Your browser doesn't support javascript.
loading
Mutations in GREB1L Cause Bilateral Kidney Agenesis in Humans and Mice.
De Tomasi, Lara; David, Pierre; Humbert, Camille; Silbermann, Flora; Arrondel, Christelle; Tores, Frédéric; Fouquet, Stéphane; Desgrange, Audrey; Niel, Olivier; Bole-Feysot, Christine; Nitschké, Patrick; Roume, Joëlle; Cordier, Marie-Pierre; Pietrement, Christine; Isidor, Bertrand; Khau Van Kien, Philippe; Gonzales, Marie; Saint-Frison, Marie-Hélène; Martinovic, Jelena; Novo, Robert; Piard, Juliette; Cabrol, Christelle; Verma, Ishwar C; Puri, Ratna; Journel, Hubert; Aziza, Jacqueline; Gavard, Laurent; Said-Menthon, Marie-Hélène; Heidet, Laurence; Saunier, Sophie; Jeanpierre, Cécile.
Afiliação
  • De Tomasi L; Laboratory of Hereditary Kidney Diseases, INSERM UMR 1163, Imagine Institute, 75015 Paris, France; Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, 75015 Paris, France; Paris Diderot University, 75013 Paris, France.
  • David P; Transgenesis Platform, Laboratoire d'Expérimentation Animale et Transgenèse (LEAT), Imagine Institute, Structure Fédérative de Recherche Necker INSERM US24/CNRS UMS3633, 75015 Paris, France.
  • Humbert C; Laboratory of Hereditary Kidney Diseases, INSERM UMR 1163, Imagine Institute, 75015 Paris, France; Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, 75015 Paris, France.
  • Silbermann F; Laboratory of Hereditary Kidney Diseases, INSERM UMR 1163, Imagine Institute, 75015 Paris, France; Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, 75015 Paris, France.
  • Arrondel C; Laboratory of Hereditary Kidney Diseases, INSERM UMR 1163, Imagine Institute, 75015 Paris, France; Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, 75015 Paris, France.
  • Tores F; Bioinformatic Platform, INSERM UMR 1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, 75015 Paris, France.
  • Fouquet S; Imaging Platform, Sorbonne Universités, UPMC Univ Paris 06, INSERM UMR_S968 and CNRS UMR7210, Institut de la Vision, 75012 Paris, France.
  • Desgrange A; Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, 75015 Paris, France; Laboratory of Heart Morphogenesis, INSERM UMR 1163, Imagine Institute-Pasteur Institute, 75015 Paris, France.
  • Niel O; APHP, Pediatric Nephrology Department, Hôpital Robert Debré, 75019 Paris, France.
  • Bole-Feysot C; Genomic Platform, INSERM UMR 1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, 75015 Paris, France.
  • Nitschké P; Bioinformatic Platform, INSERM UMR 1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, 75015 Paris, France.
  • Roume J; Unité de Génétique Médicale, Centre de Référence des Maladies rares du Développement (AnD DI Rares), CHI Poissy - St Germain en Laye, 78300 Poissy, France.
  • Cordier MP; Service de Génétique, Groupement Hospitalier Est, 69677 Bron, France.
  • Pietrement C; Unité de Néphrologie Pédiatrique, CHU Reims, 51100 Reims, France.
  • Isidor B; Service de Génétique Médicale, CHU Nantes, 44093 Nantes, France.
  • Khau Van Kien P; Unité de Génétique Médicale, Nîmes University Hospital, CHU Carémeau, 30900 Nîmes, France.
  • Gonzales M; APHP, Département de Génétique Médicale, Hôpital Armand Trousseau, Université Pierre et Marie Curie, 75571 Paris, France and APHP, Unité d'EmbryoFœtopathologie, Service d'Histologie-Embryologie-Cytogénétique, Hôpital Necker-Enfants Malades, 75015 Paris, France.
  • Saint-Frison MH; APHP, Département de Génétique, Unité de Fœtopathologie, Hôpital Robert Debré, 75019 Paris, France.
  • Martinovic J; APHP, Unit of Fetal Pathology, Antoine Béclère Hospital, 92140 Clamart, France.
  • Novo R; Centre Hospitalier Universitaire de Lille, Hôpital Jeanne de Flandre, Service de Néphrologie Pédiatrique, 59000 Lille, France.
  • Piard J; Centre de Génétique Humaine, CHU Besançon, Université de Franche-Comté, 25000 Besançon, France.
  • Cabrol C; Centre de Génétique Humaine, CHU Besançon, Université de Franche-Comté, 25000 Besançon, France.
  • Verma IC; Institute of Medical Genetics and Genomics, Sir Ganga Ram Hospital, New Delhi 110060, India.
  • Puri R; Institute of Medical Genetics and Genomics, Sir Ganga Ram Hospital, New Delhi 110060, India.
  • Journel H; Service de Génétique Médicale, Hôpital Chubert, 56000 Vannes, France.
  • Aziza J; Département d'Anatomie et Cytologie Pathologiques, IUCT-Oncopole, 31100 Toulouse, France.
  • Gavard L; Service de Gynécologie-Obstétrique, Hôpital Louis Mourier, 92700 Colombes, France.
  • Said-Menthon MH; Service de Pédiatrie, Hôpitaux du Léman, 74203 Thonon les Bains, France.
  • Heidet L; APHP, Centre de Référence des Maladies Rénales Héréditaires de l'Enfant et de l'Adulte (MARHEA), Hôpital Necker-Enfants Malades, 75015 Paris, France; APHP, Service de Néphrologie Pédiatrique, Hôpital Necker-Enfants Malades, 75015 Paris, France.
  • Saunier S; Laboratory of Hereditary Kidney Diseases, INSERM UMR 1163, Imagine Institute, 75015 Paris, France; Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, 75015 Paris, France.
  • Jeanpierre C; Laboratory of Hereditary Kidney Diseases, INSERM UMR 1163, Imagine Institute, 75015 Paris, France; Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, 75015 Paris, France. Electronic address: cecile.jeanpierre@inserm.fr.
Am J Hum Genet ; 101(5): 803-814, 2017 Nov 02.
Article em En | MEDLINE | ID: mdl-29100091
ABSTRACT
Congenital anomalies of the kidney and urinary tract (CAKUT) constitute a major cause of chronic kidney disease in children and 20% of prenatally detected anomalies. CAKUT encompass a spectrum of developmental kidney defects, including renal agenesis, hypoplasia, and cystic and non-cystic dysplasia. More than 50 genes have been reported as mutated in CAKUT-affected case subjects. However, the pathophysiological mechanisms leading to bilateral kidney agenesis (BKA) remain largely elusive. Whole-exome or targeted exome sequencing of 183 unrelated familial and/or severe CAKUT-affected case subjects, including 54 fetuses with BKA, led to the identification of 16 heterozygous variants in GREB1L (growth regulation by estrogen in breast cancer 1-like), a gene reported as a target of retinoic acid signaling. Four loss-of-function and 12 damaging missense variants, 14 being absent from GnomAD, were identified. Twelve of them were present in familial or simplex BKA-affected case subjects. Female BKA-affected fetuses also displayed uterus agenesis. We demonstrated a significant association between GREB1L variants and BKA. By in situ hybridization, we showed expression of Greb1l in the nephrogenic zone in developing mouse kidney. We generated a Greb1l knock-out mouse model by CRISPR-Cas9. Analysis at E13.5 revealed lack of kidneys and genital tract anomalies in male and female Greb1l-/- embryos and a slight decrease in ureteric bud branching in Greb1l+/- embryos. We showed that Greb1l invalidation in mIMCD3 cells affected tubulomorphogenesis in 3D-collagen culture, a phenotype rescued by expression of the wild-type human protein. This demonstrates that GREB1L plays a major role in early metanephros and genital development in mice and humans.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Anormalidades Congênitas / Proteínas / Rim / Nefropatias / Mutação / Proteínas de Neoplasias Limite: Animals / Child / Female / Humans / Male Idioma: En Ano de publicação: 2017 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Anormalidades Congênitas / Proteínas / Rim / Nefropatias / Mutação / Proteínas de Neoplasias Limite: Animals / Child / Female / Humans / Male Idioma: En Ano de publicação: 2017 Tipo de documento: Article