Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(18): e2115960119, 2022 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-35482924

RESUMO

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.


Assuntos
Ciliopatias , Doenças Renais Policísticas , Animais , Cílios/metabolismo , Ciliopatias/tratamento farmacológico , Ciliopatias/genética , Ciliopatias/metabolismo , Feminino , Humanos , Doenças Renais Císticas/congênito , Masculino , Camundongos , Doenças Renais Policísticas/metabolismo , Prostaglandinas/metabolismo , Receptores de Prostaglandina E/metabolismo , Peixe-Zebra
2.
Kidney Int ; 104(2): 245-253, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37244473

RESUMO

Nephronophthisis is an autosomal recessive tubulointerstitial nephropathy, belonging to the ciliopathy disorders, characterized by fibrosis and/or cysts. It is the most common genetic cause of kidney failure in children and young adults. Clinically and genetically heterogeneous, it is caused by variants in ciliary genes, resulting in either an isolated kidney disease or syndromic forms in association with other manifestations of ciliopathy disorders. No curative treatment is currently available. Over the past 2 decades, advances in understanding disease mechanisms have identified several dysregulated signaling pathways, some shared with other cystic kidney diseases. Notably, molecules previously developed to target these pathways have shown promising beneficial effects in orthologous mouse models. In addition to these knowledge-based repurposing approaches, unbiased "in cellulo" phenotypic screens of "repurposing" libraries identified small molecules able to rescue the ciliogenesis defects observed in nephronophthisis conditions. Those compounds appeared to act on relevant pathways and, when tested, showed beneficial nephronophthisis-associated kidney and/or extrarenal defects in mice. In this review, we have summarized those studies that highlight the drug repurposing strategies in the context of a rare disorders, such as nephronophthisis-related ciliopathies, with broad genetic heterogeneity and systemic manifestations but with shared disease mechanisms.


Assuntos
Ciliopatias , Doenças Renais Císticas , Doenças Renais Policísticas , Insuficiência Renal , Animais , Camundongos , Rim/patologia , Doenças Renais Policísticas/genética , Doenças Renais Císticas/tratamento farmacológico , Doenças Renais Císticas/genética , Ciliopatias/tratamento farmacológico , Ciliopatias/genética , Insuficiência Renal/complicações , Fibrose , Cílios/patologia
3.
Kidney Int ; 104(2): 378-387, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37230223

RESUMO

Nephronophthisis (NPH) is an autosomal-recessive ciliopathy representing one of the most frequent causes of kidney failure in childhood characterized by a broad clinical and genetic heterogeneity. Applied to one of the worldwide largest cohorts of patients with NPH, genetic analysis encompassing targeted and whole exome sequencing identified disease-causing variants in 600 patients from 496 families with a detection rate of 71%. Of 788 pathogenic variants, 40 known ciliopathy genes were identified. However, the majority of patients (53%) bore biallelic pathogenic variants in NPHP1. NPH-causing gene alterations affected all ciliary modules defined by structural and/or functional subdomains. Seventy six percent of these patients had progressed to kidney failure, of which 18% had an infantile form (under five years) and harbored variants affecting the Inversin compartment or intraflagellar transport complex A. Forty eight percent of patients showed a juvenile (5-15 years) and 34% a late-onset disease (over 15 years), the latter mostly carrying variants belonging to the Transition Zone module. Furthermore, while more than 85% of patients with an infantile form presented with extra-kidney manifestations, it only concerned half of juvenile and late onset cases. Eye involvement represented a predominant feature, followed by cerebellar hypoplasia and other brain abnormalities, liver and skeletal defects. The phenotypic variability was in a large part associated with mutation types, genes and corresponding ciliary modules with hypomorphic variants in ciliary genes playing a role in early steps of ciliogenesis associated with juvenile-to-late onset NPH forms. Thus, our data confirm a considerable proportion of late-onset NPH suggesting an underdiagnosis in adult chronic kidney disease.


Assuntos
Ciliopatias , Doenças Renais Císticas , Falência Renal Crônica , Doenças Renais Policísticas , Adulto , Humanos , Falência Renal Crônica/diagnóstico , Doenças Renais Policísticas/complicações , Doenças Renais Císticas/genética , Doenças Renais Císticas/patologia , Mutação , Ciliopatias/genética
4.
J Cell Sci ; 134(2)2021 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-33483367

RESUMO

Ciliogenesis is a coordinated process initiated by the recruitment and fusion of pre-ciliary vesicles at the distal appendages of the mother centriole through mechanisms that remain unclear. Here, we report that EFA6A (also known as PSD), an exchange factor for the small G protein Arf6, is involved in early stage of ciliogenesis by promoting the fusion of distal appendage vesicles forming the ciliary vesicle. EFA6A is present in the vicinity of the mother centriole before primary cilium assembly and prior to the arrival of Arl13B-containing vesicles. During ciliogenesis, EFA6A initially accumulates at the mother centriole and later colocalizes with Arl13B along the ciliary membrane. EFA6A depletion leads to the inhibition of ciliogenesis, the absence of centrosomal Rab8-positive structures and the accumulation of Arl13B-positive vesicles around the distal appendages. Our results uncover a novel fusion machinery, comprising EFA6A, Arf6 and Arl13B, that controls the coordinated fusion of ciliary vesicles docked at the distal appendages of the mother centriole.


Assuntos
Fatores de Ribosilação do ADP , Centríolos , Cílios , Fatores de Troca do Nucleotídeo Guanina , Animais , Linhagem Celular , Vesículas Citoplasmáticas
5.
Am J Hum Genet ; 104(2): 348-355, 2019 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-30661770

RESUMO

Steroid-resistant nephrotic syndrome (SRNS) is characterized by high-range proteinuria and most often focal and segmental glomerulosclerosis (FSGS). Identification of mutations in genes causing SRNS has improved our understanding of disease mechanisms and highlighted defects in the podocyte, a highly specialized glomerular epithelial cell, as major factors in disease pathogenesis. By exome sequencing, we identified missense mutations in TBC1D8B in two families with an X-linked early-onset SRNS with FSGS. TBC1D8B is an uncharacterized Rab-GTPase-activating protein likely involved in endocytic and recycling pathways. Immunofluorescence studies revealed TBC1D8B presence in human glomeruli, and affected individual podocytes displayed architectural changes associated with migration defects commonly found in FSGS. In zebrafish we demonstrated that both knockdown and knockout of the unique TBC1D8B ortholog-induced proteinuria and that this phenotype was rescued by human TBC1D8B mRNA injection, but not by either of the two mutated mRNAs. We also showed an interaction between TBC1D8B and Rab11b, a key protein in vesicular recycling in cells. Interestingly, both internalization and recycling processes were dramatically decreased in affected individuals' podocytes and fibroblasts, confirming the crucial role of TBC1D8B in the cellular recycling processes, probably as a Rab11b GTPase-activating protein. Altogether, these results confirmed that pathogenic variations in TBC1D8B are involved in X-linked podocytopathy and points to alterations in recycling processes as a mechanism of SRNS.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Doenças Genéticas Ligadas ao Cromossomo X/genética , Mutação com Perda de Função , Síndrome Nefrótica/genética , Proteínas de Transporte Vesicular/genética , Proteínas de Peixe-Zebra/genética , Animais , Transporte Biológico/genética , Proteínas de Ligação ao Cálcio/metabolismo , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Glomérulos Renais/metabolismo , Masculino , Podócitos/citologia , Podócitos/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Sequenciamento do Exoma , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo
6.
Hum Mol Genet ; 28(16): 2720-2737, 2019 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-31042281

RESUMO

Mutations in genes encoding components of the intraflagellar transport (IFT) complexes have previously been associated with a spectrum of diseases collectively termed ciliopathies. Ciliopathies relate to defects in the formation or function of the cilium, a sensory or motile organelle present on the surface of most cell types. IFT52 is a key component of the IFT-B complex and ensures the interaction of the two subcomplexes, IFT-B1 and IFT-B2. Here, we report novel IFT52 biallelic mutations in cases with a short-rib thoracic dysplasia (SRTD) or a congenital anomaly of kidney and urinary tract (CAKUT). Combining in vitro and in vivo studies in zebrafish, we showed that SRTD-associated missense mutation impairs IFT-B complex assembly and IFT-B2 ciliary localization, resulting in decreased cilia length. In comparison, CAKUT-associated missense mutation has a mild pathogenicity, thus explaining the lack of skeletal defects in CAKUT case. In parallel, we demonstrated that the previously reported homozygous nonsense IFT52 mutation associated with Sensenbrenner syndrome [Girisha et al. (2016) A homozygous nonsense variant in IFT52 is associated with a human skeletal ciliopathy. Clin. Genet., 90, 536-539] leads to exon skipping and results in a partially functional protein. Finally, our work uncovered a novel role for IFT52 in microtubule network regulation. We showed that IFT52 interacts and partially co-localized with centrin at the distal end of centrioles where it is involved in its recruitment and/or maintenance. Alteration of this function likely contributes to centriole splitting observed in Ift52-/- cells. Altogether, our findings allow a better comprehensive genotype-phenotype correlation among IFT52-related cases and revealed a novel, extra-ciliary role for IFT52, i.e. disruption may contribute to pathophysiological mechanisms.


Assuntos
Proteínas de Transporte/genética , Centrossomo/metabolismo , Estudos de Associação Genética , Predisposição Genética para Doença , Microtúbulos/metabolismo , Mutação , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Criança , Pré-Escolar , Cílios/metabolismo , Consanguinidade , Análise Mutacional de DNA , Feminino , Genótipo , Homozigoto , Humanos , Lactente , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Linhagem , Fenótipo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas/genética , Combinação Trimetoprima e Sulfametoxazol/metabolismo , Sequenciamento do Exoma , Peixe-Zebra
7.
Hum Mol Genet ; 28(5): 778-795, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30388224

RESUMO

Mutations in KIF14 have previously been associated with either severe, isolated or syndromic microcephaly with renal hypodysplasia (RHD). Syndromic microcephaly-RHD was strongly reminiscent of clinical ciliopathies, relating to defects of the primary cilium, a signalling organelle present on the surface of many quiescent cells. KIF14 encodes a mitotic kinesin, which plays a key role at the midbody during cytokinesis and has not previously been shown to be involved in cilia-related functions. Here, we analysed four families with fetuses presenting with the syndromic form and harbouring biallelic variants in KIF14. Our functional analyses showed that the identified variants severely impact the activity of KIF14 and likely correspond to loss-of-function mutations. Analysis in human fetal tissues further revealed the accumulation of KIF14-positive midbody remnants in the lumen of ureteric bud tips indicating a shared function of KIF14 during brain and kidney development. Subsequently, analysis of a kif14 mutant zebrafish line showed a conserved role for this mitotic kinesin. Interestingly, ciliopathy-associated phenotypes were also present in mutant embryos, supporting a potential direct or indirect role for KIF14 at cilia. However, our in vitro and in vivo analyses did not provide evidence of a direct role for KIF14 in ciliogenesis and suggested that loss of kif14 causes ciliopathy-like phenotypes through an accumulation of mitotic cells in ciliated tissues. Altogether, our results demonstrate that KIF14 mutations result in a severe syndrome associating microcephaly and RHD through its conserved function in cytokinesis during kidney and brain development.


Assuntos
Anormalidades Congênitas/genética , Estudos de Associação Genética , Predisposição Genética para Doença , Nefropatias/congênito , Rim/anormalidades , Cinesinas/genética , Mutação com Perda de Função , Microcefalia/genética , Proteínas Oncogênicas/genética , Animais , Anormalidades Congênitas/metabolismo , Citocinese/genética , Modelos Animais de Doenças , Feminino , Imunofluorescência , Genes Letais , Estudos de Associação Genética/métodos , Loci Gênicos , Humanos , Rim/metabolismo , Nefropatias/genética , Nefropatias/metabolismo , Cinesinas/química , Cinesinas/metabolismo , Masculino , Microcefalia/metabolismo , Microcefalia/patologia , Proteínas Oncogênicas/química , Proteínas Oncogênicas/metabolismo , Linhagem , Fenótipo , Relação Estrutura-Atividade , Peixe-Zebra
8.
Kidney Int ; 98(4): 958-969, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32505465

RESUMO

Biallelic mutations in MAPKBP1 were recently associated with late-onset cilia-independent nephronophthisis. MAPKBP1 was found at mitotic spindle poles but could not be detected at primary cilia or centrosomes. Here, by identification and characterization of novel MAPKBP1 variants, we aimed at further investigating its role in health and disease. Genetic analysis was done by exome sequencing, homozygosity mapping, and a targeted kidney gene panel while coimmunoprecipitation was used to explore wild-type and mutant protein-protein interactions. Expression of MAPKBP1 in non-ciliated HeLa and ciliated inner medullary collecting duct cells enabled co-localization studies by fluorescence microscopy. By next generation sequencing, we identified two novel homozygous MAPKBP1 splice-site variants in patients with nephronophthisis-related chronic kidney disease. Splice-site analyses revealed truncation of C-terminal coiled-coil domains and patient-derived deletion constructs lost their ability to homodimerize and heterodimerize with paralogous WDR62. While wild-type MAPKBP1 exhibited centrosomal, basal body, and microtubule association, mutant proteins lost the latter and showed reduced recruitment to cell cycle dependent centriolar structures. Wild-type and mutant proteins had no reciprocal influence upon co-expression excluding dominant negative effects. Thus, MAPKBP1 appears to be a novel microtubule-binding protein with cell cycle dependent centriolar localization. Truncation of its coiled-coil domain is enough to abrogate its dimerization and results in severely disturbed intracellular localizations. Delineating the impact of impaired dimerization on cell cycle regulation and intracellular kidney signaling may provide new insights into common mechanisms of kidney degeneration. Thus, due to milder clinical presentation, MAPKBP1-associated nephronophthisis should be considered in adult patients with otherwise unexplained chronic kidney disease.


Assuntos
Centrossomo , Doenças Renais Policísticas , Adulto , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Centrossomo/metabolismo , Cílios/metabolismo , Dimerização , Fibrose , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas do Tecido Nervoso/metabolismo , Doenças Renais Policísticas/metabolismo
9.
Hum Mol Genet ; 27(2): 266-282, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29121203

RESUMO

A child presenting with Mainzer-Saldino syndrome (MZSDS), characterized by renal, retinal and skeletal involvements, was also diagnosed with lung infections and airway ciliary dyskinesia. These manifestations suggested dysfunction of both primary and motile cilia, respectively. Targeted exome sequencing identified biallelic mutations in WDR19, encoding an IFT-A subunit previously associated with MZSDS-related chondrodysplasia, Jeune asphyxiating thoracic dysplasia and cranioectodermal dysplasia, linked to primary cilia dysfunction, and in TEKT1 which encodes tektin-1 an uncharacterized member of the tektin family, mutations of which may cause ciliary dyskinesia. Tektin-1 localizes at the centrosome in cycling cells, at basal bodies of both primary and motile cilia and to the axoneme of motile cilia in airway cells. The identified mutations impaired these localizations. In addition, airway cells from the affected individual showed severe motility defects without major ultrastructural changes. Knockdown of tekt1 in zebrafish resulted in phenotypes consistent with a function for tektin-1 in ciliary motility, which was confirmed by live imaging. Finally, experiments in the zebrafish also revealed a synergistic effect of tekt1 and wdr19. Altogether, our data show genetic interactions between WDR19 and TEKT1 likely contributing to the overall clinical phenotype observed in the affected individual and provide strong evidence for TEKT1 as a new candidate gene for primary ciliary dyskinesia.


Assuntos
Cílios/genética , Ciliopatias/genética , Proteínas dos Microtúbulos/genética , Animais , Osso e Ossos/anormalidades , Ataxia Cerebelar/genética , Criança , Transtornos da Motilidade Ciliar/genética , Ciliopatias/metabolismo , Craniossinostoses/genética , Proteínas do Citoesqueleto , Displasia Ectodérmica/genética , Exoma , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas dos Microtúbulos/metabolismo , Mutação , Fenótipo , Proteínas/genética , Proteínas/metabolismo , Retinose Pigmentar/genética , Sequenciamento do Exoma , Peixe-Zebra/genética
10.
Hum Mol Genet ; 27(2): 224-238, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29077851

RESUMO

Genetic findings reported by our group and others showed that de novo missense variants in the KIF2A gene underlie malformations of brain development called pachygyria and microcephaly. Though KIF2A is known as member of the Kinesin-13 family involved in the regulation of microtubule end dynamics through its ATP dependent MT-depolymerase activity, how KIF2A variants lead to brain malformations is still largely unknown. Using cellular and in utero electroporation approaches, we show here that KIF2A disease-causing variants disrupts projection neuron positioning and interneuron migration, as well as progenitors proliferation. Interestingly, further dissection of this latter process revealed that ciliogenesis regulation is also altered during progenitors cell cycle. Altogether, our data suggest that deregulation of the coupling between ciliogenesis and cell cycle might contribute to the pathogenesis of KIF2A-related brain malformations. They also raise the issue whether ciliogenesis defects are a hallmark of other brain malformations, such as those related to tubulins and MT-motor proteins variants.


Assuntos
Cílios/genética , Cinesinas/metabolismo , Malformações do Desenvolvimento Cortical/genética , Proteínas Repressoras/metabolismo , Animais , Encéfalo/metabolismo , Ciclo Celular/genética , Cílios/fisiologia , Células HeLa , Humanos , Cinesinas/genética , Malformações do Desenvolvimento Cortical/metabolismo , Camundongos , Microcefalia/metabolismo , Microtúbulos/metabolismo , Neurogênese , Proteínas Repressoras/genética , Fuso Acromático/metabolismo , Tubulina (Proteína)/metabolismo
11.
Am J Hum Genet ; 100(2): 323-333, 2017 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-28089251

RESUMO

Nephronophthisis (NPH), an autosomal-recessive tubulointerstitial nephritis, is the most common cause of hereditary end-stage renal disease in the first three decades of life. Since most NPH gene products (NPHP) function at the primary cilium, NPH is classified as a ciliopathy. We identified mutations in a candidate gene in eight individuals from five families presenting late-onset NPH with massive renal fibrosis. This gene encodes MAPKBP1, a poorly characterized scaffolding protein for JNK signaling. Immunofluorescence analyses showed that MAPKBP1 is not present at the primary cilium and that fibroblasts from affected individuals did not display ciliogenesis defects, indicating that MAPKBP1 may represent a new family of NPHP not involved in cilia-associated functions. Instead, MAPKBP1 is recruited to mitotic spindle poles (MSPs) during the early phases of mitosis where it colocalizes with its paralog WDR62, which plays a key role at MSP. Detected mutations compromise recruitment of MAPKBP1 to the MSP and/or its interaction with JNK2 or WDR62. Additionally, we show increased DNA damage response signaling in fibroblasts from affected individuals and upon knockdown of Mapkbp1 in murine cell lines, a phenotype previously associated with NPH. In conclusion, we identified mutations in MAPKBP1 as a genetic cause of juvenile or late-onset and cilia-independent NPH.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/genética , Doenças Renais Císticas/congênito , Adolescente , Alelos , Animais , Proteínas de Ciclo Celular , Criança , Cílios/genética , Dano ao DNA/genética , Modelos Animais de Doenças , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibrose , Regulação da Expressão Gênica , Humanos , Rim/citologia , Rim/metabolismo , Doenças Renais Císticas/diagnóstico , Doenças Renais Císticas/genética , Falência Renal Crônica/diagnóstico , Falência Renal Crônica/genética , Camundongos , Camundongos Knockout , Mitose , Mutação , Células NIH 3T3 , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Linhagem , Fenótipo , Transdução de Sinais , Polos do Fuso/metabolismo , Adulto Jovem , Peixe-Zebra
12.
Biol Cell ; 111(4): 79-94, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30720881

RESUMO

Cilia and flagella are microtubule-based antenna which are highly conserved among eukaryotes. In vertebrates, primary and motile cilia have evolved to exert several key functions during development and tissue homoeostasis. Ciliary dysfunction in humans causes a highly heterogeneous group of diseases called ciliopathies, a class of genetic multisystemic disorders primarily affecting kidney, skeleton, retina, lung and the central nervous system. Among key ciliary proteins, kinesin family members (KIF) are microtubule-interacting proteins involved in many diverse cellular functions, including transport of cargo (organelles, proteins and lipids) along microtubules and regulating the dynamics of cytoplasmic and spindle microtubules through their depolymerising activity. Many KIFs are also involved in diverse ciliary functions including assembly/disassembly, motility and signalling. We here review these ciliary kinesins in vertebrates and focus on their involvement in ciliopathy-related disorders.


Assuntos
Cílios , Ciliopatias , Cinesinas , Animais , Transporte Biológico , Cílios/metabolismo , Cílios/patologia , Ciliopatias/metabolismo , Ciliopatias/patologia , Humanos , Cinesinas/classificação , Cinesinas/metabolismo , Cinesinas/fisiologia
13.
Biol Cell ; 111(9): 217-231, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31177551

RESUMO

Ciliopathies are complex genetic multi-system disorders causally related to abnormal assembly or function of motile or non-motile cilia. While most human cells possess a non-motile sensory/primary cilium (PC) during development and/or in adult tissues, motile cilia are restricted to specialised cells. As a result, PC-associated ciliopathies are characterised by high phenotypic variability with extensive clinical and genetic overlaps. In the present review, we have focused on cerebral developmental anomalies, which are commonly found in PC-associated ciliopathies and which have mostly been linked to Hedgehog signalling defects. In addition, we have reviewed emerging evidence that PC dysfunctions could be directly or indirectly involved in the mechanisms underlying malformations of cerebral cortical development including primary microcephaly.


Assuntos
Agenesia do Corpo Caloso/embriologia , Cerebelo/anormalidades , Cílios/patologia , Ciliopatias/embriologia , Hidrocefalia/embriologia , Malformações do Sistema Nervoso/embriologia , Defeitos do Tubo Neural/embriologia , Animais , Cerebelo/embriologia , Deficiências do Desenvolvimento , Proteínas Hedgehog/metabolismo , Humanos , Camundongos , Transdução de Sinais
14.
Nature ; 502(7472): 567-70, 2013 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-24097348

RESUMO

In most eukaryotic cells microtubules undergo post-translational modifications such as acetylation of α-tubulin on lysine 40, a widespread modification restricted to a subset of microtubules that turns over slowly. This subset of stable microtubules accumulates in cell protrusions and regulates cell polarization, migration and invasion. However, mechanisms restricting acetylation to these microtubules are unknown. Here we report that clathrin-coated pits (CCPs) control microtubule acetylation through a direct interaction of the α-tubulin acetyltransferase αTAT1 (refs 8, 9) with the clathrin adaptor AP2. We observe that about one-third of growing microtubule ends contact and pause at CCPs and that loss of CCPs decreases lysine 40 acetylation levels. We show that αTAT1 localizes to CCPs through a direct interaction with AP2 that is required for microtubule acetylation. In migrating cells, the polarized orientation of acetylated microtubules correlates with CCP accumulation at the leading edge, and interaction of αTAT1 with AP2 is required for directional migration. We conclude that microtubules contacting CCPs become acetylated by αTAT1. In migrating cells, this mechanism ensures the acetylation of microtubules oriented towards the leading edge, thus promoting directional cell locomotion and chemotaxis.


Assuntos
Acetiltransferases/metabolismo , Clatrina/metabolismo , Invaginações Revestidas da Membrana Celular/metabolismo , Microtúbulos/metabolismo , Acetilação , Complexo 2 de Proteínas Adaptadoras/metabolismo , Biocatálise , Movimento Celular , Invaginações Revestidas da Membrana Celular/enzimologia , Células HeLa , Humanos , Microtúbulos/química , Ligação Proteica , Tubulina (Proteína)/metabolismo
15.
PLoS Genet ; 12(3): e1005894, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26967905

RESUMO

Ciliopathies are a group of genetic multi-systemic disorders related to dysfunction of the primary cilium, a sensory organelle present at the cell surface that regulates key signaling pathways during development and tissue homeostasis. In order to identify novel genes whose mutations would cause severe developmental ciliopathies, >500 patients/fetuses were analyzed by a targeted high throughput sequencing approach allowing exome sequencing of >1200 ciliary genes. NEK8/NPHP9 mutations were identified in five cases with severe overlapping phenotypes including renal cystic dysplasia/hypodysplasia, situs inversus, cardiopathy with hypertrophic septum and bile duct paucity. These cases highlight a genotype-phenotype correlation, with missense and nonsense mutations associated with hypodysplasia and enlarged cystic organs, respectively. Functional analyses of NEK8 mutations in patient fibroblasts and mIMCD3 cells showed that these mutations differentially affect ciliogenesis, proliferation/apoptosis/DNA damage response, as well as epithelial morphogenesis. Notably, missense mutations exacerbated some of the defects due to NEK8 loss of function, highlighting their likely gain-of-function effect. We also showed that NEK8 missense and loss-of-function mutations differentially affect the regulation of the main Hippo signaling effector, YAP, as well as the expression of its target genes in patient fibroblasts and renal cells. YAP imbalance was also observed in enlarged spheroids of Nek8-invalidated renal epithelial cells grown in 3D culture, as well as in cystic kidneys of Jck mice. Moreover, co-injection of nek8 MO with WT or mutated NEK8-GFP RNA in zebrafish embryos led to shortened dorsally curved body axis, similar to embryos injected with human YAP RNA. Finally, treatment with Verteporfin, an inhibitor of YAP transcriptional activity, partially rescued the 3D spheroid defects of Nek8-invalidated cells and the abnormalities of NEK8-overexpressing zebrafish embryos. Altogether, our study demonstrates that NEK8 human mutations cause major organ developmental defects due to altered ciliogenesis and cell differentiation/proliferation through deregulation of the Hippo pathway.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Cílios/genética , Fosfoproteínas/genética , Doenças Renais Policísticas/genética , Proteínas Quinases/genética , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Animais , Diferenciação Celular/genética , Cílios/patologia , Feminino , Estudos de Associação Genética , Humanos , Rim/metabolismo , Rim/patologia , Camundongos , Morfogênese/genética , Mutação , Quinases Relacionadas a NIMA , Fosfoproteínas/antagonistas & inibidores , Fosfoproteínas/biossíntese , Doenças Renais Policísticas/patologia , Porfirinas/administração & dosagem , Transdução de Sinais , Fatores de Transcrição , Verteporfina , Proteínas de Sinalização YAP , Peixe-Zebra
16.
Biochim Biophys Acta Mol Basis Dis ; 1864(7): 2448-2457, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29660491

RESUMO

Interallelic interactions of membrane proteins are not taken into account while evaluating the pathogenicity of sequence variants in autosomal recessive disorders. Podocin, a membrane-anchored component of the slit diaphragm, is encoded by NPHS2, the major gene mutated in hereditary podocytopathies. We formerly showed that its R229Q variant is only pathogenic when trans-associated to specific 3' mutations and suggested the causal role of an abnormal C-terminal dimerization. Here we show by FRET analysis and size exclusion chromatography that podocin oligomerization occurs exclusively through the C-terminal tail (residues 283-382): principally through the first C-terminal helical region (H1, 283-313), which forms a coiled coil as shown by circular dichroism spectroscopy, and through the 332-348 region. We show the principal role of the oligomerization sites in mediating interallelic interactions: while the monomer-forming R286Tfs*17 podocin remains membranous irrespective of the coexpressed podocin variant identity, podocin variants with an intact H1 significantly influence each other's localization (r2 = 0.68, P = 9.2 × 10-32). The dominant negative effect resulting in intracellular retention of the pathogenic F344Lfs*4-R229Q heterooligomer occurs in parallel with a reduction in the FRET efficiency, suggesting the causal role of a conformational rearrangement. On the other hand, oligomerization can also promote the membrane localization: it can prevent the endocytosis of F344Lfs*4 or F344* podocin mutants induced by C-terminal truncation. In conclusion, C-terminal oligomerization of podocin can mediate both a dominant negative effect and interallelic complementation. Interallelic interactions of NPHS2 are not restricted to the R229Q variant and have to be considered in compound heterozygous individuals.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular , Nefropatias , Proteínas de Membrana , Mutação de Sentido Incorreto , Podócitos/metabolismo , Multimerização Proteica/genética , Substituição de Aminoácidos , Linhagem Celular Transformada , Transferência Ressonante de Energia de Fluorescência , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Nefropatias/genética , Nefropatias/metabolismo , Nefropatias/patologia , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Podócitos/patologia , Domínios Proteicos
17.
Traffic ; 16(7): 712-26, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25753619

RESUMO

Cystinosin is a lysosomal cystine transporter defective in cystinosis, an autosomal recessive lysosomal storage disorder. It is composed of seven transmembrane (TM) domains and contains two lysosomal targeting motifs: a tyrosine-based signal (GYDQL) in its C-terminal tail and a non-classical motif in its fifth inter-TM loop. Using the yeast two-hybrid system, we showed that the GYDQL motif specifically interacted with the µ subunit of the adaptor protein complex 3 (AP-3). Moreover, cell surface biotinylation and total internal reflection fluorescence microscopy revealed that cystinosin was partially mislocalized to the plasma membrane (PM) in AP-3-depleted cells. We generated a chimeric CD63 protein to specifically analyze the function of the GYDQL motif. This chimeric protein was targeted to lysosomes in a manner similar to cystinosin and was partially mislocalized to the PM in AP-3 knockdown cells where it also accumulated in the trans-Golgi network and early endosomes. Together with the fact that the surface levels of cystinosin and of the CD63-GYDQL chimeric protein were not increased when clathrin-mediated endocytosis was impaired, our data show that the tyrosine-based motif of cystinosin is a 'strong' AP-3 interacting motif responsible for lysosomal targeting of cystinosin by a direct intracellular pathway.


Assuntos
Complexo 3 de Proteínas Adaptadoras/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Lisossomos/metabolismo , Sinais Direcionadores de Proteínas , Sistemas de Transporte de Aminoácidos Neutros/química , Endocitose , Células HeLa , Humanos , Transporte Proteico , Tetraspanina 30/metabolismo
18.
Am J Hum Genet ; 94(2): 288-94, 2014 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-24439109

RESUMO

Renal hypodysplasia (RHD) is a heterogeneous condition encompassing a spectrum of kidney development defects including renal agenesis, hypoplasia, and (cystic) dysplasia. Heterozygous mutations of several genes have been identified as genetic causes of RHD with various severity. However, these genes and mutations are not associated with bilateral renal agenesis, except for RET mutations, which could be involved in a few cases. The pathophysiological mechanisms leading to total absence of kidney development thus remain largely elusive. By using a whole-exome sequencing approach in families with several fetuses with bilateral renal agenesis, we identified recessive mutations in the integrin α8-encoding gene ITGA8 in two families. Itga8 homozygous knockout in mice is known to result in absence of kidney development. We provide evidence of a damaging effect of the human ITGA8 mutations. These results demonstrate that mutations of ITGA8 are a genetic cause of bilateral renal agenesis and that, at least in some cases, bilateral renal agenesis is an autosomal-recessive disease.


Assuntos
Anormalidades Congênitas/genética , Genes Recessivos , Cadeias alfa de Integrinas/genética , Nefropatias/congênito , Rim/anormalidades , Anormalidades Urogenitais/genética , Anormalidades Congênitas/patologia , Feminino , Feto/anormalidades , Homozigoto , Humanos , Cadeias alfa de Integrinas/metabolismo , Rim/patologia , Nefropatias/genética , Nefropatias/patologia , Masculino , Mutação , Linhagem , Anormalidades Urogenitais/patologia
19.
Am J Hum Genet ; 94(6): 905-14, 2014 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-24882706

RESUMO

Ciliopathies are a group of hereditary disorders associated with defects in cilia structure and function. The distal appendages (DAPs) of centrioles are involved in the docking and anchoring of the mother centriole to the cellular membrane during ciliogenesis. The molecular composition of DAPs was recently elucidated and mutations in two genes encoding DAPs components (CEP164/NPHP15, SCLT1) have been associated with human ciliopathies, namely nephronophthisis and orofaciodigital syndrome. To identify additional DAP components defective in ciliopathies, we independently performed targeted exon sequencing of 1,221 genes associated with cilia and 5 known DAP protein-encoding genes in 1,255 individuals with a nephronophthisis-related ciliopathy. We thereby detected biallelic mutations in a key component of DAP-encoding gene, CEP83, in seven families. All affected individuals had early-onset nephronophthisis and four out of eight displayed learning disability and/or hydrocephalus. Fibroblasts and tubular renal cells from affected individuals showed an altered DAP composition and ciliary defects. In summary, we have identified mutations in CEP83, another DAP-component-encoding gene, as a cause of infantile nephronophthisis associated with central nervous system abnormalities in half of the individuals.


Assuntos
Deficiência Intelectual/genética , Doenças Renais Císticas/genética , Proteínas Associadas aos Microtúbulos/genética , Mutação , Alelos , Sistema Nervoso Central/anormalidades , Centríolos/genética , Centríolos/metabolismo , Pré-Escolar , Cílios/metabolismo , Éxons , Feminino , Humanos , Lactente , Masculino , Proteínas Associadas aos Microtúbulos/metabolismo , Síndromes Orofaciodigitais/genética
20.
EMBO J ; 30(13): 2557-68, 2011 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-21642958

RESUMO

The tumour suppressor PTEN (phosphatase and tensin deleted on chromosome 10) regulates major cellular functions via lipid phosphatase-dependent and -independent mechanisms. Despite its fundamental pathophysiological importance, how PTEN's cellular activity is regulated has only been partially elucidated. We report that the scaffolding proteins ß-arrestins (ß-arrs) are important regulators of PTEN. Downstream of receptor-activated RhoA/ROCK signalling, ß-arrs activate the lipid phosphatase activity of PTEN to negatively regulate Akt and cell proliferation. In contrast, following wound-induced RhoA activation, ß-arrs inhibit the lipid phosphatase-independent anti-migratory effects of PTEN. ß-arrs can thus differentially control distinct functional outputs of PTEN important for cell proliferation and migration.


Assuntos
Arrestinas/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , PTEN Fosfo-Hidrolase/fisiologia , Animais , Arrestinas/antagonistas & inibidores , Arrestinas/genética , Arrestinas/fisiologia , Células COS , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Chlorocebus aethiops , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Camundongos , PTEN Fosfo-Hidrolase/genética , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Ligação Proteica/fisiologia , RNA Interferente Pequeno/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , beta-Arrestinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA