Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 9 de 9
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Elife ; 102021 02 02.
Article de Anglais | MEDLINE | ID: mdl-33527896

RÉSUMÉ

Recent genomic and scRNA-seq analyses of melanoma demonstrated a lack of recurrent genetic drivers of metastasis, while identifying common transcriptional states correlating with invasion or drug resistance. To test whether transcriptional adaptation can drive melanoma progression, we made use of a zebrafish mitfa:BRAFV600E;tp53-/- model, in which malignant progression is characterized by minimal genetic evolution. We undertook an overexpression-screen of 80 epigenetic/transcriptional regulators and found neural crest-mesenchyme developmental regulator SATB2 to accelerate aggressive melanoma development. Its overexpression induces invadopodia formation and invasion in zebrafish tumors and human melanoma cell lines. SATB2 binds and activates neural crest-regulators, including pdgfab and snai2. The transcriptional program induced by SATB2 overlaps with known MITFlowAXLhigh and AQP1+NGFR1high drug-resistant states and functionally drives enhanced tumor propagation and resistance to Vemurafenib in vivo. In summary, we show that melanoma transcriptional rewiring by SATB2 to a neural crest mesenchyme-like program can drive invasion and drug resistance in autochthonous tumors.


Sujet(s)
Résistance aux médicaments antinéoplasiques/génétique , Protéines de liaison aux séquences d'ADN MAR/métabolisme , Mélanome/génétique , Invasion tumorale/génétique , Facteurs de transcription/métabolisme , Protéines de poisson-zèbre/métabolisme , Animaux , Systèmes CRISPR-Cas , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Régulation de l'expression des gènes tumoraux , Humains , Protéines de liaison aux séquences d'ADN MAR/génétique , Mélanome/traitement médicamenteux , Mélanome/métabolisme , Crête neurale/cytologie , Facteurs de transcription/génétique , Danio zébré , Protéines de poisson-zèbre/génétique
2.
Cells ; 9(4)2020 04 19.
Article de Anglais | MEDLINE | ID: mdl-32325871

RÉSUMÉ

A common feature of tumorigenesis is the upregulation of angiogenesis pathways in order to supply nutrients via the blood for the growing tumor. Understanding how cells promote angiogenesis and how to control these processes pharmaceutically are of great clinical interest. Clear cell renal cell carcinoma (ccRCC) is the most common form of sporadic and inherited kidney cancer which is associated with excess neovascularization. ccRCC is highly associated with biallelic mutations in the von Hippel-Lindau (VHL) tumor suppressor gene. Although upregulation of the miR-212/132 family and disturbed VHL signaling have both been linked with angiogenesis, no evidence of a possible connection between the two has yet been made. We show that miRNA-212/132 levels are increased after loss of functional pVHL, the protein product of the VHL gene, in vivo and in vitro. Furthermore, we show that blocking miRNA-212/132 with anti-miRs can significantly alleviate the excessive vascular branching phenotype characteristic of vhl-/- mutant zebrafish. Moreover, using human umbilical vascular endothelial cells (HUVECs) and an endothelial cell/pericyte coculture system, we observed that VHL knockdown promotes endothelial cells neovascularization capacity in vitro, an effect which can be inhibited by anti-miR-212/132 treatment. Taken together, our results demonstrate an important role for miRNA-212/132 in angiogenesis induced by loss of VHL. Intriguingly, this also presents a possibility for the pharmaceutical manipulation of angiogenesis by modulating levels of MiR212/132.


Sujet(s)
Régulation de l'expression des gènes tumoraux/génétique , microARN/génétique , Protéine Von Hippel-Lindau supresseur de tumeur/génétique , Agents angiogéniques/métabolisme , Néphrocarcinome/génétique , Néphrocarcinome/anatomopathologie , Lignée cellulaire tumorale , Cellules endothéliales/métabolisme , Humains , Rein/anatomopathologie , Tumeurs du rein/génétique , Tumeurs du rein/anatomopathologie , Régulation positive
3.
Nephron ; 138(4): 310-323, 2018.
Article de Anglais | MEDLINE | ID: mdl-29342457

RÉSUMÉ

BACKGROUND: von Hippel-Lindau (VHL) disease is characterized by the development of benign and malignant tumours in many organ systems, including renal cysts and clear cell renal cell carcinoma. It is not completely understood what underlies the development of renal pathology, and the use of murine Vhl models has been challenging due to limitations in disease conservation. We previously described a zebrafish model bearing inactivating mutations in the orthologue of the human VHL gene. METHODS: We used histopathological and functional assays to investigate the pronephric and glomerular developmental defects in vhl mutant zebrafish, supported by human cell culture assays. RESULTS: Here, we report that vhl is required to maintain pronephric tubule and glomerulus integrity in zebrafish embryos. vhl mutant glomeruli are enlarged, cxcr4a+ capillary loops are dilated and the Bowman space is widened. While we did not observe pronephric cysts, the cells of the proximal convoluted and anterior proximal straight tubule are enlarged, periodic acid schiff (PAS) and Oil Red O positive, and display a clear cytoplasm after hematoxylin and eosine staining. Ultrastructural analysis showed the vhl-/- tubule to accumulate large numbers of vesicles of variable size and electron density. Microinjection of the endocytic fluorescent marker AM1-43 in zebrafish embryos revealed an accumulation of endocytic vesicles in the vhl mutant pronephric tubule, which we can recapitulate in human cells lacking VHL. CONCLUSIONS: Our data indicates that vhl is required to maintain pronephric tubule and glomerulus integrity during zebrafish development, and suggests a role for VHL in endocytic vesicle trafficking.


Sujet(s)
Glomérule rénal/métabolisme , Tubules contournés proximaux/métabolisme , Protéines suppresseurs de tumeurs/génétique , Protéines de poisson-zèbre/génétique , Danio zébré/physiologie , Animaux , Développement embryonnaire/génétique , Glomérule rénal/malformations , Glomérule rénal/croissance et développement , Tubules contournés proximaux/malformations , Tubules contournés proximaux/croissance et développement , Larve , Mutation , Récepteurs aux facteurs de croissance endothéliale vasculaire/antagonistes et inhibiteurs
4.
Hum Mol Genet ; 27(3): 529-545, 2018 02 01.
Article de Anglais | MEDLINE | ID: mdl-29228333

RÉSUMÉ

DNAAF1 (LRRC50) is a cytoplasmic protein required for dynein heavy chain assembly and cilia motility, and DNAAF1 mutations cause primary ciliary dyskinesia (PCD; MIM 613193). We describe four families with DNAAF1 mutations and complex congenital heart disease (CHD). In three families, all affected individuals have typical PCD phenotypes. However, an additional family demonstrates isolated CHD (heterotaxy) in two affected siblings, but no clinical evidence of PCD. We identified a homozygous DNAAF1 missense mutation, p.Leu191Phe, as causative for heterotaxy in this family. Genetic complementation in dnaaf1-null zebrafish embryos demonstrated the rescue of normal heart looping with wild-type human DNAAF1, but not the p.Leu191Phe variant, supporting the conserved pathogenicity of this DNAAF1 missense mutation. This observation points to a phenotypic continuum between CHD and PCD, providing new insights into the pathogenesis of isolated CHD. In further investigations of the function of DNAAF1 in dynein arm assembly, we identified interactions with members of a putative dynein arm assembly complex. These include the ciliary intraflagellar transport protein IFT88 and the AAA+ (ATPases Associated with various cellular Activities) family proteins RUVBL1 (Pontin) and RUVBL2 (Reptin). Co-localization studies support these findings, with the loss of RUVBL1 perturbing the co-localization of DNAAF1 with IFT88. We show that RUVBL1 orthologues have an asymmetric left-sided distribution at both the mouse embryonic node and the Kupffer's vesicle in zebrafish embryos, with the latter asymmetry dependent on DNAAF1. These results suggest that DNAAF1-RUVBL1 biochemical and genetic interactions have a novel functional role in symmetry breaking and cardiac development.


Sujet(s)
ATPases associated with diverse cellular activities/métabolisme , Protéines de transport/métabolisme , Cils vibratiles/métabolisme , Helicase/métabolisme , Protéines associées aux microtubules/métabolisme , ATPases associated with diverse cellular activities/génétique , Animaux , Protéines de transport/génétique , Cils vibratiles/physiologie , Helicase/génétique , Femelle , Génotype , Cellules HEK293 , Humains , Mâle , Protéines associées aux microtubules/génétique , Mutation faux-sens/génétique , Pedigree , Phénotype , Protéines suppresseurs de tumeurs/génétique , Protéines suppresseurs de tumeurs/métabolisme , /méthodes , Danio zébré , Protéines de poisson-zèbre/génétique , Protéines de poisson-zèbre/métabolisme
5.
Circ Res ; 120(8): 1263-1275, 2017 Apr 14.
Article de Anglais | MEDLINE | ID: mdl-28179432

RÉSUMÉ

RATIONALE: Lymphatic vessel formation and function constitutes a physiologically and pathophysiologically important process, but its genetic control is not well understood. OBJECTIVE: Here, we identify the secreted Polydom/Svep1 protein as essential for the formation of the lymphatic vasculature. We analyzed mutants in mice and zebrafish to gain insight into the role of Polydom/Svep1 in the lymphangiogenic process. METHODS AND RESULTS: Phenotypic analysis of zebrafish polydom/svep1 mutants showed a decrease in venous and lymphovenous sprouting, which leads to an increased number of intersegmental arteries. A reduced number of primordial lymphatic cells populated the horizontal myoseptum region but failed to migrate dorsally or ventrally, resulting in severe reduction of the lymphatic trunk vasculature. Corresponding mutants in the mouse Polydom/Svep1 gene showed normal egression of Prox-1+ cells from the cardinal vein at E10.5, but at E12.5, the tight association between the cardinal vein and lymphatic endothelial cells at the first lymphovenous contact site was abnormal. Furthermore, mesenteric lymphatic structures at E18.5 failed to undergo remodeling events in mutants and lacked lymphatic valves. In both fish and mouse embryos, the expression of the gene suggests a nonendothelial and noncell autonomous mechanism. CONCLUSIONS: Our data identify zebrafish and mouse Polydom/Svep1 as essential extracellular factors for lymphangiogenesis. Expression of the respective genes by mesenchymal cells in intimate proximity with venous and lymphatic endothelial cells is required for sprouting and migratory events in zebrafish and for remodeling events of the lymphatic intraluminal valves in mouse embryos.


Sujet(s)
Cellules endothéliales/métabolisme , Évolution moléculaire , Lymphangiogenèse , Vaisseaux lymphatiques/métabolisme , Protéines/métabolisme , Protéines de poisson-zèbre/métabolisme , Danio zébré/métabolisme , Animaux , Animal génétiquement modifié , Protéines de liaison au calcium , Molécules d'adhérence cellulaire , Communication cellulaire , Mouvement cellulaire , Cellules endothéliales/anatomopathologie , Endothélium lymphatique/malformations , Endothélium lymphatique/métabolisme , Endothélium lymphatique/physiopathologie , Régulation de l'expression des gènes au cours du développement , Génotype , Vaisseaux lymphatiques/malformations , Vaisseaux lymphatiques/physiopathologie , Mésoderme/métabolisme , Mutation , Phénotype , Protéines/génétique , Transduction du signal , Facteurs temps , Danio zébré/génétique , Protéines de poisson-zèbre/génétique
6.
Hum Genet ; 134(8): 905-16, 2015 Aug.
Article de Anglais | MEDLINE | ID: mdl-26026792

RÉSUMÉ

Congenital anomalies of the kidney and urinary tract (CAKUT) account for 40-50% of chronic kidney disease that manifests in the first two decades of life. Thus far, 31 monogenic causes of isolated CAKUT have been described, explaining ~12% of cases. To identify additional CAKUT-causing genes, we performed whole-exome sequencing followed by a genetic burden analysis in 26 genetically unsolved families with CAKUT. We identified two heterozygous mutations in SRGAP1 in 2 unrelated families. SRGAP1 is a small GTPase-activating protein in the SLIT2-ROBO2 signaling pathway, which is essential for development of the metanephric kidney. We then examined the pathway-derived candidate gene SLIT2 for mutations in cohort of 749 individuals with CAKUT and we identified 3 unrelated individuals with heterozygous mutations. The clinical phenotypes of individuals with mutations in SLIT2 or SRGAP1 were cystic dysplastic kidneys, unilateral renal agenesis, and duplicated collecting system. We show that SRGAP1 is expressed in early mouse nephrogenic mesenchyme and that it is coexpressed with ROBO2 in SIX2-positive nephron progenitor cells of the cap mesenchyme in developing rat kidney. We demonstrate that the newly identified mutations in SRGAP1 lead to an augmented inhibition of RAC1 in cultured human embryonic kidney cells and that the SLIT2 mutations compromise the ability of the SLIT2 ligand to inhibit cell migration. Thus, we report on two novel candidate genes for causing monogenic isolated CAKUT in humans.


Sujet(s)
Protéines d'activation de la GTPase , Protéines et peptides de signalisation intercellulaire , Mutation , Protéines de tissu nerveux , Récepteurs immunologiques , Transduction du signal/génétique , Malformations urogénitales , Reflux vésico-urétéral , Animaux , Exome , Protéines d'activation de la GTPase/biosynthèse , Protéines d'activation de la GTPase/génétique , Cellules HEK293 , Humains , Protéines et peptides de signalisation intercellulaire/biosynthèse , Protéines et peptides de signalisation intercellulaire/métabolisme , Mésoderme/métabolisme , Souris , Protéines de tissu nerveux/biosynthèse , Protéines de tissu nerveux/métabolisme , Rats , Récepteurs immunologiques/biosynthèse , Récepteurs immunologiques/génétique , Récepteurs immunologiques/métabolisme , Facteurs de risque , Malformations urogénitales/embryologie , Malformations urogénitales/génétique , Reflux vésico-urétéral/embryologie , Reflux vésico-urétéral/génétique
7.
Nephron ; 129(1): 62-7, 2015.
Article de Anglais | MEDLINE | ID: mdl-25531169

RÉSUMÉ

Congenital anomalies of the kidney and urinary tract (CAKUT) constitute one of the most common developmental diseases in humans; however, the cause for most patients remains unknown. Efforts to identify novel genetic causes for CAKUT through next-generation sequencing techniques have led to the discovery of new genes and risk factors. Concomitantly, these same efforts have generated large gene candidate lists requiring individual functional characterization. Appropriate model systems are needed to assess the functionality of genes and pathogenicity of genetic variants discovered in CAKUT patients. In this review, we discuss how cellular, animal, and personal (human) models are being used to study CAKUT candidate genes and what their major advantages and disadvantages are with respect to relevance and throughput.


Sujet(s)
Rein/malformations , Modèles biologiques , Voies urinaires/malformations , Animaux , Variation génétique , Séquençage nucléotidique à haut débit , Humains , Techniques in vitro , Souris , Modèles animaux , Mutation , Médecine de précision , Protéomique , Facteurs de risque , Cellules souches/cytologie , Danio zébré
8.
PLoS Genet ; 10(10): e1004594, 2014 Oct.
Article de Anglais | MEDLINE | ID: mdl-25340510

RÉSUMÉ

We recently reported that centrosomal protein 164 (CEP164) regulates both cilia and the DNA damage response in the autosomal recessive polycystic kidney disease nephronophthisis. Here we examine the functional role of CEP164 in nephronophthisis-related ciliopathies and concomitant fibrosis. Live cell imaging of RPE-FUCCI (fluorescent, ubiquitination-based cell cycle indicator) cells after siRNA knockdown of CEP164 revealed an overall quicker cell cycle than control cells, although early S-phase was significantly longer. Follow-up FACS experiments with renal IMCD3 cells confirm that Cep164 siRNA knockdown promotes cells to accumulate in S-phase. We demonstrate that this effect can be rescued by human wild-type CEP164, but not disease-associated mutants. siRNA of CEP164 revealed a proliferation defect over time, as measured by CyQuant assays. The discrepancy between accelerated cell cycle and inhibited overall proliferation could be explained by induction of apoptosis and epithelial-to-mesenchymal transition. Reduction of CEP164 levels induces apoptosis in immunofluorescence, FACS and RT-QPCR experiments. Furthermore, knockdown of Cep164 or overexpression of dominant negative mutant allele CEP164 Q525X induces epithelial-to-mesenchymal transition, and concomitant upregulation of genes associated with fibrosis. Zebrafish injected with cep164 morpholinos likewise manifest developmental abnormalities, impaired DNA damage signaling, apoptosis and a pro-fibrotic response in vivo. This study reveals a novel role for CEP164 in the pathogenesis of nephronophthisis, in which mutations cause ciliary defects coupled with DNA damage induced replicative stress, cell death, and epithelial-to-mesenchymal transition, and suggests that these events drive the characteristic fibrosis observed in nephronophthisis kidneys.


Sujet(s)
Cils vibratiles/génétique , Fibrose/génétique , Maladies kystiques rénales/génétique , Protéines microtubulaires/génétique , Animaux , Apoptose/génétique , Cycle cellulaire/génétique , Cils vibratiles/anatomopathologie , Altération de l'ADN/génétique , Transition épithélio-mésenchymateuse , Fibrose/anatomopathologie , Techniques de knock-down de gènes , Humains , Maladies kystiques rénales/anatomopathologie , Protéines microtubulaires/biosynthèse , Petit ARN interférent , Transduction du signal , Danio zébré
9.
Mol Cell Biol ; 30(14): 3620-34, 2010 Jul.
Article de Anglais | MEDLINE | ID: mdl-20479124

RÉSUMÉ

The lymphatic vascular system maintains tissue fluid homeostasis, helps mediate afferent immune responses, and promotes cancer metastasis. To address the role microRNAs (miRNAs) play in the development and function of the lymphatic vascular system, we defined the in vitro miRNA expression profiles of primary human lymphatic endothelial cells (LECs) and blood vascular endothelial cells (BVECs) and identified four BVEC signature and two LEC signature miRNAs. Their vascular lineage-specific expression patterns were confirmed in vivo by quantitative real-time PCR and in situ hybridization. Functional characterization of the BVEC signature miRNA miR-31 identified a novel BVEC-specific posttranscriptional regulatory mechanism that inhibits the expression of lymphatic lineage-specific transcripts in vitro. We demonstrate that suppression of lymphatic differentiation is partially mediated via direct repression of PROX1, a transcription factor that functions as a master regulator of lymphatic lineage-specific differentiation. Finally, in vivo studies of Xenopus and zebrafish demonstrated that gain of miR-31 function impaired venous sprouting and lymphatic vascular development, thus highlighting the importance of miR-31 as a negative regulator of lymphatic development. Collectively, our findings identify miR-31 is a potent regulator of vascular lineage-specific differentiation and development in vertebrates.


Sujet(s)
Cellules endothéliales/cytologie , Cellules endothéliales/métabolisme , Système lymphatique/cytologie , Système lymphatique/croissance et développement , microARN/génétique , Animaux , Animal génétiquement modifié , Séquence nucléotidique , Vaisseaux sanguins/cytologie , Vaisseaux sanguins/croissance et développement , Vaisseaux sanguins/métabolisme , Différenciation cellulaire/génétique , Différenciation cellulaire/physiologie , Cellules cultivées , Femelle , Analyse de profil d'expression de gènes , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Humains , Hybridation in situ , Techniques in vitro , Système lymphatique/métabolisme , Souris , microARN/métabolisme , Réaction de polymérisation en chaîne , Maturation post-transcriptionnelle des ARN , ARN messager/génétique , ARN messager/métabolisme , Protéines suppresseurs de tumeurs/génétique , Protéines suppresseurs de tumeurs/métabolisme , Xenopus laevis/embryologie , Xenopus laevis/génétique , Xenopus laevis/métabolisme , Danio zébré/embryologie , Danio zébré/génétique , Danio zébré/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE