showed a similar impairment to differentiate into mid-hindbrain and cerebellar granule cells when compared to healthy controls. In addition, analysis of primary cilium count and morphology showed notable ciliary defects in all differentiating JS patient-derived iPSCs compared to controls. These results confirm that patient-derived iPSCs are an accessible and relevant in vitro model to analyze cellular phenotypes connected to the presence of JS gene mutations in a neuronal context.


Subject(s)
Abnormalities, Multiple , Cell Differentiation , Cerebellum , Cerebellum/abnormalities , Eye Abnormalities , Induced Pluripotent Stem Cells , Kidney Diseases, Cystic , Neurons , Retina , Retina/abnormalities , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Humans , Eye Abnormalities/genetics , Eye Abnormalities/pathology , Cerebellum/pathology , Cerebellum/metabolism , Neurons/metabolism , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Retina/metabolism , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/pathology , Kidney Diseases, Cystic/metabolism , Male , Female , Mutation/genetics , Cilia/metabolism
15.
Am J Kidney Dis ; 84(1): 62-72.e1, 2024 07.
Article in English | MEDLINE | ID: mdl-38280640

ABSTRACT

RATIONALE & OBJECTIVE: Simple kidney cysts, which are common and usually considered of limited clinical relevance, are associated with older age and lower glomerular filtration rate (GFR), but little has been known of their association with progressive chronic kidney disease (CKD). STUDY DESIGN: Observational cohort study. SETTING & PARTICIPANTS: Patients with presurgical computed tomography or magnetic resonance imaging who underwent a radical nephrectomy for a tumor; we reviewed the retained kidney images to characterize parenchymal cysts at least 5mm in diameter according to size and location. EXPOSURE: Parenchymal cysts at least 5mm in diameter in the retained kidney. Cyst characteristics were correlated with microstructural findings on kidney histology. OUTCOME: Progressive CKD defined by dialysis, kidney transplantation, a sustained≥40% decline in eGFR for at least 3 months, or an eGFR<10mL/min/1.73m2 that was at least 5mL/min/1.73m2 below the postnephrectomy baseline for at least 3 months. ANALYTICAL APPROACH: Cox models assessed the risk of progressive CKD. Models adjusted for baseline age, sex, body mass index, hypertension, diabetes, eGFR, proteinuria, and tumor volume. Nonparametric Spearman's correlations were used to examine the association of the number and size of the cysts with clinical characteristics, kidney function, and kidney volumes. RESULTS: There were 1,195 patients with 50 progressive CKD events over a median 4.4 years of follow-up evaluation. On baseline imaging, 38% had at least 1 cyst, 34% had at least 1 cortical cyst, and 8.7% had at least 1 medullary cyst. A higher number of cysts was associated with progressive CKD and was modestly correlated with larger nephrons and more nephrosclerosis on kidney histology. The number of medullary cysts was more strongly associated with progressive CKD than the number of cortical cysts. LIMITATIONS: Patients who undergo a radical nephrectomy may differ from the general population. A radical nephrectomy may accelerate the risk of progressive CKD. Genetic testing was not performed. CONCLUSIONS: Cysts in the kidney, particularly the medulla, should be further examined as a potentially useful imaging biomarker of progressive CKD beyond the current clinical evaluation of kidney function and common CKD risk factors. PLAIN-LANGUAGE SUMMARY: Kidney cysts are common and often are considered of limited clinical relevance despite being associated with lower glomerular filtration rate. We studied a large cohort of patients who had a kidney removed due to a tumor to determine whether cysts in the retained kidney were associated with kidney health in the future. We found that more cysts in the kidney and, in particular, cysts in the deepest tissue of the kidney (the medulla) were associated with progressive kidney disease, including kidney failure where dialysis or a kidney transplantation is needed. Patients with cysts in the kidney medulla may benefit from closer monitoring.


Subject(s)
Disease Progression , Glomerular Filtration Rate , Kidney Diseases, Cystic , Nephrectomy , Renal Insufficiency, Chronic , Humans , Male , Female , Middle Aged , Renal Insufficiency, Chronic/epidemiology , Renal Insufficiency, Chronic/etiology , Kidney Diseases, Cystic/diagnostic imaging , Kidney Diseases, Cystic/pathology , Kidney Diseases, Cystic/surgery , Kidney Diseases, Cystic/etiology , Aged , Kidney Neoplasms/surgery , Kidney Neoplasms/pathology , Cohort Studies , Magnetic Resonance Imaging , Postoperative Complications/etiology , Postoperative Complications/epidemiology , Retrospective Studies , Tomography, X-Ray Computed
16.
Calcif Tissue Int ; 114(2): 137-146, 2024 02.
Article in English | MEDLINE | ID: mdl-37981601

ABSTRACT

Hereditary hypophosphatemic rickets with hypercalciuria (HHRH) is a rare disorder of phosphate homeostasis. We describe a single-center experience of genetically proven HHRH families and perform systematic review phenotype-genotype correlation in reported biallelic probands and their monoallelic relatives. Detailed clinical, biochemical, radiological, and genetic data were retrieved from our center and a systematic review of Pub-Med and Embase databases for patients and relatives who were genetically proven. Total of nine subjects (probands:5) carrying biallelic SLC34A3 mutations (novel:2) from our center had a spectrum from rickets/osteomalacia to normal BMD, with hypophosphatemia and hypercalciuria in all. We describe the first case of genetically proven HHRH with enthesopathy. Elevated FGF23 in another patient with hypophosphatemia, iron deficiency anemia, and noncirrhotic periportal fibrosis led to initial misdiagnosis as tumoral osteomalacia. On systematic review of 58 probands (with biallelic SLC34A3 mutations; 35 males), early-onset HHRH and renal calcification were present in ~ 70% and late-onset HHRH in 10%. c.575C > T p.(Ser192Leu) variant occurred in 53% of probands without skeletal involvement. Among 110 relatives harboring monoallelic SLC34A3 mutation at median age 38 years, renal calcification, hypophosphatemia, high 1,25(OH)2D, and hypercalciuria were observed in ~30%, 22.3%, 40%, and 38.8%, respectively. Renal calcifications correlated with age but were similar across truncating and non-truncating variants. Although most relatives were asymptomatic for bone involvement, 6/12(50%) had low bone mineral density. We describe the first monocentric HHRH case series from India with varied phenotypes. In a systematic review, frequent renal calcifications and low BMD in relatives with monoallelic variants (HHRH trait) merit identification.


Subject(s)
Enthesopathy , Familial Hypophosphatemic Rickets , Hypophosphatemia , Kidney Diseases, Cystic , Nephrocalcinosis , Osteomalacia , Male , Humans , Adult , Familial Hypophosphatemic Rickets/complications , Familial Hypophosphatemic Rickets/diagnosis , Familial Hypophosphatemic Rickets/genetics , Hypercalciuria/complications , Hypercalciuria/genetics , Osteomalacia/complications , Osteomalacia/genetics
17.
FASEB J ; 37(1): e22696, 2023 01.
Article in English | MEDLINE | ID: mdl-36520027

ABSTRACT

Mutations or deletions in transcription factor hepatocyte nuclear factor 1 homeobox ß (HNF1ß) cause renal cysts and/or malformation, maturity-onset diabetes of the young and electrolyte disturbances. Here, we applied a comprehensive bioinformatic approach on ChIP-seq, RNA-seq, and gene expression array studies to identify novel transcriptional targets of HNF1ß explaining the kidney phenotype of HNF1ß patients. We identified BAR/IMD Domain Containing Adaptor Protein 2 Like 2 (BAIAP2L2), as a novel transcriptional target of HNF1ß and validated direct transcriptional activation of the BAIAP2L2 promoter by a reporter luciferase assay. Using mass spectrometry analysis, we show that BAIAP2L2 binds to other members of the I-BAR domain-containing family: BAIAP2 and BAIAP2L1. Subsequently, the role of BAIAP2L2 in maintaining epithelial cell integrity in the kidney was assessed using Baiap2l2 knockout cell and mouse models. Kidney epithelial cells lacking functional BAIAP2L2 displayed normal F-actin distribution at cell-cell contacts and formed polarized three-dimensional spheroids with a lumen. In vivo, Baiap2l2 knockout mice displayed normal kidney and colon tissue morphology and serum and urine electrolyte concentrations were not affected. Altogether, our study is the first to characterize the function of BAIAP2L2 in the kidney in vivo and we report that mice lacking BAIAP2L2 exhibit normal electrolyte homeostasis and tissue morphology under physiological conditions.


Subject(s)
Cysts , Kidney Diseases, Cystic , Animals , Humans , Mice , Cysts/genetics , Cysts/metabolism , Electrolytes/metabolism , Kidney/metabolism , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/metabolism , Mice, Knockout , Transcription Factors/metabolism , Transcriptional Activation
18.
Cerebellum ; 23(2): 579-588, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37351729

ABSTRACT

The neuropsychological characteristics of the cerebellar cognitive affective syndrome (CCAS) in congenital, non-progressive malformations of the cerebellum have been scarcely investigated, and even less is known for Joubert syndrome (JS), an inherited, non-progressive cerebellar ataxia characterized by the so-called molar tooth sign. The few studies on this topic reported inconsistent results about intellectual functioning and specific neuropsychological impairments. The aim of this research is to examine the neuropsychological profile of JS compared to other congenital cerebellar malformations (CM), considering individual variability of intellectual quotient (IQ) in the two groups. Fourteen patients with JS and 15 patients with CM aged 6-25 years were tested through a comprehensive, standardized neuropsychological battery. Their scores in the neuropsychological domains were inspected through descriptive analysis and compared by mean of MANOVA and ANOVA models, then replicated inserting IQ as covariate. The two groups showed a largely overlapping neuropsychological profile, consistent with CCAS. However, the JS group showed worse performance in visual-spatial memory compared to CM patients, although this difference was mitigated when considering IQ. These findings highlight a divergence between JS and other CM in visual-spatial memory, which might suggest a critical role of the cerebellum in recalling task-relevant memories and might inform rehabilitative interventions.


Subject(s)
Abnormalities, Multiple , Cerebellar Diseases , Cerebellum/abnormalities , Eye Abnormalities , Kidney Diseases, Cystic , Retina/abnormalities , Humans , Abnormalities, Multiple/psychology , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/psychology , Eye Abnormalities/psychology
19.
Cerebellum ; 23(4): 1736-1740, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38334877

ABSTRACT

KBG syndrome is a rare genetic disorder caused by heterozygous pathogenic variants in ANKRD11. Affected individuals have developmental delay, short stature, characteristic facial features, and other dysmorphic findings. To date, a spectrum of unspecific neuroradiological defects has been reported in KBG patients, such as cortical defects, white matter abnormalities, corpus callosum, and cerebellar vermis hypoplasia.Deep clinical and neuroradiological phenotyping and genotype of a patient presenting with mild cognitive and behavioral problems were obtained after written informed consent.We herein describe the first KBG patient presenting with cerebellar heterotopia, a heterogeneous malformation characterized by the presence of clusters of neurons within the white matter of cerebellar hemispheres.This novel association broadens the neuroradiological spectrum of KBG syndrome, and further prompts to investigate the potential functions of ANKRD11 in cerebellar development.


Subject(s)
Cerebellum , Humans , Cerebellum/diagnostic imaging , Cerebellum/abnormalities , Cerebellum/pathology , Intellectual Disability/genetics , Intellectual Disability/diagnostic imaging , Intellectual Disability/pathology , Abnormalities, Multiple/genetics , Abnormalities, Multiple/diagnostic imaging , Abnormalities, Multiple/pathology , Magnetic Resonance Imaging , Male , Facies , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/diagnostic imaging , Kidney Diseases, Cystic/pathology , Female , Neurocutaneous Syndromes/diagnostic imaging , Neurocutaneous Syndromes/genetics , Neurocutaneous Syndromes/pathology , Child , Choristoma/diagnostic imaging , Choristoma/pathology , Bone Diseases, Developmental , Tooth Abnormalities
20.
Nephrol Dial Transplant ; 39(8): 1288-1298, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-38178635

ABSTRACT

BACKGROUND: Autosomal dominant Alport Syndrome (ADAS), also known as thin basement membrane disease (TBMD), is caused by pathogenic variants in the COL4A3 and COL4A4 genes. A cystic phenotype has been described in some patients with TBMD, but no genetic studies have been performed. We conducted a genetic and radiologic investigation in a cohort of ADAS patients to analyze the prevalence of multicystic kidney disease (MKD) and its association with chronic kidney disease (CKD). METHODS: This was a retrospective single-center cohort study. Thirty-one patients showing pathogenic or likely pathogenic variants in COL4A3 or COL4A4 from a cohort of 79 patients with persistent microscopic hematuria were included. Mean follow-up was 9.4 ± 9.6 years. The primary objective of the study was to determine the prevalence of MKD in the cohort of ADAS patients. Secondary objectives were to determine risk factors associated with an estimated glomerular filtration rate (eGFR) <45 mL/min/1.73 m2 at the time of genetic and radiologic evaluation and to investigate the coexistence of other genetic abnormalities associated with familial hematuria and cystic kidney disease. RESULTS: MKD was found in 16 patients (52%). Mean number of cysts per kidney was 12.7 ± 5.5. No genetic abnormalities were found in a panel of 101 other genes related to familial hematuria, focal segmental glomerulosclerosis and cystic kidney disease. A greater number of patients with MKD had an eGFR <45 mL/min/1.73 m2 (63% vs 7%, P = .006) and more advanced CKD than patients without MKD. The annual rate of eGFR decline was greater in patients with MKD: -1.8 vs 0.06 mL/min/1.73 m2/year (P = .009). By multivariable linear regression analysis, the main determinants of eGFR change per year were time-averaged proteinuria (P = .002) and MKD (P = .02). CONCLUSION: MKD is commonly found in ADAS and is associated with a worse kidney outcome. No pathogenic variants were found in genes other than COL4A3/COL4A4.


Subject(s)
Collagen Type IV , Glomerular Filtration Rate , Nephritis, Hereditary , Phenotype , Renal Insufficiency, Chronic , Humans , Nephritis, Hereditary/genetics , Nephritis, Hereditary/complications , Female , Male , Retrospective Studies , Adult , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/epidemiology , Collagen Type IV/genetics , Follow-Up Studies , Middle Aged , Autoantigens/genetics , Prognosis , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/epidemiology , Kidney Diseases, Cystic/complications , Prevalence
SELECTION OF CITATIONS
SEARCH DETAIL