Your browser doesn't support javascript.
loading
Lowe syndrome patient cells display mTOR- and RhoGTPase-dependent phenotypes alleviated by rapamycin and statins.
Madhivanan, Kayalvizhi; Ramadesikan, Swetha; Hsieh, Wen-Chieh; Aguilar, Mariana C; Hanna, Claudia B; Bacallao, Robert L; Aguilar, R Claudio.
Affiliation
  • Madhivanan K; Department of Biological Sciences, Purdue University, Hansen Life Sciences Building, Room 321, 201 S. University street, West Lafayette, IN 47907, USA.
  • Ramadesikan S; Department of Biological Sciences, Purdue University, Hansen Life Sciences Building, Room 321, 201 S. University street, West Lafayette, IN 47907, USA.
  • Hsieh WC; Department of Biological Sciences, Purdue University, Hansen Life Sciences Building, Room 321, 201 S. University street, West Lafayette, IN 47907, USA.
  • Aguilar MC; Department of Biological Sciences, Purdue University, Hansen Life Sciences Building, Room 321, 201 S. University street, West Lafayette, IN 47907, USA.
  • Hanna CB; Department of Biological Sciences, Purdue University, Hansen Life Sciences Building, Room 321, 201 S. University street, West Lafayette, IN 47907, USA.
  • Bacallao RL; Division of Nephrology, Indiana University School of Medicine, 340 W 10th St #6200, Indianapolis, IN 46202, USA.
  • Aguilar RC; Department of Biological Sciences, Purdue University, Hansen Life Sciences Building, Room 321, 201 S. University street, West Lafayette, IN 47907, USA.
Hum Mol Genet ; 29(10): 1700-1715, 2020 06 27.
Article in En | MEDLINE | ID: mdl-32391547
ABSTRACT
Lowe syndrome (LS) is an X-linked developmental disease characterized by cognitive deficiencies, bilateral congenital cataracts and renal dysfunction. Unfortunately, this disease leads to the early death of affected children often due to kidney failure. Although this condition was first described in the early 1950s and the affected gene (OCRL1) was identified in the early 1990s, its pathophysiological mechanism is not fully understood and there is no LS-specific cure available to patients. Here we report two important signaling pathways affected in LS patient cells. While RhoGTPase signaling abnormalities led to adhesion and spreading defects as compared to normal controls, PI3K/mTOR hyperactivation interfered with primary cilia assembly (scenario also observed in other ciliopathies with compromised kidney function). Importantly, we identified two FDA-approved drugs able to ameliorate these phenotypes. Specifically, statins mitigated adhesion and spreading abnormalities while rapamycin facilitated ciliogenesis in LS patient cells. However, no single drug was able to alleviate both phenotypes. Based on these and other observations, we speculate that Ocrl1 has dual, independent functions supporting proper RhoGTPase and PI3K/mTOR signaling. Therefore, this study suggest that Ocrl1-deficiency leads to signaling defects likely to require combinatorial drug treatment to suppress patient phenotypes and symptoms.
Subject(s)

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Phosphoric Monoester Hydrolases / Genetic Diseases, X-Linked / TOR Serine-Threonine Kinases / Oculocerebrorenal Syndrome Limits: Humans Language: En Journal: Hum Mol Genet Journal subject: BIOLOGIA MOLECULAR / GENETICA MEDICA Year: 2020 Document type: Article Affiliation country: United States

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Phosphoric Monoester Hydrolases / Genetic Diseases, X-Linked / TOR Serine-Threonine Kinases / Oculocerebrorenal Syndrome Limits: Humans Language: En Journal: Hum Mol Genet Journal subject: BIOLOGIA MOLECULAR / GENETICA MEDICA Year: 2020 Document type: Article Affiliation country: United States
...