Your browser doesn't support javascript.
loading
AMBRA1 p.Gln30Arg Mutation, Identified in a Cowden Syndrome Family, Exhibits Hyperproliferative Potential in hTERT-RPE1 Cells.
Revathidevi, Sundaramoorthy; Hosomichi, Kazuyoshi; Natsume, Toyoaki; Nakaoka, Hirofumi; Fujito, Naoko T; Akatsuka, Hisako; Sato, Takehito; Munirajan, Arasambattu Kannan; Inoue, Ituro.
Afiliação
  • Revathidevi S; Human Genetics Laboratory, National Institute of Genetics, Mishima 411-8540, Japan.
  • Hosomichi K; Laboratory of Computational Genomics, Tokyo University of Pharmacy and Life Sciences, Hachioji 192-0392, Japan.
  • Natsume T; Molecular Cell Engineering Laboratory, National Institute of Genetics, Mishima 411-8540, Japan.
  • Nakaoka H; Research Center for Genome & Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan.
  • Fujito NT; Department of Cancer Genome Research, Sasaki Institute, Sasaki Foundation, Chiyoda 101-0062, Japan.
  • Akatsuka H; Human Genetics Laboratory, National Institute of Genetics, Mishima 411-8540, Japan.
  • Sato T; Division of Host Defense Mechanism, Tokai University School of Medicine, Isehara 259-1193, Japan.
  • Munirajan AK; Division of Host Defense Mechanism, Tokai University School of Medicine, Isehara 259-1193, Japan.
  • Inoue I; Department of Genetics, Dr. ALM PG Institute of Basic Medical Sciences, University of Madras, Taramani, Chennai 600113, India.
Int J Mol Sci ; 23(19)2022 Sep 22.
Article em En | MEDLINE | ID: mdl-36232425
ABSTRACT
Cowden syndrome (CS) is a rare autosomal dominant disorder associated with multiple hamartomatous and neoplastic lesions in various organs. Most CS patients have been found to have germline mutations in the PTEN tumor suppressor. In the present study, we investigated the causative gene of CS in a family of PTEN (phosphatase and tensin homolog deleted on chromosome 10) -negative CS patients. Whole exome sequencing analysis revealed AMBRA1 (Autophagy and Beclin 1 Regulator 1) as a novel candidate gene harboring two germline variants p.Gln30Arg (Q30R) and p.Arg1195Ser (R1195S). AMBRA1 is a key regulator of the autophagy signaling network and a tumor suppressor. To functionally validate the role of AMBRA1 in the clinical manifestations of CS, we generated AMBRA1 depletion and Q30R mutation in hTERT-RPE1 (humanTelomerase Reverse Transcriptase-immortalized Retinal Pigmented Epithelial cells) using the CRISPR-Cas9 gene editing system. We observed that both AMBRA1-depleted and mutant cells showed accumulation in the S phase, leading to hyperproliferation, which is a characteristic of hamartomatous lesions. Specifically, the AMBRA1 Q30R mutation disturbed the G1/S transition of cells, leading to continuous mitotic entry of mutant cells, irrespective of the extracellular condition. From our analysis of primary ciliogenesis in these cells, we speculated that the mitotic entry of AMBRA1 Q30R mutants could be due to non-functional primary cilia that lead to impaired processing of extracellular sensory signals. Additionally, we observed a situs inversus phenotype in ambra1-depleted zebrafish, a developmental abnormality resulting from dysregulated primary ciliogenesis. Taken together, we established that the AMBRA1 Q30R mutation that we observed in CS patients might play an important role in inducing the hyperproliferative potential of cells through regulating primary ciliogenesis.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Síndrome do Hamartoma Múltiplo Tipo de estudo: Prognostic_studies Limite: Animals Idioma: En Ano de publicação: 2022 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Síndrome do Hamartoma Múltiplo Tipo de estudo: Prognostic_studies Limite: Animals Idioma: En Ano de publicação: 2022 Tipo de documento: Article