Abstract

The suprachiasmatic nucleus (SCN) is the principal circadian clock of mammals, coordinating daily rhythms of physiology and behavior. Circadian timing pivots around self-sustaining transcriptional-translational negative feedback loops (TTFLs), whereby CLOCK and BMAL1 drive the expression of the negative regulators Period and Cryptochrome (Cry). Global deletion of Cry1 and Cry2 disables the TTFL, resulting in arrhythmicity in downstream behaviors. We used this highly tractable biology to further develop genetic code expansion (GCE) as a translational switch to achieve reversible control of a biologically relevant protein, Cry1, in the SCN. This employed an orthogonal aminoacyl-tRNA synthetase/tRNACUA pair delivered to the SCN by adeno-associated virus (AAV) vectors, allowing incorporation of a noncanonical amino acid (ncAA) into AAV-encoded Cry1 protein carrying an ectopic amber stop codon. Thus, translational readthrough and Cry1 expression were conditional on the supply of ncAA via culture medium or drinking water and were restricted to neurons by synapsin-dependent expression of aminoacyl tRNA-synthetase. Activation of Cry1 translation by ncAA in neurons of arrhythmic Cry-null SCN slices immediately and dose-dependently initiated TTFL circadian rhythms, which dissipated rapidly after ncAA withdrawal. Moreover, genetic activation of the TTFL in SCN neurons rapidly and reversibly initiated circadian behavior in otherwise arrhythmic Cry-null mice, with rhythm amplitude being determined by the number of transduced SCN neurons. Thus, Cry1 does not specify the development of circadian circuitry and competence but is essential for its labile and rapidly reversible activation. This demonstrates reversible control of mammalian behavior using GCE-based translational switching, a method of potentially broad neurobiological interest.

Highlights

  • ADivision of Neurobiology, Medical Research Council Laboratory of Molecular Biology, CB2 0QH Cambridge, United Kingdom; and bDivision of Protein and Nucleic Acid Chemistry, Medical Research Council Laboratory of Molecular Biology, CB2 0QH Cambridge, United Kingdom

  • To test the potential of translational switching for conditional expression of Cry1, suprachiasmatic nucleus (SCN) organotypic slices were cotransduced with two associated virus (AAV)-expressed constructs (Fig. 1A)

  • Cry1 is expressed from the minimal Cry1 promoter [18, 19], and for translational conditionality it carries a TAG mutation to confer amber dependence

Read more

Summary

Introduction

ADivision of Neurobiology, Medical Research Council Laboratory of Molecular Biology, CB2 0QH Cambridge, United Kingdom; and bDivision of Protein and Nucleic Acid Chemistry, Medical Research Council Laboratory of Molecular Biology, CB2 0QH Cambridge, United Kingdom. Global deletion of Cry and Cry disables the TTFL, resulting in arrhythmicity in downstream behaviors We used this highly tractable biology to further develop genetic code expansion (GCE) as a translational switch to achieve reversible control of a biologically relevant protein, Cry, in the SCN. Cry does not specify the development of circadian circuitry and competence but is essential for its labile and rapidly reversible activation This demonstrates reversible control of mammalian behavior using GCE-based translational switching, a method of potentially broad neurobiological interest. Cry translation in SCN neurons was sufficient to initiate circadian behavior rapidly and reversibly in arrhythmic Cry-null mice This demonstrates control of mammalian behavior using translational switching, a method of broad applicability

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call