Neurobiology of Disease (Jun 2022)

Deficiency of autism-related Scn2a gene in mice disrupts sleep patterns and circadian rhythms

  • Zhixiong Ma,
  • Muriel Eaton,
  • Yushuang Liu,
  • Jingliang Zhang,
  • Xiaoling Chen,
  • Xinyu Tu,
  • Yiqiang Shi,
  • Zhefu Que,
  • Kyle Wettschurack,
  • Zaiyang Zhang,
  • Riyi Shi,
  • Yueyi Chen,
  • Adam Kimbrough,
  • Nadia A. Lanman,
  • Leah Schust,
  • Zhuo Huang,
  • Yang Yang

Journal volume & issue
Vol. 168
p. 105690

Abstract

Read online

Autism spectrum disorder (ASD) affects ~2% of the population in the US, and monogenic forms of ASD often result in the most severe manifestation of the disorder. Recently, SCN2A has emerged as a leading gene associated with ASD, of which abnormal sleep pattern is a common comorbidity. SCN2A encodes the voltage-gated sodium channel NaV1.2. Predominantly expressed in the brain, NaV1.2 mediates the action potential firing of neurons. Clinical studies found that a large portion of children with SCN2A deficiency have sleep disorders, which severely impact the quality of life of affected individuals and their caregivers. The underlying mechanism of sleep disturbances related to NaV1.2 deficiency, however, is not known. Using a gene-trap Scn2a-deficient mouse model (Scn2atrap), we found that Scn2a deficiency results in increased wakefulness and reduced non-rapid-eye-movement (NREM) sleep. Brain region-specific Scn2a deficiency in the suprachiasmatic nucleus (SCN) containing region, which is involved in circadian rhythms, partially recapitulates the sleep disturbance phenotypes. At the cellular level, we found that Scn2a deficiency disrupted the firing pattern of spontaneously firing neurons in the SCN region. At the molecular level, RNA-sequencing analysis revealed differentially expressed genes in the circadian entrainment pathway including core clock genes Per1 and Per2. Performing a transcriptome-based compound discovery, we identified dexanabinol (HU-211), a putative glutamate receptor modulator, that can partially reverse the sleep disturbance in mice. Overall, our study reveals possible molecular and cellular mechanisms underlying Scn2a deficiency-related sleep disturbances, which may inform the development of potential pharmacogenetic interventions for the affected individuals.

Keywords