20 research outputs found

    IFT74 variants cause skeletal ciliopathy and motile cilia defects in mice and humans

    Get PDF
    Motile and non-motile cilia play critical roles in mammalian development and health. These organelles are composed of a 1000 or more unique proteins, but their assembly depends entirely on proteins synthesized in the cell body and transported into the cilium by intraflagellar transport (IFT). In mammals, malfunction of non-motile cilia due to IFT dysfunction results in complex developmental phenotypes that affect most organs. In contrast, disruption of motile cilia function causes subfertility, disruption of the left-right body axis, and recurrent airway infections with progressive lung damage. In this work, we characterize allele specific phenotypes resulting from IFT74 dysfunction in human and mice. We identified two families carrying a deletion encompassing IFT74 exon 2, the first coding exon, resulting in a protein lacking the first 40 amino acids and two individuals carrying biallelic splice site mutations. Homozygous exon 2 deletion cases presented a ciliary chondrodysplasia with narrow thorax and progressive growth retardation along with a mucociliary clearance disorder phenotype with severely shorted cilia. Splice site variants resulted in a lethal skeletal chondrodysplasia phenotype. In mice, removal of the first 40 amino acids likewise results in a motile cilia phenotype but with little effect on primary cilia structure. Mice carrying this allele are born alive but are growth restricted and developed hydrocephaly in the first month of life. In contrast, a strong, likely null, allele of Ift74 in mouse completely blocks ciliary assembly and causes severe heart defects and midgestational lethality. In vitro studies suggest that the first 40 amino acids of IFT74 are dispensable for binding of other IFT subunits but are important for tubulin binding. Higher demands on tubulin transport in motile cilia compared to primary cilia resulting from increased mechanical stress and repair needs could account for the motile cilia phenotype observed in human and mice

    IFT74 variants cause skeletal ciliopathy and motile cilia defects in mice and humans

    Get PDF
    Motile and non-motile cilia play critical roles in mammalian development and health. These organelles are composed of a 1000 or more unique proteins, but their assembly depends entirely on proteins synthesized in the cell body and transported into the cilium by intraflagellar transport (IFT). In mammals, malfunction of non-motile cilia due to IFT dysfunction results in complex developmental phenotypes that affect most organs. In contrast, disruption of motile cilia function causes subfertility, disruption of the left-right body axis, and recurrent airway infections with progressive lung damage. In this work, we characterize allele specific phenotypes resulting from IFT74 dysfunction in human and mice. We identified two families carrying a deletion encompassing IFT74 exon 2, the first coding exon, resulting in a protein lacking the first 40 amino acids and two individuals carrying biallelic splice site mutations. Homozygous exon 2 deletion cases presented a ciliary chondrodysplasia with narrow thorax and progressive growth retardation along with a mucociliary clearance disorder phenotype with severely shorted cilia. Splice site variants resulted in a lethal skeletal chondrodysplasia phenotype. In mice, removal of the first 40 amino acids likewise results in a motile cilia phenotype but with little effect on primary cilia structure. Mice carrying this allele are born alive but are growth restricted and developed hydrocephaly in the first month of life. In contrast, a strong, likely null, allele of Ift74 in mouse completely blocks ciliary assembly and causes severe heart defects and midgestational lethality. In vitro studies suggest that the first 40 amino acids of IFT74 are dispensable for binding of other IFT subunits but are important for tubulin binding. Higher demands on tubulin transport in motile cilia compared to primary cilia resulting from increased mechanical stress and repair needs could account for the motile cilia phenotype observed in human and mice

    Mimicry and well known genetic friends: molecular diagnosis in an Iranian cohort of suspected Bartter syndrome and proposition of an algorithm for clinical differential diagnosis.

    Get PDF
    BACKGROUND: Bartter Syndrome is a rare, genetically heterogeneous, mainly autosomal recessively inherited condition characterized by hypochloremic hypokalemic metabolic alkalosis. Mutations in several genes encoding for ion channels localizing to the renal tubules including SLC12A1, KCNJ1, BSND, CLCNKA, CLCNKB, MAGED2 and CASR have been identified as underlying molecular cause. No genetically defined cases have been described in the Iranian population to date. Like for other rare genetic disorders, implementation of Next Generation Sequencing (NGS) technologies has greatly facilitated genetic diagnostics and counseling over the last years. In this study, we describe the clinical, biochemical and genetic characteristics of patients from 15 Iranian families with a clinical diagnosis of Bartter Syndrome. RESULTS: Age range of patients included in this study was 3 months to 6 years and all patients showed hypokalemic metabolic alkalosis. 3 patients additionally displayed hypercalciuria, with evidence of nephrocalcinosis in one case. Screening by Whole Exome Sequencing (WES) and long range PCR revealed that 12/17 patients (70%) had a deletion of the entire CLCNKB gene that was previously identified as the most common cause of Bartter Syndrome in other populations. 4/17 individuals (approximately 25% of cases) were found to suffer in fact from pseudo-Bartter syndrome resulting from congenital chloride diarrhea due to a novel homozygous mutation in the SLC26A3 gene, Pendred syndrome due to a known homozygous mutation in SLC26A4, Cystic Fibrosis (CF) due to a novel mutation in CFTR and apparent mineralocorticoid excess syndrome due to a novel homozygous loss of function mutation in HSD11B2 gene. 1 case (5%) remained unsolved. CONCLUSIONS: Our findings demonstrate deletion of CLCNKB is the most common cause of Bartter syndrome in Iranian patients and we show that age of onset of clinical symptoms as well as clinical features amongst those patients are variable. Further, using WES we were able to prove that nearly 1/4 patients in fact suffered from Pseudo-Bartter Syndrome, reversing the initial clinical diagnosis with important impact on the subsequent treatment and clinical follow up pathway. Finally, we propose an algorithm for clinical differential diagnosis of Bartter Syndrome

    Roles of anks 6 and the matrix metalloprotease 9 in polycystic kidney diseases

    No full text
    Dans ce travail de thèse, nous avons mené deux projets différents concernant la polykystose rénale. Dans le premier travail, nous avons caractérisé un nouveau modèle murin de polykystose rénale autosomique dominante muté dans le domaine SAM de la protéine ANKS6 (ANKS6I747N), qui confirme le rôle du domaine SAM dans la fonction rénale. Nos souris développent une maladie kystique différente de celle observée dans le modèle de rat cy/+ muté aussi dans le domaine SAM d'ANKS6 (R823W) (à 6 acides aminés d'intervalle). Nous suggérons que la protéine ANKS6 mutée sur le domaine SAM (I747N) séquestre BICC1, un antagoniste du miARN17 qui dégrade le transcrit de la polycystine2. Cette action induirait une diminution de l'expression de polycystine2 ce qui activerait la voie Wnt/Beta caténine, une voie induite dans les maladies kystiques rénales. Chez le rat, la mutation(R823W) altère l'interaction entre ANKS6 et ANKS3. Dans le deuxième travail, nous avons montré pour la première fois un rôle protecteur de la MMP9 sur la formation des kystes. Nous avons croisé les souris jck, un modèle de polykystose rénale, avec les souris déficientes en MMP9. Les souris déficientes en MMP9 développent une insuffisance rénale plus sévère dès l'âge de 1 mois. Ceci est dû à des lésions plus sévères qui se manifestent par une augmentation très significative du poids des reins rapporté au poids corporel et à une augmentation du nombre de kystes. En revanche, les kystes sont significativement plus petits chez les animaux déficients en MMP9 par rapport aux témoins, suggérant que la MMP9 exerce un effet protecteur sur la formation des kystes et un rôle délétère sur leur croissance.In this thesis, we conducted two different projects about polycystic kidney disease. In the first study, we characterized a novel mouse model of polycystic. We screened an ENU treated mouse library and identified a strain carrying a missense mutation leading to an amino acid replacement (I747N) within the SAM domain of ANKS6, 6 amino acid away from the mutation R823W in the rat. Because the mice develop PKD, this mutation confirms the role of the SAM domain of ANKS6 in kidney function. Comparison of PKD/Mhm(Cy/+) rat and of our Anks6(I747N) mouse model further shows that the two models display noticeably different PKD phenotypes and that cystic enlargement is due to defective interaction with different protein partners, ANKS3 and BICC1, respectively in the rat and in the mouse. In the second study, we showed for the first time a protective role of MMP9 on the formation of cysts. We generated jck-MMP9 deficient mice inbred on C57Bl/6J background and compared them to jck littermates. Apoptosis and cell proliferation were similar in both groups of mice, while MMP9 deficiency induced more severe renal lesions, characterized by an increase in cyst number and in the amount of fibrosis. Indeed, life span of these mice was reduced. In addition, the mice exhibited shorter cilia length, probably due to the presence of MMP9 in this organelle. The mechanism of the protective effect of MMP9 on cystic phenotype is still not elucidated and is most likely due to the anti fibrotic activity of the enzyme

    Cleavage of periostin by MMP9 protects mice from kidney cystic disease.

    No full text
    The matrix metalloproteinase MMP9 influences cellular morphology and function, and plays important roles in organogenesis and disease. It exerts both protective and deleterious effects in renal pathology, depending upon its specific substrates. To explore new functions for MMP9 in kidney cysts formation and disease progression, we generated a mouse model by breeding juvenile cystic kidney (jck) mice with MMP9 deficient mice. Specifically, we provide evidence that MMP9 is overexpressed in cystic tissue where its enzymatic activity is increased 7-fold. MMP9 deficiency in cystic kidney worsen cystic kidney diseases by decreasing renal function, favoring cyst expansion and fibrosis. In addition, we find that periostin is a new critical substrate for MMP9 and in its absence periostin accumulates in cystic lining cells. As periostin promotes renal cyst growth and interstitial fibrosis in polycystic kidney diseases, we propose that the control of periostin by MMP9 and its associated intracellular signaling pathways including integrins, integrin-linked kinase and focal adhesion kinase confers to MMP9 a protective effect on the severity of the disease

    ANKS3 Co-Localises with ANKS6 in Mouse Renal Cilia and Is Associated with Vasopressin Signaling and Apoptosis In Vivo in Mice

    Get PDF
    International audienceMutations in Ankyrin repeat and sterile alpha motif domain containing 6 (ANKS6) play a causative role in renal cyst formation in the PKD/Mhm(cy/+) rat model of polycystic kidney disease and in nephronophthisis in humans. A network of protein partners of ANKS6 is emerging and their functional characterization provides important clues to understand the role of ANKS6 in renal biology and in mechanisms involved in the formation of renal cysts. Following experimental confirmation of interaction between ANKS6and ANKS3 using a Yeast two hybrid system, we demonstrated that binding between the two proteins occurs through their sterile alpha motif (SAM) and that the amino acid 823 in rat ANSK6 is key for this interaction. We further showed their interaction by co-immunoprecipitation and showed in vivo in mice that ANKS3 is present in renal cilia. Downregulated expression of Anks3 in vivo in mice by Locked Nucleic Acid (LNA) modified antisense oligonucleotides was associated with increased transcription of vasopressin-induced genes, suggesting changes in renal water permeability, and altered transcription of genes encoding proteins involved in cilium structure, apoptosis and cell proliferation. These data provide experimental evidence of ANKS3-ANKS6 direct interaction through their SAM domain and co-localisation in mouse renal cilia, and shed light on molecular mechanisms indirectly mediated by ANKS6 in the mouse kidney, that may be affected by altered ANKS3-ANKS6 interaction. Our results contribute to improved knowledge of the structure and function of the network of proteins interacting with ANKS6, which may represent therapeutic targets in cystic diseases

    <i>Anks3</i> expression knock-down <i>in vivo</i> stimulates expression of genes involved in the vasopressin signaling pathway.

    No full text
    <p>(<b>A</b>-<b>B</b>) Renal mRNA expression of <i>Vit32</i>, <i>Aqp1</i>, <i>Aqp2</i> and <i>Aqp3</i> was evaluated by quantitative RT-PCR in mice four days (n = 3)(<b>A</b>) and twelve days (n = 5)(<b>B</b>) after the final injection of Locked Nucleic Acid modified AntiSense Oligonucleotides (LNA ASO). Quantification of each cDNA was performed in duplicate and normalized to <i>Gusb</i> gene expression level. (<b>C</b>) AQP2 expression in kidney tissues. Representative Western blots performed with protein extracts from 5 kidneys treated with scrambled (SCR) or ANKS3 ASO (top) and quantitative analysis of blots normalized to β-actin expression (bottom) show that AQP2 expression is significantly increased in mice twelve days after the final injection of ANKS3 ASO when compared to mice injected with SCR ASO. (<b>D</b>, <b>E</b>) Immunofluorescence staining of the papilla for AQP2 (<b>D</b>) and AVPR2 (<b>E</b>) show a dramatic increase of the proteins in kidneys of ANKS3 ASO treated mice twelve days after the final injection compared to SCR ASO controls. Scale bar: 300 μm. Data are means ± SEM. Non-parametric Mann-Whitney U test was used to assess differences between ANKS3 ASO and control mice injected with saline (Ctr)- or SCR ASO. *P<0.05; **P<0.01 significantly different to controls.</p

    The mutation (R823W) in PKD/Mhm(<i>cy/cy</i>) rats does not alter ANKS6-ANKS3 renal co-localisation.

    No full text
    <p>Immunohistochemical staining of renal cortex and medulla was performed in 3 week-old wild-type and PKD/Mhm(<i>cy/cy</i>) rats for ANKS3 (upper panel) and ANKS6 (lower panel).</p

    <i>In vivo</i> LNA ASO-induced downregulation of<i>Anks3</i> expression in mouse kidney.

    No full text
    <p>(<b>A</b>) Presence of ANKS3 in kidneys treated with saline (CTR) and scrambled AntiSense Oligonucleotides (SCR ASO) 12 days post injection and with ANKS3 Locked Nucleic Acid modified AntiSense Oligonucleotides (ANKS3 LNA ASOs). Representative photomicrographs of kidney sections from 5 mice four and twelve days after the last injection of LNA ASOs (D4 post inj., D12 post inj.). Kidneys were stained with ANKS3 (revealed by Alexa Fluor 488 in green) and the LNA ASOs were localised with the Alexa Fluor 647 marker in red. (<b>B-D</b>) <i>Anks3</i> renal decreased expression does not affect expression of <i>Anks6</i>. Abundance of <i>Anks3</i> transcripts (<b>B</b>) and protein (<b>C</b>), and <i>Anks6</i> transcripts (<b>D</b>) were evaluated in mouse kidneys four days (D4 post inj., n = 3) and twelve days (D12 post inj., n = 5) after the final injection. cDNA quantification was performed in duplicate and normalized to <i>Actb</i> gene expression level. (<b>C</b>) Western blots (upper panel) and quantitative protein analysis (lower panel) performed with extracts from kidneys of 5 mice treated with saline (Ctr), scramble ASO (SCR ASO) or ANKS3 ASO four and twelve days after the last injection showed that expression of ANKS3 normalized to that of β-actin is significantly decreased in mice treated with ANKS3 ASO. Data are mean ± SEM. Non-parametric Mann-Whitney U test was used to assess differences between ANKS3 ASO and SCR ASO treated mice.**P<0.01; *P<0.05 significantly different to control mice treated with saline (Ctr) or SCR ASO.</p
    corecore