2018
DOI: 10.1016/j.cmet.2018.05.023
|View full text |Cite
|
Sign up to set email alerts
|

Autophagy Regulates the Liver Clock and Glucose Metabolism by Degrading CRY1

Abstract: The circadian clock coordinates behavioral and circadian cues with availability and utilization of nutrients. Proteasomal degradation of clock repressors, such as cryptochrome (CRY)1, maintains periodicity. Whether macroautophagy, a quality control pathway, degrades circadian proteins remains unknown. Here we show that circadian proteins BMAL1, CLOCK, REV-ERBα, and CRY1 are lysosomal targets, and that macroautophagy affects the circadian clock by selectively degrading CRY1. Autophagic degradation of CRY1, an i… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
2
2
1

Citation Types

1
81
2

Year Published

2019
2019
2024
2024

Publication Types

Select...
7
1

Relationship

0
8

Authors

Journals

citations
Cited by 132 publications
(84 citation statements)
references
References 47 publications
1
81
2
Order By: Relevance
“…31,40,41 The autophagy pathway was shown to play a modulatory role in degradation of CRY1 and BMAL1 (mammalian homolog of Cycle) during circadian regulation of glucose metabolism in mice liver. 42,43 The autophagy pathway and TOR pathway (a negative regulator of autophagy) were shown to regulate circadian locomotor rhythms. 44 Ectopic expression of mutant Shibire (Drosophila homolog of Dynamin)-an ATPase required for scission of plasma membrane to form endocytic vesicles, causes disruption of circadian rhythm.…”
Section: Gmr-driven Chmp2b Intron5 Expression Causes Reduction In Tmentioning
confidence: 99%
“…31,40,41 The autophagy pathway was shown to play a modulatory role in degradation of CRY1 and BMAL1 (mammalian homolog of Cycle) during circadian regulation of glucose metabolism in mice liver. 42,43 The autophagy pathway and TOR pathway (a negative regulator of autophagy) were shown to regulate circadian locomotor rhythms. 44 Ectopic expression of mutant Shibire (Drosophila homolog of Dynamin)-an ATPase required for scission of plasma membrane to form endocytic vesicles, causes disruption of circadian rhythm.…”
Section: Gmr-driven Chmp2b Intron5 Expression Causes Reduction In Tmentioning
confidence: 99%
“…(LC3)-interacting region (LIR) motifs are found in the CRY1 sequence, and their role has been confirmed by the observation that mice in which autophagy is genetically blocked exhibit accumulation of CRY1 and disruption of the circadian clock in the liver (Toledo et al, 2018). Moreover, autophagic degradation of CRY1 is important in maintaining blood glucose levels by driving gluconeogenesis (Toledo et al, 2018). As in mammals (Turek et al, 2005;Green et al, 2008), the circadian clock is involved in fat storage and mobilization also in Drosophila.…”
Section: Cry and The Regulation Of Metabolic Processesmentioning
confidence: 96%
“…Moreover, CRY1 interacts with the autophagosome marker light chain 3 (LC3), responsible for its time-dependent autophagic degradation (Toledo et al, 2018). (LC3)-interacting region (LIR) motifs are found in the CRY1 sequence, and their role has been confirmed by the observation that mice in which autophagy is genetically blocked exhibit accumulation of CRY1 and disruption of the circadian clock in the liver (Toledo et al, 2018). Moreover, autophagic degradation of CRY1 is important in maintaining blood glucose levels by driving gluconeogenesis (Toledo et al, 2018).…”
Section: Cry and The Regulation Of Metabolic Processesmentioning
confidence: 99%
“…124 CRY1 and CRY2 proteins reduce fasting glucose production by inhibiting glucagon-induced DIBNER liver gluconeogenesis. 110,125,126 Moreover, CRY1 levels are decreased by autophagy in rodent liver, providing a link between autophagy, increased gluconeogenesis and the overall maintenance of glucose homeostasis. 126 Furthermore, diet-induced obesity in wild-type mice causes remodelling of the circadian liver enhancer landscape, leading to massive changes in lipid metabolism via SREBP and PPARα.…”
Section: Dibnermentioning
confidence: 99%
“…110,125,126 Moreover, CRY1 levels are decreased by autophagy in rodent liver, providing a link between autophagy, increased gluconeogenesis and the overall maintenance of glucose homeostasis. 126 Furthermore, diet-induced obesity in wild-type mice causes remodelling of the circadian liver enhancer landscape, leading to massive changes in lipid metabolism via SREBP and PPARα. 127 Additionally, the RNA-binding protein NONO situated in liver cell nuclei couples the rhythmic expression of metabolic genes with nutrient levels, with NONO-deficient mice exhibiting impaired glucose tolerance as well as lower hepatic glycogen and lipid content.…”
Section: Dibnermentioning
confidence: 99%