Volume 8, Issue 2, Pages (July 2014)

Slides:



Advertisements
Similar presentations
Fig. 1. SR-202 Is a Specific PPARγ Antagonist A, Structure of SR-202
Advertisements

Volume 6, Issue 7, Pages (July 2017)
Volume 9, Issue 5, Pages (December 2014)
Volume 26, Issue 14, Pages (July 2016)
Canonical Wnt/β-catenin signaling mediates transforming growth factor-β1-driven podocyte injury and proteinuria  Dan Wang, Chunsun Dai, Yingjian Li, Youhua.
Catabolic cytokines disrupt the circadian clock and the expression of clock-controlled genes in cartilage via an NFкB-dependent pathway  B. Guo, N. Yang,
Volume 23, Issue 4, Pages (April 2018)
Volume 131, Issue 2, Pages (August 2006)
Volume 137, Issue 2, Pages e2 (August 2009)
In Vivo Monitoring of Peripheral Circadian Clocks in the Mouse
Volume 16, Issue 10, Pages (September 2016)
Volume 12, Issue 7, Pages (April 2002)
Rhythmic Oxygen Levels Reset Circadian Clocks through HIF1α
Volume 13, Issue 8, Pages (November 2015)
Volume 12, Issue 3, Pages (July 2015)
Volume 134, Issue 2, Pages (July 2008)
Volume 24, Issue 11, Pages (June 2014)
Manipulating the Cellular Circadian Period of Arginine Vasopressin Neurons Alters the Behavioral Circadian Period  Michihiro Mieda, Hitoshi Okamoto, Takeshi.
Volume 21, Issue 11, Pages (December 2017)
Volume 85, Issue 5, Pages (March 2015)
Gopal K. Pattanayak, Guillaume Lambert, Kevin Bernat, Michael J. Rust 
Volume 24, Issue 6, Pages (March 2014)
IL-1R1 Signaling Facilitates Munro’s Microabscess Formation in Psoriasiform Imiquimod-Induced Skin Inflammation  Mireia Uribe-Herranz, Li-Hua Lian, Kirsten.
Volume 24, Issue 7, Pages (March 2014)
Volume 43, Issue 5, Pages (September 2004)
Volume 79, Issue 4, Pages (August 2013)
Volume 19, Issue 4, Pages (February 2009)
Matthew P. Pando, David Morse, Nicolas Cermakian, Paolo Sassone-Corsi 
Visualizing Ligand Binding to a GPCR In Vivo Using NanoBRET
Volume 18, Issue 13, Pages (March 2017)
Volume 11, Issue 4, Pages (April 2010)
Volume 23, Issue 4, Pages (April 2016)
Volume 13, Issue 6, Pages (June 2011)
Volume 5, Issue 4, Pages (November 2013)
Protection against High-Fat-Diet-Induced Obesity in MDM2C305F Mice Due to Reduced p53 Activity and Enhanced Energy Expenditure  Shijie Liu, Tae-Hyung.
Volume 14, Issue 10, Pages (March 2016)
Meal Timing Regulates the Human Circadian System
A Circadian Clock in HaCaT Keratinocytes
Heat Shock Transcription Factor 1 Is a Key Determinant of HCC Development by Regulating Hepatic Steatosis and Metabolic Syndrome  Xiongjie Jin, Demetrius.
PER1 Phosphorylation Specifies Feeding Rhythm in Mice
Circadian Disruption Leads to Insulin Resistance and Obesity
Volume 13, Issue 8, Pages (November 2015)
Volume 17, Issue 8, Pages (November 2016)
Volume 20, Issue 24, Pages (December 2010)
Volume 20, Issue 5, Pages (August 2017)
Volume 1, Issue 4, Pages (April 2005)
Chen Liu, Steven M Reppert  Neuron 
Blocking Ca2+ Channel β3 Subunit Reverses Diabetes
Essential Role of TGF-β Signaling in Glucose-Induced Cell Hypertrophy
Differential Gene Induction of Human β-Defensins (hBD-1, -2, -3, and -4) in Keratinocytes Is Inhibited by Retinoic Acid  Jürgen Harder, Ulf Meyer-Hoffert,
Kanyan Xu, Xiangzhong Zheng, Amita Sehgal  Cell Metabolism 
Connective tissue growth factor expression is regulated by histamine in lung fibroblasts: Potential role of histamine in airway remodeling  Steffen Kunzmann,
PAS Kinase Drives Lipogenesis through SREBP-1 Maturation
Hung-Chun Chang, Leonard Guarente  Cell 
Volume 12, Issue 4, Pages (July 2015)
Circadian Dysfunction Induces Leptin Resistance in Mice
Volume 27, Issue 7, Pages (April 2017)
Transient Receptor Potential Vanilloid-1 Mediates Heat-Shock-Induced Matrix Metalloproteinase-1 Expression in Human Epidermal Keratinocytes  Wen H. Li,
Smith-Magenis Syndrome Results in Disruption of CLOCK Gene Transcription and Reveals an Integral Role for RAI1 in the Maintenance of Circadian Rhythmicity 
Volume 20, Issue 4, Pages (October 2014)
A Mathematical Model of the Liver Circadian Clock Linking Feeding and Fasting Cycles to Clock Function  Aurore Woller, Hélène Duez, Bart Staels, Marc.
Arisa Hirano, Daniel Braas, Ying-Hui Fu, Louis J. Ptáček  Cell Reports 
Rongmin Chen, Matthew D’Alessandro, Choogon Lee  Current Biology 
Volume 6, Issue 5, Pages (November 2007)
Volume 129, Issue 3, Pages (May 2007)
Adipose Fatty Acid Oxidation Is Required for Thermogenesis and Potentiates Oxidative Stress-Induced Inflammation  Jieun Lee, Jessica M. Ellis, Michael J.
Volume 46, Issue 5, Pages (June 2012)
Volume 27, Issue 17, Pages e2 (September 2017)
Volume 91, Issue 7, Pages (December 1997)
Volume 24, Issue 11, Pages (June 2014)
Presentation transcript:

Volume 8, Issue 2, Pages 393-401 (July 2014) The Role of the Endocrine System in Feeding-Induced Tissue-Specific Circadian Entrainment  Miho Sato, Mariko Murakami, Koichi Node, Ritsuko Matsumura, Makoto Akashi  Cell Reports  Volume 8, Issue 2, Pages 393-401 (July 2014) DOI: 10.1016/j.celrep.2014.06.015 Copyright © 2014 The Authors Terms and Conditions

Cell Reports 2014 8, 393-401DOI: (10.1016/j.celrep.2014.06.015) Copyright © 2014 The Authors Terms and Conditions

Figure 1 In Vivo Monitoring of Peripheral Tissues from Individual Animals during Restricted Feeding (A–C) Per2-luciferase (Per2Luc) mice during the restricted feeding schedule were monitored with an in vivo imaging system. Feeding schedule (A, left). Per2Luc mice received food during the nighttime (21:00–9:00; light on at 9:00 and off at 21:00) for 1 week and then had restricted food access for 6 hr (9:00–15:00) under continuous dark conditions. Black-and-white bars represent light-dark (LD) conditions. Pink shadow represents feeding time. Images were taken at the time indicated by the black triangles. Right, representative in vivo images from the ventral aspect. Yellow arrowheads denote the submandibular gland (Sub Gla). (B) Averaged quantitative data of Sub Gla and the liver from (A). Rhythmic expression of Per2 in the liver was entrained to restricted feeding, although not in the Sub Gla. ∗p < 0.05, ∗∗p < 0.01 versus corresponding nighttime. The data represent the mean ± SE (n = 8). (C) Individual quantitative data of liver from (A). Cell Reports 2014 8, 393-401DOI: (10.1016/j.celrep.2014.06.015) Copyright © 2014 The Authors Terms and Conditions

Figure 2 Involvement of Insulin in Feeding-Induced Circadian Phase Shift in the Liver (A) Acute induction of Per2 transcripts after feeding was blocked by S961. Per2 mRNA expression levels in liver at ZT2 (60 min after feeding onset) were determined with RT-PCR. Each value was normalized with β-actin. The data represent the mean ± SE (n = 3). ∗p < 0.05. (B) Representative in vivo image of Per2Luc mice liver injected with S961 30 min before the start of feeding every day during restricted feeding. S961 inhibited the entrainment of Per2. (C) Quantitative data of bioluminescence in the liver from (B). The data represent the mean ± SE (n = 8). ∗∗p < 0.01. (D) Acrophases in a representative animal from (B) were calculated by a cosine curve fitting. The data at right represent the mean ± SE (n = 8). (E) Quantitative RT-PCR of Per2, Bmal1, and Rev-erbα mRNA in the liver during RF (ZT1-7) in LD; n = 2. Black circles represent individual values of mice given PBS mice and green circles represent mice given S961. S961 was used at a concentration of 30 nmol/kg in all experiments. Cell Reports 2014 8, 393-401DOI: (10.1016/j.celrep.2014.06.015) Copyright © 2014 The Authors Terms and Conditions

Figure 3 Insulin-Mediated Circadian Phase Shift of Per2 Expression Rhythms in Insulin-Sensitive Tissues (A–E) Explants derived from Per2Luc mice were treated with insulin at specific phases. Bioluminescence was measured in real time. (A) Representative data of liver explants pretreated with 200 nM S961 from 30 min before administration of insulin. Gray shadows represent the presence of insulin and S961 in culture media. (B) Oil Red O staining and representative data of explants derived from high-fat diet (HFD)-fed Per2Luc mice. Gray shadows represent the presence of insulin in culture media. CV, central vein; NC, normal control. (C) Phase-dependent alteration of mice liver explants with insulin. Experiments with a transient treatment (1 hr) of insulin are shown on the left, and experiments with a continuous treatment at right. Gray shadows represent the presence of insulin in culture media. Representative data are shown. (D) Phase response plot of time interval between peaks in liver from (C). (E) Representative data and phase response plot of time interval between peaks of insulin-administered submandibular gland (Sub Gla), lung, aorta, and white adipose tissue (WAT). All explants were treated with dexamethasone (Dex) and the second peak after Dex treatment was defined as peak2 (∗), and the next peak as peak3. Peak2-time was referred to as time = 0. Peak interval was calculated from the time difference between peak4 and peak2. Arrowheads indicate the time of administration. For the analysis of slices other than WAT, data were detrended by subtracting a 24 hr running average from raw data. Cell Reports 2014 8, 393-401DOI: (10.1016/j.celrep.2014.06.015) Copyright © 2014 The Authors Terms and Conditions

Figure 4 Acquisition of Insulin Sensitivity by Ectopic Receptor Expression and Phase-Dependent Circadian Phase Shift by Transient Induction of Per2 (A) NIH 3T3 fibroblasts were transfected with the Bmal1-driven luciferase and human insulin receptor α (hIRα) subunit expression vectors, and then stimulated with 60 nM insulin or vehicle. Arrowheads indicate the time of administration. To synchronize cellular clocks, the cells were treated with 50 nM dexamethasone more than 24 hr before insulin stimulation. (B) Phase differences were calculated by comparing the first peak or trough after administration of insulin. DL, delay phase administration (increasing phase of Bmal1 expression); ADV, advance phase administration (decreasing phase of Bmal1 expression). Data are represented as the mean ± SE for triplicate samples. (C and D) NIH 3T3 and U2OS cells were transfected with the Bmal1-driven luciferase vector and a Lac repressor-expressing vector in the presence or absence of an IPTG-inducible Per2 expression vector. The cells were treated with 50 nM dexamethasone (2 hr) to synchronize cellular clocks. Bmal1 transcription was monitored in real-time using a cell culture-based luminescent monitoring system in the presence of luciferin. Approximately 18–20 hr (decreasing phase of Bmal1 expression, for “phase advance” experiments) or 30–32 hr (increasing phase of Bmal1 expression, for “phase delay” experiments) after the dexamethasone shock, 2 mM IPTG was added to the culture media to induce Per2 for 1 hr. Arrows indicate the time of administration. One hour after the administration, IPTG was removed. (E and F) Phase differences were calculated by comparing the first peak or trough after administration of IPTG. Data are represented as the mean ± SE for triplicate samples. DL, delay phase administration (increasing phase of Bmal1 expression); ADV, advance phase administration (decreasing phase of Bmal1 expression). (G and H) Liver and white adipose tissue (WAT) explants from Per2Luc mice were pretreated with 50 μM LY294002 (LY) or 20 μM U0126 (U) before administration of insulin. Explants were incubated with insulin for 2 hr. The data represent the mean ± SE. Gray shadows represent the presence of insulin and inhibitors in culture media. For synchronization of cellular clocks, all explants were treated with 100 nM dexamethasone (Dex). The second peak after Dex treatment was defined as peak2 (∗) and the next peak as peak3. Peak2-time was referred to as time = 0. Peak interval was calculated from the time difference between peak3 and peak2. Arrowheads indicate the time of administration. For the analysis of slices other than WAT, data were detrended by subtracting a 24 hr running average from raw data. Cell Reports 2014 8, 393-401DOI: (10.1016/j.celrep.2014.06.015) Copyright © 2014 The Authors Terms and Conditions