Your browser doesn't support javascript.
loading
Early adolescent Rai1 reactivation reverses transcriptional and social interaction deficits in a mouse model of Smith-Magenis syndrome.
Huang, Wei-Hsiang; Wang, David C; Allen, William E; Klope, Matthew; Hu, Hailan; Shamloo, Mehrdad; Luo, Liqun.
Afiliação
  • Huang WH; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305; weihsiah@stanford.edu lluo@stanford.edu.
  • Wang DC; Department of Biology, Stanford University, Stanford, CA 94305.
  • Allen WE; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305.
  • Klope M; Department of Biology, Stanford University, Stanford, CA 94305.
  • Hu H; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305.
  • Shamloo M; Neurosciences Program, Stanford University, Stanford, CA 94305.
  • Luo L; Stanford Behavioral and Functional Neuroscience Laboratory, Stanford University School of Medicine, Stanford, CA 94305.
Proc Natl Acad Sci U S A ; 115(42): 10744-10749, 2018 10 16.
Article em En | MEDLINE | ID: mdl-30275311
ABSTRACT
Haploinsufficiency of Retinoic Acid Induced 1 (RAI1) causes Smith-Magenis syndrome (SMS), a syndromic autism spectrum disorder associated with craniofacial abnormalities, intellectual disability, and behavioral problems. There is currently no cure for SMS. Here, we generated a genetic mouse model to determine the reversibility of SMS-like neurobehavioral phenotypes in Rai1 heterozygous mice. We show that normalizing the Rai1 level 3-4 wk after birth corrected the expression of genes related to neural developmental pathways and fully reversed a social interaction deficit caused by Rai1 haploinsufficiency. In contrast, Rai1 reactivation 7-8 wk after birth was not beneficial. We also demonstrated that the correct Rai1 dose is required in both excitatory and inhibitory neurons for proper social interactions. Finally, we found that Rai1 heterozygous mice exhibited a reduction of dendritic spines in the medial prefrontal cortex (mPFC) and that optogenetic activation of mPFC neurons in adults improved the social interaction deficit of Rai1 heterozygous mice. Together, these results suggest the existence of a postnatal temporal window during which restoring Rai1 can improve the transcriptional and social behavioral deficits in a mouse model of SMS. It is possible that circuit-level interventions would be beneficial beyond this critical window.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Transtornos do Comportamento Social / Transativadores / Modelos Animais de Doenças / Síndrome de Smith-Magenis / Haploinsuficiência / Relações Interpessoais Tipo de estudo: Prognostic_studies Limite: Adolescent / Animals / Humans / Male Idioma: En Revista: Proc Natl Acad Sci U S A Ano de publicação: 2018 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Transtornos do Comportamento Social / Transativadores / Modelos Animais de Doenças / Síndrome de Smith-Magenis / Haploinsuficiência / Relações Interpessoais Tipo de estudo: Prognostic_studies Limite: Adolescent / Animals / Humans / Male Idioma: En Revista: Proc Natl Acad Sci U S A Ano de publicação: 2018 Tipo de documento: Article