201
|
Maboudi Afkham H, Qiu X, The M, Käll L. Uncertainty estimation of predictions of peptides' chromatographic retention times in shotgun proteomics. Bioinformatics 2017; 33:508-513. [PMID: 27797755 DOI: 10.1093/bioinformatics/btw619] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 09/20/2016] [Indexed: 12/17/2022] Open
Abstract
Motivation Liquid chromatography is frequently used as a means to reduce the complexity of peptide-mixtures in shotgun proteomics. For such systems, the time when a peptide is released from a chromatography column and registered in the mass spectrometer is referred to as the peptide's retention time . Using heuristics or machine learning techniques, previous studies have demonstrated that it is possible to predict the retention time of a peptide from its amino acid sequence. In this paper, we are applying Gaussian Process Regression to the feature representation of a previously described predictor E lude . Using this framework, we demonstrate that it is possible to estimate the uncertainty of the prediction made by the model. Here we show how this uncertainty relates to the actual error of the prediction. Results In our experiments, we observe a strong correlation between the estimated uncertainty provided by Gaussian Process Regression and the actual prediction error. This relation provides us with new means for assessment of the predictions. We demonstrate how a subset of the peptides can be selected with lower prediction error compared to the whole set. We also demonstrate how such predicted standard deviations can be used for designing adaptive windowing strategies. Contact lukas.kall@scilifelab.se. Availability and Implementation Our software and the data used in our experiments is publicly available and can be downloaded from https://github.com/statisticalbiotechnology/GPTime .
Collapse
Affiliation(s)
- Heydar Maboudi Afkham
- Science for Life Laboratory, School of Biotechnology, KTH - Royal Institute of Technology, 17121 Solna, Sweden
| | - Xuanbin Qiu
- Science for Life Laboratory, School of Biotechnology, KTH - Royal Institute of Technology, 17121 Solna, Sweden
| | - Matthew The
- Science for Life Laboratory, School of Biotechnology, KTH - Royal Institute of Technology, 17121 Solna, Sweden
| | - Lukas Käll
- Science for Life Laboratory, School of Biotechnology, KTH - Royal Institute of Technology, 17121 Solna, Sweden
| |
Collapse
|
202
|
Abstract
In most organisms, gene expression over the course of the day is under the control of the circadian clock. The canonical clock operates as a gene expression circuit that is controlled at the level of transcription, and transcriptional control is also a major clock output. However, rhythmic transcription cannot explain all the observed rhythms in protein accumulation. Although it is clear that rhythmic gene expression also involves RNA processing and protein turnover, until two years ago little was known in any eukaryote about diel dynamics of mRNA translation into protein. A recent series of studies in animals and plants demonstrated that diel cycles of translation efficiency are widespread across the tree of life and its transcriptomes. There are surprising parallels between the patterns of diel translation in mammals and plants. For example, ribosomal proteins and mitochondrial proteins are under translational control in mouse liver, human tissue culture, and Arabidopsis seedlings. In contrast, the way in which the circadian clock, light-dark changes, and other environmental factors such as nutritional signals interact to drive the cycles of translation may differ between organisms. Further investigation is needed to identify the signaling pathways, biochemical mechanisms, RNA sequence features, and the physiological implications of diel translation.
Collapse
Affiliation(s)
- Sarah Catherine Mills
- a Department of Biochemistry and Cellular & Molecular Biology , The University of Tennessee , Knoxville , TN , USA
| | - Ramya Enganti
- a Department of Biochemistry and Cellular & Molecular Biology , The University of Tennessee , Knoxville , TN , USA
| | - Albrecht G von Arnim
- a Department of Biochemistry and Cellular & Molecular Biology , The University of Tennessee , Knoxville , TN , USA.,b UT-ORNL Graduate School of Genome Science and Technology , The University of Tennessee , Knoxville , TN , USA
| |
Collapse
|
203
|
Affiliation(s)
- Filipa Rijo-Ferreira
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Howard Hughes Medical Institute, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Joseph S. Takahashi
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Howard Hughes Medical Institute, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- * E-mail: (JST); (LMF)
| | - Luisa M. Figueiredo
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- * E-mail: (JST); (LMF)
| |
Collapse
|
204
|
Fijalkowska D, Verbruggen S, Ndah E, Jonckheere V, Menschaert G, Van Damme P. eIF1 modulates the recognition of suboptimal translation initiation sites and steers gene expression via uORFs. Nucleic Acids Res 2017; 45:7997-8013. [PMID: 28541577 PMCID: PMC5570006 DOI: 10.1093/nar/gkx469] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 05/11/2017] [Indexed: 12/25/2022] Open
Abstract
Alternative translation initiation mechanisms such as leaky scanning and reinitiation potentiate the polycistronic nature of human transcripts. By allowing for reprogrammed translation, these mechanisms can mediate biological responses to stimuli. We combined proteomics with ribosome profiling and mRNA sequencing to identify the biological targets of translation control triggered by the eukaryotic translation initiation factor 1 (eIF1), a protein implicated in the stringency of start codon selection. We quantified expression changes of over 4000 proteins and 10 000 actively translated transcripts, leading to the identification of 245 transcripts undergoing translational control mediated by upstream open reading frames (uORFs) upon eIF1 deprivation. Here, the stringency of start codon selection and preference for an optimal nucleotide context were largely diminished leading to translational upregulation of uORFs with suboptimal start. Interestingly, genes affected by eIF1 deprivation were implicated in energy production and sensing of metabolic stress.
Collapse
Affiliation(s)
- Daria Fijalkowska
- VIB-UGent Center for Medical Biotechnology, B-9000 Ghent, Belgium.,Department of Biochemistry, Ghent University, B-9000 Ghent, Belgium
| | - Steven Verbruggen
- Lab of Bioinformatics and Computational Genomics, Department of Mathematical Modelling, Statistics and Bioinformatics, Faculty of Bioscience Engineering, Ghent University, B-9000 Ghent, Belgium
| | - Elvis Ndah
- VIB-UGent Center for Medical Biotechnology, B-9000 Ghent, Belgium.,Department of Biochemistry, Ghent University, B-9000 Ghent, Belgium.,Lab of Bioinformatics and Computational Genomics, Department of Mathematical Modelling, Statistics and Bioinformatics, Faculty of Bioscience Engineering, Ghent University, B-9000 Ghent, Belgium
| | - Veronique Jonckheere
- VIB-UGent Center for Medical Biotechnology, B-9000 Ghent, Belgium.,Department of Biochemistry, Ghent University, B-9000 Ghent, Belgium
| | - Gerben Menschaert
- Lab of Bioinformatics and Computational Genomics, Department of Mathematical Modelling, Statistics and Bioinformatics, Faculty of Bioscience Engineering, Ghent University, B-9000 Ghent, Belgium
| | - Petra Van Damme
- VIB-UGent Center for Medical Biotechnology, B-9000 Ghent, Belgium.,Department of Biochemistry, Ghent University, B-9000 Ghent, Belgium
| |
Collapse
|
205
|
Manoogian ENC, Panda S. Circadian rhythms, time-restricted feeding, and healthy aging. Ageing Res Rev 2017; 39:59-67. [PMID: 28017879 PMCID: PMC5814245 DOI: 10.1016/j.arr.2016.12.006] [Citation(s) in RCA: 209] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Revised: 12/16/2016] [Accepted: 12/18/2016] [Indexed: 12/29/2022]
Abstract
Circadian rhythms optimize physiology and health by temporally coordinating cellular function, tissue function, and behavior. These endogenous rhythms dampen with age and thus compromise temporal coordination. Feeding-fasting patterns are an external cue that profoundly influence the robustness of daily biological rhythms. Erratic eating patterns can disrupt the temporal coordination of metabolism and physiology leading to chronic diseases that are also characteristic of aging. However, sustaining a robust feeding-fasting cycle, even without altering nutrition quality or quantity, can prevent or reverse these chronic diseases in experimental models. In humans, epidemiological studies have shown erratic eating patterns increase the risk of disease, whereas sustained feeding-fasting cycles, or prolonged overnight fasting, is correlated with protection from breast cancer. Therefore, optimizing the timing of external cues with defined eating patterns can sustain a robust circadian clock, which may prevent disease and improve prognosis.
Collapse
Affiliation(s)
- Emily N C Manoogian
- Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd, La Jolla, CA 92037, USA.
| | - Satchidananda Panda
- Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd, La Jolla, CA 92037, USA; University of California Center for Circadian Biology, 9500, Gilman Drive, La Jolla, 92093, USA.
| |
Collapse
|
206
|
Zhang K, Xu P, Sowers JL, Machuca DF, Mirfattah B, Herring J, Tang H, Chen Y, Tian B, Brasier AR, Sowers LC. Proteome Analysis of Hypoxic Glioblastoma Cells Reveals Sequential Metabolic Adaptation of One-Carbon Metabolic Pathways. Mol Cell Proteomics 2017; 16:1906-1921. [PMID: 28874504 DOI: 10.1074/mcp.ra117.000154] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Indexed: 02/04/2023] Open
Abstract
Rapidly proliferating tumors are exposed to a hypoxic microenvironment because of their density, high metabolic consumption, and interruptions in blood flow because of immature angiogenesis. Cellular responses to hypoxia promote highly malignant and metastatic behavior, as well as a chemotherapy-resistant state. To better understand the complex relationships between hypoxic adaptations and cancer progression, we studied the dynamic proteome responses of glioblastoma cells exposed to hypoxia via an innovative approach: quantification of newly synthesized proteins using heavy stable-isotope arginine labeling combined with accurate assessment of cell replication by quantification of the light/heavy arginine ratio of peptides in histone H4. We found that hypoxia affects cancer cells in multiple intertwined ways: inflammation, typically with over-expressed glucose transporter (GLUT1), DUSP4/MKP2, and RelA proteins; a metabolic adaptation with overexpression of all glycolytic pathway enzymes for pyruvate/lactate synthesis; and the EMT (epithelial-mesenchymal transition) and cancer stem cell (CSC) renewal with characteristic morphological changes and mesenchymal/CSC protein expression profiles. For the first time, we identified the vitamin B12 transporter protein TCN2, which is essential for one-carbon metabolism, as being significantly downregulated. Further, we found, by knockdown and overexpression experiments, that TCN2 plays an important role in controlling cancer cell transformation toward the highly aggressive mesenchymal/CSC stage; low expression of TCN2 has an effect similar to hypoxia, whereas high expression of TCN2 can reverse it. We conclude that hypoxia induces sequential metabolic responses of one-carbon metabolism in tumor cells. Our mass spectrometry data are available via ProteomeXchange with identifiers PXD005487 (TMT-labeling) and PXD007280 (label-free).
Collapse
Affiliation(s)
- Kangling Zhang
- From the ‡Department of Pharmacology, University of Texas Medical Branch, Galveston (UTMB), Texas, 77555; .,¶Sealy Center for Molecular Medicine, UTMB, Galveston, Texas, 77555
| | - Pei Xu
- ‖Department of Neuroscience and Cell Biology, UTMB, Galveston, Texas, 77555
| | - James L Sowers
- ‖Department of Neuroscience and Cell Biology, UTMB, Galveston, Texas, 77555
| | - Daniel F Machuca
- From the ‡Department of Pharmacology, University of Texas Medical Branch, Galveston (UTMB), Texas, 77555
| | - Barsam Mirfattah
- From the ‡Department of Pharmacology, University of Texas Medical Branch, Galveston (UTMB), Texas, 77555
| | - Jason Herring
- From the ‡Department of Pharmacology, University of Texas Medical Branch, Galveston (UTMB), Texas, 77555
| | - Hui Tang
- From the ‡Department of Pharmacology, University of Texas Medical Branch, Galveston (UTMB), Texas, 77555
| | - Yan Chen
- ‖Department of Neuroscience and Cell Biology, UTMB, Galveston, Texas, 77555
| | - Bing Tian
- §Institute for Translational Sciences, UTMB, Galveston, Texas, 77555.,¶Sealy Center for Molecular Medicine, UTMB, Galveston, Texas, 77555
| | - Allan R Brasier
- §Institute for Translational Sciences, UTMB, Galveston, Texas, 77555.,¶Sealy Center for Molecular Medicine, UTMB, Galveston, Texas, 77555
| | - Lawrence C Sowers
- From the ‡Department of Pharmacology, University of Texas Medical Branch, Galveston (UTMB), Texas, 77555.,§Institute for Translational Sciences, UTMB, Galveston, Texas, 77555
| |
Collapse
|
207
|
Mauvoisin D, Atger F, Dayon L, Núñez Galindo A, Wang J, Martin E, Da Silva L, Montoliu I, Collino S, Martin FP, Ratajczak J, Cantó C, Kussmann M, Naef F, Gachon F. Circadian and Feeding Rhythms Orchestrate the Diurnal Liver Acetylome. Cell Rep 2017; 20:1729-1743. [PMID: 28813682 PMCID: PMC5568034 DOI: 10.1016/j.celrep.2017.07.065] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 06/12/2017] [Accepted: 07/24/2017] [Indexed: 12/24/2022] Open
Abstract
Lysine acetylation is involved in various biological processes and is considered a key reversible post-translational modification in the regulation of gene expression, enzyme activity, and subcellular localization. This post-translational modification is therefore highly relevant in the context of circadian biology, but its characterization on the proteome-wide scale and its circadian clock dependence are still poorly described. Here, we provide a comprehensive and rhythmic acetylome map of the mouse liver. Rhythmic acetylated proteins showed subcellular localization-specific phases that correlated with the related metabolites in the regulated pathways. Mitochondrial proteins were over-represented among the rhythmically acetylated proteins and were highly correlated with SIRT3-dependent deacetylation. SIRT3 activity being nicotinamide adenine dinucleotide (NAD)+ level-dependent, we show that NAD+ is orchestrated by both feeding rhythms and the circadian clock through the NAD+ salvage pathway but also via the nicotinamide riboside pathway. Hence, the diurnal acetylome relies on a functional circadian clock and affects important diurnal metabolic pathways in the mouse liver.
Collapse
Affiliation(s)
- Daniel Mauvoisin
- Diabetes and Circadian Rhythms Department, Nestlé Institute of Health Sciences, 1015 Lausanne, Switzerland; Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Florian Atger
- Diabetes and Circadian Rhythms Department, Nestlé Institute of Health Sciences, 1015 Lausanne, Switzerland; Department of Pharmacology and Toxicology, University of Lausanne, 1015 Lausanne, Switzerland
| | - Loïc Dayon
- Systems Nutrition, Metabonomics & Proteomics, Nestlé Institute of Health Sciences, 1015 Lausanne, Switzerland
| | - Antonio Núñez Galindo
- Systems Nutrition, Metabonomics & Proteomics, Nestlé Institute of Health Sciences, 1015 Lausanne, Switzerland
| | - Jingkui Wang
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Eva Martin
- Diabetes and Circadian Rhythms Department, Nestlé Institute of Health Sciences, 1015 Lausanne, Switzerland
| | - Laetitia Da Silva
- Systems Nutrition, Metabonomics & Proteomics, Nestlé Institute of Health Sciences, 1015 Lausanne, Switzerland
| | - Ivan Montoliu
- Systems Nutrition, Metabonomics & Proteomics, Nestlé Institute of Health Sciences, 1015 Lausanne, Switzerland
| | - Sebastiano Collino
- Systems Nutrition, Metabonomics & Proteomics, Nestlé Institute of Health Sciences, 1015 Lausanne, Switzerland
| | - Francois-Pierre Martin
- Systems Nutrition, Metabonomics & Proteomics, Nestlé Institute of Health Sciences, 1015 Lausanne, Switzerland
| | - Joanna Ratajczak
- Diabetes and Circadian Rhythms Department, Nestlé Institute of Health Sciences, 1015 Lausanne, Switzerland; School of Life Sciences, Ecole Polytechnique Fédérale Lausanne, 1015 Lausanne, Switzerland
| | - Carles Cantó
- Diabetes and Circadian Rhythms Department, Nestlé Institute of Health Sciences, 1015 Lausanne, Switzerland; School of Life Sciences, Ecole Polytechnique Fédérale Lausanne, 1015 Lausanne, Switzerland
| | - Martin Kussmann
- Systems Nutrition, Metabonomics & Proteomics, Nestlé Institute of Health Sciences, 1015 Lausanne, Switzerland
| | - Felix Naef
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Frédéric Gachon
- Diabetes and Circadian Rhythms Department, Nestlé Institute of Health Sciences, 1015 Lausanne, Switzerland; School of Life Sciences, Ecole Polytechnique Fédérale Lausanne, 1015 Lausanne, Switzerland.
| |
Collapse
|
208
|
Kriebs A, Jordan SD, Soto E, Henriksson E, Sandate CR, Vaughan ME, Chan AB, Duglan D, Papp SJ, Huber AL, Afetian ME, Yu RT, Zhao X, Downes M, Evans RM, Lamia KA. Circadian repressors CRY1 and CRY2 broadly interact with nuclear receptors and modulate transcriptional activity. Proc Natl Acad Sci U S A 2017; 114:8776-8781. [PMID: 28751364 PMCID: PMC5565439 DOI: 10.1073/pnas.1704955114] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Nuclear hormone receptors (NRs) regulate physiology by sensing lipophilic ligands and adapting cellular transcription appropriately. A growing understanding of the impact of circadian clocks on mammalian transcription has sparked interest in the interregulation of transcriptional programs. Mammalian clocks are based on a transcriptional feedback loop featuring the transcriptional activators circadian locomotor output cycles kaput (CLOCK) and brain and muscle ARNT-like 1 (BMAL1), and transcriptional repressors cryptochrome (CRY) and period (PER). CRY1 and CRY2 bind independently of other core clock factors to many genomic sites, which are enriched for NR recognition motifs. Here we report that CRY1/2 serve as corepressors for many NRs, indicating a new facet of circadian control of NR-mediated regulation of metabolism and physiology, and specifically contribute to diurnal modulation of drug metabolism.
Collapse
Affiliation(s)
- Anna Kriebs
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Sabine D Jordan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Erin Soto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Emma Henriksson
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
- Department of Clinical Sciences, Clinical Research Centre, Lund University, Malmö 20502, Sweden
| | - Colby R Sandate
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Megan E Vaughan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Alanna B Chan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Drew Duglan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Stephanie J Papp
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Anne-Laure Huber
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Megan E Afetian
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Ruth T Yu
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Xuan Zhao
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Michael Downes
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Ronald M Evans
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037
- Howard Hughes Medical Institute, The Salk Institute for Biological Studies, La Jolla, CA 92037
- Center for Circadian Biology, University of California, San Diego, CA 92161
| | - Katja A Lamia
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037;
- Center for Circadian Biology, University of California, San Diego, CA 92161
| |
Collapse
|
209
|
Rhythmic Behavior Is Controlled by the SRm160 Splicing Factor in Drosophila melanogaster. Genetics 2017; 207:593-607. [PMID: 28801530 DOI: 10.1534/genetics.117.300139] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 08/02/2017] [Indexed: 02/06/2023] Open
Abstract
Circadian clocks organize the metabolism, physiology, and behavior of organisms throughout the day-night cycle by controlling daily rhythms in gene expression at the transcriptional and post-transcriptional levels. While many transcription factors underlying circadian oscillations are known, the splicing factors that modulate these rhythms remain largely unexplored. A genome-wide assessment of the alterations of gene expression in a null mutant of the alternative splicing regulator SR-related matrix protein of 160 kDa (SRm160) revealed the extent to which alternative splicing impacts on behavior-related genes. We show that SRm160 affects gene expression in pacemaker neurons of the Drosophila brain to ensure proper oscillations of the molecular clock. A reduced level of SRm160 in adult pacemaker neurons impairs circadian rhythms in locomotor behavior, and this phenotype is caused, at least in part, by a marked reduction in period (per) levels. Moreover, rhythmic accumulation of the neuropeptide PIGMENT DISPERSING FACTOR in the dorsal projections of these neurons is abolished after SRm160 depletion. The lack of rhythmicity in SRm160-downregulated flies is reversed by a fully spliced per construct, but not by an extra copy of the endogenous locus, showing that SRm160 positively regulates per levels in a splicing-dependent manner. Our findings highlight the significant effect of alternative splicing on the nervous system and particularly on brain function in an in vivo model.
Collapse
|
210
|
Liu Y, Gonzàlez-Porta M, Santos S, Brazma A, Marioni JC, Aebersold R, Venkitaraman AR, Wickramasinghe VO. Impact of Alternative Splicing on the Human Proteome. Cell Rep 2017; 20:1229-1241. [PMID: 28768205 PMCID: PMC5554779 DOI: 10.1016/j.celrep.2017.07.025] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 06/02/2017] [Accepted: 07/12/2017] [Indexed: 02/02/2023] Open
Abstract
Alternative splicing is a critical determinant of genome complexity and, by implication, is assumed to engender proteomic diversity. This notion has not been experimentally tested in a targeted, quantitative manner. Here, we have developed an integrative approach to ask whether perturbations in mRNA splicing patterns alter the composition of the proteome. We integrate RNA sequencing (RNA-seq) (to comprehensively report intron retention, differential transcript usage, and gene expression) with a data-independent acquisition (DIA) method, SWATH-MS (sequential window acquisition of all theoretical spectra-mass spectrometry), to capture an unbiased, quantitative snapshot of the impact of constitutive and alternative splicing events on the proteome. Whereas intron retention is accompanied by decreased protein abundance, alterations in differential transcript usage and gene expression alter protein abundance proportionate to transcript levels. Our findings illustrate how RNA splicing links isoform expression in the human transcriptome with proteomic diversity and provides a foundation for studying perturbations associated with human diseases.
Collapse
Affiliation(s)
- Yansheng Liu
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Mar Gonzàlez-Porta
- European Molecular Biology Laboratory-European Bioinformatics Institute (EMBL-EBI), Hinxton, UK
| | - Sergio Santos
- European Molecular Biology Laboratory-European Bioinformatics Institute (EMBL-EBI), Hinxton, UK
| | - Alvis Brazma
- European Molecular Biology Laboratory-European Bioinformatics Institute (EMBL-EBI), Hinxton, UK
| | - John C Marioni
- European Molecular Biology Laboratory-European Bioinformatics Institute (EMBL-EBI), Hinxton, UK
| | - Ruedi Aebersold
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland.
| | - Ashok R Venkitaraman
- The Medical Research Council Cancer Unit, University of Cambridge, Cambridge CB2 0XZ, UK.
| | - Vihandha O Wickramasinghe
- The Medical Research Council Cancer Unit, University of Cambridge, Cambridge CB2 0XZ, UK; RNA Biology and Cancer Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia.
| |
Collapse
|
211
|
Stare T, Stare K, Weckwerth W, Wienkoop S, Gruden K. Comparison between Proteome and Transcriptome Response in Potato (Solanum tuberosum L.) Leaves Following Potato Virus Y (PVY) Infection. Proteomes 2017; 5:proteomes5030014. [PMID: 28684682 PMCID: PMC5620531 DOI: 10.3390/proteomes5030014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 06/27/2017] [Accepted: 07/01/2017] [Indexed: 12/17/2022] Open
Abstract
Plant diseases caused by viral infection are affecting all major crops. Being an obligate intracellular organisms, chemical control of these pathogens is so far not applied in the field except to control the insect vectors of the viruses. Understanding of molecular responses of plant immunity is therefore economically important, guiding the enforcement of crop resistance. To disentangle complex regulatory mechanisms of the plant immune responses, understanding system as a whole is a must. However, integrating data from different molecular analysis (transcriptomics, proteomics, metabolomics, smallRNA regulation etc.) is not straightforward. We evaluated the response of potato (Solanum tuberosum L.) following the infection with potato virus Y (PVY). The response has been analyzed on two molecular levels, with microarray transcriptome analysis and mass spectroscopy-based proteomics. Within this report, we performed detailed analysis of the results on both levels and compared two different approaches for analysis of proteomic data (spectral count versus MaxQuant). To link the data on different molecular levels, each protein was mapped to the corresponding potato transcript according to StNIB paralogue grouping. Only 33% of the proteins mapped to microarray probes in a one-to-one relation and additionally many showed discordance in detected levels of proteins with corresponding transcripts. We discussed functional importance of true biological differences between both levels and showed that the reason for the discordance between transcript and protein abundance lies partly in complexity and structure of biological regulation of proteome and transcriptome and partly in technical issues contributing to it.
Collapse
Affiliation(s)
- Tjaša Stare
- Department of Biotechnology and Systems Biology, National Institute of Biology, 1000 Ljubljana, Slovenia.
| | - Katja Stare
- Department of Biotechnology and Systems Biology, National Institute of Biology, 1000 Ljubljana, Slovenia.
| | - Wolfram Weckwerth
- Department of Ecogenomics and Systems Biology, Faculty of Life Sciences, University of Vienna, 1010 Wien, Austria.
| | - Stefanie Wienkoop
- Department of Ecogenomics and Systems Biology, Faculty of Life Sciences, University of Vienna, 1010 Wien, Austria.
| | - Kristina Gruden
- Department of Biotechnology and Systems Biology, National Institute of Biology, 1000 Ljubljana, Slovenia.
| |
Collapse
|
212
|
Castelo-Szekely V, Arpat AB, Janich P, Gatfield D. Translational contributions to tissue specificity in rhythmic and constitutive gene expression. Genome Biol 2017. [PMID: 28622766 PMCID: PMC5473967 DOI: 10.1186/s13059-017-1222-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background The daily gene expression oscillations that underlie mammalian circadian rhythms show striking differences between tissues and involve post-transcriptional regulation. Both aspects remain poorly understood. We have used ribosome profiling to explore the contribution of translation efficiency to temporal gene expression in kidney and contrasted our findings with liver data available from the same mice. Results Rhythmic translation of constantly abundant messenger RNAs (mRNAs) affects largely non-overlapping transcript sets with distinct phase clustering in the two organs. Moreover, tissue differences in translation efficiency modulate the timing and amount of protein biosynthesis from rhythmic mRNAs, consistent with organ specificity in clock output gene repertoires and rhythmicity parameters. Our comprehensive datasets provided insights into translational control beyond temporal regulation. Between tissues, many transcripts show differences in translation efficiency, which are, however, of markedly smaller scale than mRNA abundance differences. Tissue-specific changes in translation efficiency are associated with specific transcript features and, intriguingly, globally counteracted and compensated transcript abundance variations, leading to higher similarity at the level of protein biosynthesis between both tissues. Conclusions We show that tissue specificity in rhythmic gene expression extends to the translatome and contributes to define the identities, the phases and the expression levels of rhythmic protein biosynthesis. Moreover, translational compensation of transcript abundance divergence leads to overall higher similarity at the level of protein production across organs. The unique resources provided through our study will serve to address fundamental questions of post-transcriptional control and differential gene expression in vivo. Electronic supplementary material The online version of this article (doi:10.1186/s13059-017-1222-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Violeta Castelo-Szekely
- Center for Integrative Genomics, University of Lausanne, Génopode, 1015, Lausanne, Switzerland
| | - Alaaddin Bulak Arpat
- Center for Integrative Genomics, University of Lausanne, Génopode, 1015, Lausanne, Switzerland.,Vital-IT, Swiss Institute of Bioinformatics, Génopode, 1015, Lausanne, Switzerland
| | - Peggy Janich
- Center for Integrative Genomics, University of Lausanne, Génopode, 1015, Lausanne, Switzerland
| | - David Gatfield
- Center for Integrative Genomics, University of Lausanne, Génopode, 1015, Lausanne, Switzerland.
| |
Collapse
|
213
|
Henriksson E, Huber AL, Soto EK, Kriebs A, Vaughan ME, Duglan D, Chan AB, Papp SJ, Nguyen M, Afetian ME, Lamia KA. The Liver Circadian Clock Modulates Biochemical and Physiological Responses to Metformin. J Biol Rhythms 2017; 32:345-358. [PMID: 28816632 DOI: 10.1177/0748730417710348] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Metformin is widely used in the treatment of type 2 diabetes to lower blood glucose. Although metformin is a relatively safe and effective drug, its clinical efficacy is variable and under certain circumstances it may contribute to life-threatening lactic acidosis. Thus, additional understanding of metformin pharmacokinetics and pharmacodynamics could provide important information regarding therapeutic use of this widely prescribed drug. Here we report a significant effect of time of day on acute blood glucose reduction in response to metformin administration and on blood lactate levels in healthy mice. Furthermore, we demonstrate that while metformin transport into hepatocytes is unaltered by time of day, the kinetics of metformin-induced activation of AMP-activated protein kinase (AMPK) in the liver are remarkably altered with circadian time. Liver-specific ablation of Bmal1 expression alters metformin induction of AMPK and blood glucose response but does not completely abolish time of day differences. Together, these data demonstrate that circadian rhythms affect the biological responses to metformin in a complex manner.
Collapse
Affiliation(s)
- Emma Henriksson
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA.,Department of Clinical Sciences, CRC, Lund University, Malmö, Sweden
| | - Anne-Laure Huber
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Erin K Soto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Anna Kriebs
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Megan E Vaughan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Drew Duglan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Alanna B Chan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Stephanie J Papp
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Madelena Nguyen
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Megan E Afetian
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Katja A Lamia
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| |
Collapse
|
214
|
Zhu B, Zhang Q, Pan Y, Mace EM, York B, Antoulas AC, Dacso CC, O'Malley BW. A Cell-Autonomous Mammalian 12 hr Clock Coordinates Metabolic and Stress Rhythms. Cell Metab 2017; 25:1305-1319.e9. [PMID: 28591634 PMCID: PMC5526350 DOI: 10.1016/j.cmet.2017.05.004] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 12/11/2016] [Accepted: 05/18/2017] [Indexed: 02/08/2023]
Abstract
Besides circadian rhythms, oscillations cycling with a 12 hr period exist. However, the prevalence, origin, regulation, and function of mammalian 12 hr rhythms remain elusive. Utilizing an unbiased mathematical approach identifying all superimposed oscillations, we uncovered prevalent 12 hr gene expression and metabolic rhythms in mouse liver, coupled with a physiological 12 hr unfolded protein response oscillation. The mammalian 12 hr rhythm is cell autonomous, driven by a dedicated 12 hr pacemaker distinct from the circadian clock, and can be entrained in vitro by metabolic and ER stress cues. Mechanistically, we identified XBP1s as a transcriptional regulator of the mammalian 12 hr clock. Downregulation of the 12 hr gene expression strongly correlates with human hepatic steatosis and steatohepatitis, implying its importance in maintaining metabolic homeostasis. The mammalian 12 hr rhythm of gene expression also is conserved in nematodes and crustaceans, indicating an ancient origin of the 12 hr clock. Our work sheds new light on how perturbed biological rhythms contribute to human disease.
Collapse
Affiliation(s)
- Bokai Zhu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Qiang Zhang
- Department of Electrical and Computer Engineering, Rice University, Houston, TX 77005, USA
| | - Yinghong Pan
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77004, USA
| | - Emily M Mace
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Brian York
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Athanasios C Antoulas
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Electrical and Computer Engineering, Rice University, Houston, TX 77005, USA; Max Planck Fellow Group for Data-Driven System Reduction and Identification, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg 39106, Germany
| | - Clifford C Dacso
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Electrical and Computer Engineering, Rice University, Houston, TX 77005, USA
| | - Bert W O'Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
215
|
Chaix A, Zarrinpar A, Panda S. The circadian coordination of cell biology. J Cell Biol 2017; 215:15-25. [PMID: 27738003 PMCID: PMC5057284 DOI: 10.1083/jcb.201603076] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 09/21/2016] [Indexed: 02/07/2023] Open
Abstract
Chaix et al. review how cells generate circadian oscillations and how circadian clocks control cell biology. Circadian clocks are cell-autonomous timing mechanisms that organize cell functions in a 24-h periodicity. In mammals, the main circadian oscillator consists of transcription–translation feedback loops composed of transcriptional regulators, enzymes, and scaffolds that generate and sustain daily oscillations of their own transcript and protein levels. The clock components and their targets impart rhythmic functions to many gene products through transcriptional, posttranscriptional, translational, and posttranslational mechanisms. This, in turn, temporally coordinates many signaling pathways, metabolic activity, organelles’ structure and functions, as well as the cell cycle and the tissue-specific functions of differentiated cells. When the functions of these circadian oscillators are disrupted by age, environment, or genetic mutation, the temporal coordination of cellular functions is lost, reducing organismal health and fitness.
Collapse
Affiliation(s)
- Amandine Chaix
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Amir Zarrinpar
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037 Division of Gastroenterology, University of California, San Diego, La Jolla, CA 92093
| | - Satchidananda Panda
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037
| |
Collapse
|
216
|
Mellor J. The molecular basis of metabolic cycles and their relationship to circadian rhythms. Nat Struct Mol Biol 2017; 23:1035-1044. [PMID: 27922609 DOI: 10.1038/nsmb.3311] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 09/23/2016] [Indexed: 12/12/2022]
Abstract
Metabolic cycles result from the partitioning of oxidative and reductive metabolism into rhythmic phases of gene expression and oscillating post-translational protein modifications. Relatively little is known about how these switches in gene expression are controlled, although recent studies have suggested that transcription itself may play a central role. This review explores the molecular basis of the metabolic and gene-expression oscillations in the yeast Saccharomyces cerevisiae, as well as how they relate to other biological time-keeping mechanisms, such as circadian rhythms.
Collapse
Affiliation(s)
- Jane Mellor
- Department of Biochemistry, University of Oxford, Oxford, UK
| |
Collapse
|
217
|
Inhibition of expression of the circadian clock gene Period causes metabolic abnormalities including repression of glycometabolism in Bombyx mori cells. Sci Rep 2017; 7:46258. [PMID: 28393918 PMCID: PMC5385517 DOI: 10.1038/srep46258] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 03/13/2017] [Indexed: 02/07/2023] Open
Abstract
Abnormalities in the circadian clock system are known to affect the body’s metabolic functions, though the molecular mechanisms responsible remain uncertain. In this study, we achieved continuous knockdown of B. mori Period (BmPer) gene expression in the B. mori ovary cell line (BmN), and generated a Per-KD B. mori model with developmental disorders including small individual cells and slow growth. We conducted cell metabolomics assays by gas chromatography/liquid chromatography-mass spectrometry and showed that knockdown of BmPer gene expression resulted in significant inhibition of glycometabolism. Amino acids that used glucose metabolites as a source were also down-regulated, while lipid metabolism and nucleotide metabolism were significantly up-regulated. Metabolite correlation analysis showed that pyruvate and lactate were closely related to glycometabolism, as well as to metabolites such as aspartate, alanine, and xanthine in other pathways. Further validation experiments showed that the activities of the key enzymes of glucose metabolism, hexokinase, phosphofructokinase, and citrate synthase, were significantly decreased and transcription of their encoding genes, as well as that of pyruvate kinase, were also significantly down-regulated. We concluded that inhibition of the circadian clock gene BmPer repressed glycometabolism, and may be associated with changes in cellular amino acid metabolism, and in cell growth and development.
Collapse
|
218
|
Krishnaiah SY, Wu G, Altman BJ, Growe J, Rhoades SD, Coldren F, Venkataraman A, Olarerin-George AO, Francey LJ, Mukherjee S, Girish S, Selby CP, Cal S, Er U, Sianati B, Sengupta A, Anafi RC, Kavakli IH, Sancar A, Baur JA, Dang CV, Hogenesch JB, Weljie AM. Clock Regulation of Metabolites Reveals Coupling between Transcription and Metabolism. Cell Metab 2017; 25:961-974.e4. [PMID: 28380384 PMCID: PMC5479132 DOI: 10.1016/j.cmet.2017.03.019] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 01/12/2017] [Accepted: 03/22/2017] [Indexed: 01/09/2023]
Abstract
The intricate connection between the circadian clock and metabolism remains poorly understood. We used high temporal resolution metabolite profiling to explore clock regulation of mouse liver and cell-autonomous metabolism. In liver, ∼50% of metabolites were circadian, with enrichment of nucleotide, amino acid, and methylation pathways. In U2 OS cells, 28% were circadian, including amino acids and NAD biosynthesis metabolites. Eighteen metabolites oscillated in both systems and a subset of these in primary hepatocytes. These 18 metabolites were enriched in methylation and amino acid pathways. To assess clock dependence of these rhythms, we used genetic perturbation. BMAL1 knockdown diminished metabolite rhythms, while CRY1 or CRY2 perturbation generally shortened or lengthened rhythms, respectively. Surprisingly, CRY1 knockdown induced 8 hr rhythms in amino acid, methylation, and vitamin metabolites, decoupling metabolite from transcriptional rhythms, with potential impact on nutrient sensing in vivo. These results provide the first comprehensive views of circadian liver and cell-autonomous metabolism.
Collapse
Affiliation(s)
- Saikumari Y Krishnaiah
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute of Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gang Wu
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Brian J Altman
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jacqueline Growe
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Seth D Rhoades
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute of Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Faith Coldren
- Institute of Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anand Venkataraman
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anthony O Olarerin-George
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lauren J Francey
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Sarmistha Mukherjee
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Saiveda Girish
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christopher P Selby
- Department of Biochemistry and Biophysics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sibel Cal
- Chemical and Biological Engineering and Molecular Biology and Genetics, Koc University, Rumeli Feneri Yolu, 34450 Sariyer, Istanbul, Turkey
| | - Ubeydullah Er
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute of Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bahareh Sianati
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute of Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Arjun Sengupta
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute of Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ron C Anafi
- Department of Medicine and Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - I Halil Kavakli
- Chemical and Biological Engineering and Molecular Biology and Genetics, Koc University, Rumeli Feneri Yolu, 34450 Sariyer, Istanbul, Turkey
| | - Aziz Sancar
- Department of Biochemistry and Biophysics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Joseph A Baur
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Chi V Dang
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John B Hogenesch
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| | - Aalim M Weljie
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute of Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
219
|
Abstract
Chronotherapeutics aim at treating illnesses according to the endogenous biologic rhythms, which moderate xenobiotic metabolism and cellular drug response. The molecular clocks present in individual cells involve approximately fifteen clock genes interconnected in regulatory feedback loops. They are coordinated by the suprachiasmatic nuclei, a hypothalamic pacemaker, which also adjusts the circadian rhythms to environmental cycles. As a result, many mechanisms of diseases and drug effects are controlled by the circadian timing system. Thus, the tolerability of nearly 500 medications varies by up to fivefold according to circadian scheduling, both in experimental models and/or patients. Moreover, treatment itself disrupted, maintained, or improved the circadian timing system as a function of drug timing. Improved patient outcomes on circadian-based treatments (chronotherapy) have been demonstrated in randomized clinical trials, especially for cancer and inflammatory diseases. However, recent technological advances have highlighted large interpatient differences in circadian functions resulting in significant variability in chronotherapy response. Such findings advocate for the advancement of personalized chronotherapeutics through interdisciplinary systems approaches. Thus, the combination of mathematical, statistical, technological, experimental, and clinical expertise is now shaping the development of dedicated devices and diagnostic and delivery algorithms enabling treatment individualization. In particular, multiscale systems chronopharmacology approaches currently combine mathematical modeling based on cellular and whole-body physiology to preclinical and clinical investigations toward the design of patient-tailored chronotherapies. We review recent systems research works aiming to the individualization of disease treatment, with emphasis on both cancer management and circadian timing system-resetting strategies for improving chronic disease control and patient outcomes.
Collapse
Affiliation(s)
- Annabelle Ballesta
- Warwick Medical School (A.B., P.F.I., R.D., F.A.L.) and Warwick Mathematics Institute (A.B., D.A.R.), University of Warwick, Coventry, United Kingdom; Warwick Systems Biology and Infectious Disease Epidemiological Research Centre, Senate House, Coventry, United Kingdom (A.B., P.F.I., R.D., D.A.R., F.A.L.); INSERM-Warwick European Associated Laboratory "Personalising Cancer Chronotherapy through Systems Medicine" (C2SysMed), Unité mixte de Recherche Scientifique 935, Centre National de Recherche Scientifique Campus, Villejuif, France (A.B., P.F.I., R.D., D.A.R., F.A.L.); and Queen Elisabeth Hospital Birmingham, University Hospitals Birmingham National Health Service Foundation Trust, Cancer Unit, Edgbaston Birmingham, United Kingdom (P.F.I., F.A.L.)
| | - Pasquale F Innominato
- Warwick Medical School (A.B., P.F.I., R.D., F.A.L.) and Warwick Mathematics Institute (A.B., D.A.R.), University of Warwick, Coventry, United Kingdom; Warwick Systems Biology and Infectious Disease Epidemiological Research Centre, Senate House, Coventry, United Kingdom (A.B., P.F.I., R.D., D.A.R., F.A.L.); INSERM-Warwick European Associated Laboratory "Personalising Cancer Chronotherapy through Systems Medicine" (C2SysMed), Unité mixte de Recherche Scientifique 935, Centre National de Recherche Scientifique Campus, Villejuif, France (A.B., P.F.I., R.D., D.A.R., F.A.L.); and Queen Elisabeth Hospital Birmingham, University Hospitals Birmingham National Health Service Foundation Trust, Cancer Unit, Edgbaston Birmingham, United Kingdom (P.F.I., F.A.L.)
| | - Robert Dallmann
- Warwick Medical School (A.B., P.F.I., R.D., F.A.L.) and Warwick Mathematics Institute (A.B., D.A.R.), University of Warwick, Coventry, United Kingdom; Warwick Systems Biology and Infectious Disease Epidemiological Research Centre, Senate House, Coventry, United Kingdom (A.B., P.F.I., R.D., D.A.R., F.A.L.); INSERM-Warwick European Associated Laboratory "Personalising Cancer Chronotherapy through Systems Medicine" (C2SysMed), Unité mixte de Recherche Scientifique 935, Centre National de Recherche Scientifique Campus, Villejuif, France (A.B., P.F.I., R.D., D.A.R., F.A.L.); and Queen Elisabeth Hospital Birmingham, University Hospitals Birmingham National Health Service Foundation Trust, Cancer Unit, Edgbaston Birmingham, United Kingdom (P.F.I., F.A.L.)
| | - David A Rand
- Warwick Medical School (A.B., P.F.I., R.D., F.A.L.) and Warwick Mathematics Institute (A.B., D.A.R.), University of Warwick, Coventry, United Kingdom; Warwick Systems Biology and Infectious Disease Epidemiological Research Centre, Senate House, Coventry, United Kingdom (A.B., P.F.I., R.D., D.A.R., F.A.L.); INSERM-Warwick European Associated Laboratory "Personalising Cancer Chronotherapy through Systems Medicine" (C2SysMed), Unité mixte de Recherche Scientifique 935, Centre National de Recherche Scientifique Campus, Villejuif, France (A.B., P.F.I., R.D., D.A.R., F.A.L.); and Queen Elisabeth Hospital Birmingham, University Hospitals Birmingham National Health Service Foundation Trust, Cancer Unit, Edgbaston Birmingham, United Kingdom (P.F.I., F.A.L.)
| | - Francis A Lévi
- Warwick Medical School (A.B., P.F.I., R.D., F.A.L.) and Warwick Mathematics Institute (A.B., D.A.R.), University of Warwick, Coventry, United Kingdom; Warwick Systems Biology and Infectious Disease Epidemiological Research Centre, Senate House, Coventry, United Kingdom (A.B., P.F.I., R.D., D.A.R., F.A.L.); INSERM-Warwick European Associated Laboratory "Personalising Cancer Chronotherapy through Systems Medicine" (C2SysMed), Unité mixte de Recherche Scientifique 935, Centre National de Recherche Scientifique Campus, Villejuif, France (A.B., P.F.I., R.D., D.A.R., F.A.L.); and Queen Elisabeth Hospital Birmingham, University Hospitals Birmingham National Health Service Foundation Trust, Cancer Unit, Edgbaston Birmingham, United Kingdom (P.F.I., F.A.L.)
| |
Collapse
|
220
|
Koppenol-Raab M, Sjoelund V, Manes NP, Gottschalk RA, Dutta B, Benet ZL, Fraser IDC, Nita-Lazar A. Proteome and Secretome Analysis Reveals Differential Post-transcriptional Regulation of Toll-like Receptor Responses. Mol Cell Proteomics 2017; 16:S172-S186. [PMID: 28235783 PMCID: PMC5393387 DOI: 10.1074/mcp.m116.064261] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 02/24/2017] [Indexed: 12/27/2022] Open
Abstract
The innate immune system is the organism's first line of defense against pathogens. Pattern recognition receptors (PRRs) are responsible for sensing the presence of pathogen-associated molecules. The prototypic PRRs, the membrane-bound receptors of the Toll-like receptor (TLR) family, recognize pathogen-associated molecular patterns (PAMPs) and initiate an innate immune response through signaling pathways that depend on the adaptor molecules MyD88 and TRIF. Deciphering the differences in the complex signaling events that lead to pathogen recognition and initiation of the correct response remains challenging. Here we report the discovery of temporal changes in the protein signaling components involved in innate immunity. Using an integrated strategy combining unbiased proteomics, transcriptomics and macrophage stimulations with three different PAMPs, we identified differences in signaling between individual TLRs and revealed specifics of pathway regulation at the protein level.
Collapse
Affiliation(s)
- Marijke Koppenol-Raab
- From the ‡Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892
| | - Virginie Sjoelund
- From the ‡Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892
| | - Nathan P Manes
- From the ‡Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892
| | - Rachel A Gottschalk
- From the ‡Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892
| | - Bhaskar Dutta
- From the ‡Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892
| | - Zachary L Benet
- From the ‡Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892
| | - Iain D C Fraser
- From the ‡Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892
| | - Aleksandra Nita-Lazar
- From the ‡Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892
| |
Collapse
|
221
|
Chiang CK, Xu B, Mehta N, Mayne J, Sun WYL, Cheng K, Ning Z, Dong J, Zou H, Cheng HYM, Figeys D. Phosphoproteome Profiling Reveals Circadian Clock Regulation of Posttranslational Modifications in the Murine Hippocampus. Front Neurol 2017; 8:110. [PMID: 28382018 PMCID: PMC5360755 DOI: 10.3389/fneur.2017.00110] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 03/08/2017] [Indexed: 12/20/2022] Open
Abstract
The circadian clock is an endogenous oscillator that drives daily rhythms in physiology, behavior, and gene expression. The underlying mechanisms of circadian timekeeping are cell-autonomous and involve oscillatory expression of core clock genes that is driven by interconnecting transcription–translation feedback loops (TTFLs). Circadian clock TTFLs are further regulated by posttranslational modifications, in particular, phosphorylation. The hippocampus plays an important role in spatial memory and the conversion of short- to long-term memory. Several studies have reported the presence of a peripheral oscillator in the hippocampus and have highlighted the importance of circadian regulation in memory formation. Given the general importance of phosphorylation in circadian clock regulation, we performed global quantitative proteome and phosphoproteome analyses of the murine hippocampus across the circadian cycle, applying spiked-in labeled reference and high accuracy mass spectrometry (MS). Of the 3,052 proteins and 2,868 phosphosites on 1,368 proteins that were accurately quantified, 1.7% of proteins and 5.2% of phosphorylation events exhibited time-of-day-dependent expression profiles. The majority of circadian phosphopeptides displayed abrupt fluctuations at mid-to-late day without underlying rhythms of protein abundance. Bioinformatic analysis of cyclic phosphorylation events revealed their diverse distribution in different biological pathways, most notably, cytoskeletal organization and neuronal morphogenesis. This study provides the first large-scale, quantitative MS analysis of the circadian phosphoproteome and proteome of the murine hippocampus and highlights the significance of rhythmic regulation at the posttranslational level in this peripheral oscillator. In addition to providing molecular insights into the hippocampal circadian clock, our results will assist in the understanding of genetic factors that underlie rhythms-associated pathological states of the hippocampus.
Collapse
Affiliation(s)
- Cheng-Kang Chiang
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada; Department of Chemistry, National Dong Hwa University, Shoufeng, Hualien, Taiwan
| | - Bo Xu
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa , Ottawa, ON , Canada
| | - Neel Mehta
- Department of Biology, University of Toronto Mississauga , Mississauga, ON , Canada
| | - Janice Mayne
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa , Ottawa, ON , Canada
| | - Warren Y L Sun
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa , Ottawa, ON , Canada
| | - Kai Cheng
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa , Ottawa, ON , Canada
| | - Zhibin Ning
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa , Ottawa, ON , Canada
| | - Jing Dong
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences , Dalian , China
| | - Hanfa Zou
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences , Dalian , China
| | - Hai-Ying Mary Cheng
- Department of Biology, University of Toronto Mississauga , Mississauga, ON , Canada
| | - Daniel Figeys
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada; Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON, Canada; Canadian Institute for Advanced Research, Toronto, ON, Canada
| |
Collapse
|
222
|
Mermet J, Yeung J, Naef F. Systems Chronobiology: Global Analysis of Gene Regulation in a 24-Hour Periodic World. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a028720. [PMID: 27920039 DOI: 10.1101/cshperspect.a028720] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Mammals have evolved an internal timing system, the circadian clock, which synchronizes physiology and behavior to the daily light and dark cycles of the Earth. The master clock, located in the suprachiasmatic nucleus (SCN) of the brain, takes fluctuating light input from the retina and synchronizes other tissues to the same internal rhythm. The molecular clocks that drive these circadian rhythms are ticking in nearly all cells in the body. Efforts in systems chronobiology are now being directed at understanding, on a comprehensive scale, how the circadian clock controls different layers of gene regulation to provide robust timing cues at the cellular and tissue level. In this review, we introduce some basic concepts underlying periodicity of gene regulation, and then highlight recent genome-wide investigations on the propagation of rhythms across multiple regulatory layers in mammals, all the way from chromatin conformation to protein accumulation.
Collapse
Affiliation(s)
- Jérôme Mermet
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Jake Yeung
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Felix Naef
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| |
Collapse
|
223
|
Abstract
This review summarizes various mathematical models of cell-autonomous mammalian circadian clock. We present the basics necessary for understanding of the cell-autonomous mammalian circadian oscillator, modern experimental data essential for its reconstruction and some special problems related to the validation of mathematical circadian oscillator models. This work compares existing mathematical models of circadian oscillator and the results of the computational studies of the oscillating systems. Finally, we discuss applications of the mathematical models of mammalian circadian oscillator for solving specific problems in circadian rhythm biology.
Collapse
|
224
|
Millius A, Ueda HR. Systems Biology-Derived Discoveries of Intrinsic Clocks. Front Neurol 2017; 8:25. [PMID: 28220104 PMCID: PMC5292584 DOI: 10.3389/fneur.2017.00025] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 01/17/2017] [Indexed: 12/19/2022] Open
Abstract
A systems approach to studying biology uses a variety of mathematical, computational, and engineering tools to holistically understand and model properties of cells, tissues, and organisms. Building from early biochemical, genetic, and physiological studies, systems biology became established through the development of genome-wide methods, high-throughput procedures, modern computational processing power, and bioinformatics. Here, we highlight a variety of systems approaches to the study of biological rhythms that occur with a 24-h period-circadian rhythms. We review how systems methods have helped to elucidate complex behaviors of the circadian clock including temperature compensation, rhythmicity, and robustness. Finally, we explain the contribution of systems biology to the transcription-translation feedback loop and posttranslational oscillator models of circadian rhythms and describe new technologies and "-omics" approaches to understand circadian timekeeping and neurophysiology.
Collapse
Affiliation(s)
- Arthur Millius
- Laboratory for Synthetic Biology, RIKEN Quantitative Biology Center, Suita, Osaka, Japan
| | - Hiroki R. Ueda
- Laboratory for Synthetic Biology, RIKEN Quantitative Biology Center, Suita, Osaka, Japan
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
225
|
Abstract
Circadian rhythms govern multiple aspects of animal metabolism. Transcriptome-, proteome- and metabolome-wide measurements have revealed widespread circadian rhythms in metabolism governed by a cellular genetic oscillator, the circadian core clock. However, it remains unclear if and under which conditions transcriptional rhythms cause rhythms in particular metabolites and metabolic fluxes. Here, we analyzed the circadian orchestration of metabolic pathways by direct measurement of enzyme activities, analysis of transcriptome data, and developing a theoretical method called circadian response analysis. Contrary to a common assumption, we found that pronounced rhythms in metabolic pathways are often favored by separation rather than alignment in the times of peak activity of key enzymes. This property holds true for a set of metabolic pathway motifs (e.g., linear chains and branching points) and also under the conditions of fast kinetics typical for metabolic reactions. By circadian response analysis of pathway motifs, we determined exact timing separation constraints on rhythmic enzyme activities that allow for substantial rhythms in pathway flux and metabolite concentrations. Direct measurements of circadian enzyme activities in mouse skeletal muscle confirmed that such timing separation occurs in vivo.
Collapse
|
226
|
What computational non-targeted mass spectrometry-based metabolomics can gain from shotgun proteomics. Curr Opin Biotechnol 2017; 43:141-146. [DOI: 10.1016/j.copbio.2016.11.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 11/25/2016] [Accepted: 11/28/2016] [Indexed: 01/25/2023]
|
227
|
Robles MS, Humphrey SJ, Mann M. Phosphorylation Is a Central Mechanism for Circadian Control of Metabolism and Physiology. Cell Metab 2017; 25:118-127. [PMID: 27818261 DOI: 10.1016/j.cmet.2016.10.004] [Citation(s) in RCA: 264] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 09/12/2016] [Accepted: 10/05/2016] [Indexed: 12/21/2022]
Abstract
Circadian clocks are self-sustainable endogenous oscillators, present in virtually every cell, driving daily cycles of metabolism and physiology. The molecular mechanism of the circadian clock is based on interconnected transcriptional and translational feedback loops. While many studies have addressed circadian rhythms of the transcriptome and, to a lesser extent, the proteome, none have investigated the phosphoproteome. We apply mass spectrometry-based phosphoproteomics to obtain the first global in vivo quantification of circadian phosphorylation in mammals. Of more than 20,000 phosphosites, 25% significantly oscillate in the mouse liver, including novel sites on core clock proteins. The extent and amplitude of phosphorylation cycles far exceeds those observed in RNA and protein abundance. Our data indicate a dominant regulatory role for phosphorylation-dependent circadian tuning of signaling pathways. This allows the organism to integrate different signals and rapidly and economically respond to daily changes in nutrient availability and physiological states.
Collapse
Affiliation(s)
- Maria S Robles
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Sean J Humphrey
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried 82152, Germany.
| |
Collapse
|
228
|
Abstract
Circadian clocks regulate most aspects of physiology and metabolism. Genome-wide approaches have uncovered widespread circadian rhythms in the transcriptome, cistrome, and epigenome of mice, and now two proteomics studies in this issue (Robles et al., 2016; Wang et al., 2016) reveal extensive circadian regulation of the nuclear and phosphoproteome.
Collapse
Affiliation(s)
- Joseph S Takahashi
- Howard Hughes Medical Institute and Department of Neuroscience, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9111 USA.
| |
Collapse
|
229
|
Wang J, Mauvoisin D, Martin E, Atger F, Galindo AN, Dayon L, Sizzano F, Palini A, Kussmann M, Waridel P, Quadroni M, Dulić V, Naef F, Gachon F. Nuclear Proteomics Uncovers Diurnal Regulatory Landscapes in Mouse Liver. Cell Metab 2017; 25:102-117. [PMID: 27818260 PMCID: PMC5241201 DOI: 10.1016/j.cmet.2016.10.003] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 07/25/2016] [Accepted: 10/05/2016] [Indexed: 12/12/2022]
Abstract
Diurnal oscillations of gene expression controlled by the circadian clock and its connected feeding rhythm enable organisms to coordinate their physiologies with daily environmental cycles. While available techniques yielded crucial insights into regulation at the transcriptional level, much less is known about temporally controlled functions within the nucleus and their regulation at the protein level. Here, we quantified the temporal nuclear accumulation of proteins and phosphoproteins from mouse liver by SILAC proteomics. We identified around 5,000 nuclear proteins, over 500 of which showed a diurnal accumulation. Parallel analysis of the nuclear phosphoproteome enabled the inference of the temporal activity of kinases accounting for rhythmic phosphorylation. Many identified rhythmic proteins were parts of nuclear complexes involved in transcriptional regulation, ribosome biogenesis, DNA repair, and the cell cycle and its potentially associated diurnal rhythm of hepatocyte polyploidy. Taken together, these findings provide unprecedented insights into the diurnal regulatory landscape of the mouse liver nucleus.
Collapse
Affiliation(s)
- Jingkui Wang
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Daniel Mauvoisin
- Department of Diabetes and Circadian Rhythms, Nestlé Institute of Health Sciences, CH-1015 Lausanne, Switzerland
| | - Eva Martin
- Department of Diabetes and Circadian Rhythms, Nestlé Institute of Health Sciences, CH-1015 Lausanne, Switzerland
| | - Florian Atger
- Department of Diabetes and Circadian Rhythms, Nestlé Institute of Health Sciences, CH-1015 Lausanne, Switzerland; Department of Pharmacology and Toxicology, University of Lausanne, CH-1015 Lausanne, Switzerland
| | - Antonio Núñez Galindo
- Systems Nutrition, Metabonomics, and Proteomics, Nestlé Institute of Health Sciences, CH-1015 Lausanne, Switzerland
| | - Loïc Dayon
- Systems Nutrition, Metabonomics, and Proteomics, Nestlé Institute of Health Sciences, CH-1015 Lausanne, Switzerland
| | - Federico Sizzano
- Department of Cell Biology, Nestlé Institute of Health Sciences, CH-1015 Lausanne, Switzerland
| | - Alessio Palini
- Department of Cell Biology, Nestlé Institute of Health Sciences, CH-1015 Lausanne, Switzerland
| | - Martin Kussmann
- Systems Nutrition, Metabonomics, and Proteomics, Nestlé Institute of Health Sciences, CH-1015 Lausanne, Switzerland
| | - Patrice Waridel
- Protein Analysis Facility, University of Lausanne, CH-1015 Lausanne, Switzerland
| | - Manfredo Quadroni
- Protein Analysis Facility, University of Lausanne, CH-1015 Lausanne, Switzerland
| | - Vjekoslav Dulić
- CNRS, UMR 5535, Institut de Génétique Moléculaire de Montpellier, 34090 Montpellier, France
| | - Felix Naef
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland.
| | - Frédéric Gachon
- Department of Diabetes and Circadian Rhythms, Nestlé Institute of Health Sciences, CH-1015 Lausanne, Switzerland; School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland.
| |
Collapse
|
230
|
Atger F, Mauvoisin D, Weger B, Gobet C, Gachon F. Regulation of Mammalian Physiology by Interconnected Circadian and Feeding Rhythms. Front Endocrinol (Lausanne) 2017; 8:42. [PMID: 28337174 PMCID: PMC5340782 DOI: 10.3389/fendo.2017.00042] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 02/17/2017] [Indexed: 12/29/2022] Open
Abstract
Circadian clocks are endogenous timekeeping systems that adapt in an anticipatory fashion the physiology and behavior of most living organisms. In mammals, the master pacemaker resides in the suprachiasmatic nucleus and entrains peripheral clocks using a wide range of signals that differentially schedule physiology and gene expression in a tissue-specific manner. The peripheral clocks, such as those found in the liver, are particularly sensitive to rhythmic external cues like feeding behavior, which modulate the phase and amplitude of rhythmic gene expression. Consequently, the liver clock temporally tunes the expression of many genes involved in metabolism and physiology. However, the circadian modulation of cellular functions also relies on multiple layers of posttranscriptional and posttranslational regulation. Strikingly, these additional regulatory events may happen independently of any transcriptional oscillations, showing that complex regulatory networks ultimately drive circadian output functions. These rhythmic events also integrate feeding-related cues and adapt various metabolic processes to food availability schedules. The importance of such temporal regulation of metabolism is illustrated by metabolic dysfunctions and diseases resulting from circadian clock disruption or inappropriate feeding patterns. Therefore, the study of circadian clocks and rhythmic feeding behavior should be of interest to further advance our understanding of the prevention and therapy of metabolic diseases.
Collapse
Affiliation(s)
- Florian Atger
- Department of Diabetes and Circadian Rhythms, Nestlé Institute of Health Sciences, Lausanne, Switzerland
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Daniel Mauvoisin
- Department of Diabetes and Circadian Rhythms, Nestlé Institute of Health Sciences, Lausanne, Switzerland
- School of Life Sciences, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Benjamin Weger
- Department of Diabetes and Circadian Rhythms, Nestlé Institute of Health Sciences, Lausanne, Switzerland
| | - Cédric Gobet
- Department of Diabetes and Circadian Rhythms, Nestlé Institute of Health Sciences, Lausanne, Switzerland
- School of Life Sciences, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Frédéric Gachon
- Department of Diabetes and Circadian Rhythms, Nestlé Institute of Health Sciences, Lausanne, Switzerland
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- *Correspondence: Frédéric Gachon,
| |
Collapse
|
231
|
Liu Y, Beyer A, Aebersold R. On the Dependency of Cellular Protein Levels on mRNA Abundance. Cell 2016; 165:535-50. [PMID: 27104977 DOI: 10.1016/j.cell.2016.03.014] [Citation(s) in RCA: 2013] [Impact Index Per Article: 223.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Indexed: 12/30/2022]
Abstract
The question of how genomic information is expressed to determine phenotypes is of central importance for basic and translational life science research and has been studied by transcriptomic and proteomic profiling. Here, we review the relationship between protein and mRNA levels under various scenarios, such as steady state, long-term state changes, and short-term adaptation, demonstrating the complexity of gene expression regulation, especially during dynamic transitions. The spatial and temporal variations of mRNAs, as well as the local availability of resources for protein biosynthesis, strongly influence the relationship between protein levels and their coding transcripts. We further discuss the buffering of mRNA fluctuations at the level of protein concentrations. We conclude that transcript levels by themselves are not sufficient to predict protein levels in many scenarios and to thus explain genotype-phenotype relationships and that high-quality data quantifying different levels of gene expression are indispensable for the complete understanding of biological processes.
Collapse
Affiliation(s)
- Yansheng Liu
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Andreas Beyer
- Cellular Networks and Systems Biology, University of Cologne, CECAD, Joseph-Stelzmann-Strasse 26, Cologne 50931, Germany.
| | - Ruedi Aebersold
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, 8093 Zurich, Switzerland; Faculty of Science, University of Zurich, 8057 Zurich, Switzerland.
| |
Collapse
|
232
|
Pett JP, Korenčič A, Wesener F, Kramer A, Herzel H. Feedback Loops of the Mammalian Circadian Clock Constitute Repressilator. PLoS Comput Biol 2016; 12:e1005266. [PMID: 27942033 PMCID: PMC5189953 DOI: 10.1371/journal.pcbi.1005266] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 12/27/2016] [Accepted: 11/27/2016] [Indexed: 11/18/2022] Open
Abstract
Mammals evolved an endogenous timing system to coordinate their physiology and behaviour to the 24h period of the solar day. While it is well accepted that circadian rhythms are generated by intracellular transcriptional feedback loops, it is still debated which network motifs are necessary and sufficient for generating self-sustained oscillations. Here, we systematically explore a data-based circadian oscillator model with multiple negative and positive feedback loops and identify a series of three subsequent inhibitions known as "repressilator" as a core element of the mammalian circadian oscillator. The central role of the repressilator motif is consistent with time-resolved ChIP-seq experiments of circadian clock transcription factors and loss of rhythmicity in core clock gene knockouts.
Collapse
Affiliation(s)
- J. Patrick Pett
- Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Anja Korenčič
- Center for Functional Genomics and Biochips, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Felix Wesener
- Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Achim Kramer
- Laboratory of Chronobiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Hanspeter Herzel
- Institute for Theoretical Biology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- * E-mail:
| |
Collapse
|
233
|
Potter GDM, Skene DJ, Arendt J, Cade JE, Grant PJ, Hardie LJ. Circadian Rhythm and Sleep Disruption: Causes, Metabolic Consequences, and Countermeasures. Endocr Rev 2016; 37:584-608. [PMID: 27763782 PMCID: PMC5142605 DOI: 10.1210/er.2016-1083] [Citation(s) in RCA: 372] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Circadian (∼24-hour) timing systems pervade all kingdoms of life and temporally optimize behavior and physiology in humans. Relatively recent changes to our environments, such as the introduction of artificial lighting, can disorganize the circadian system, from the level of the molecular clocks that regulate the timing of cellular activities to the level of synchronization between our daily cycles of behavior and the solar day. Sleep/wake cycles are intertwined with the circadian system, and global trends indicate that these, too, are increasingly subject to disruption. A large proportion of the world's population is at increased risk of environmentally driven circadian rhythm and sleep disruption, and a minority of individuals are also genetically predisposed to circadian misalignment and sleep disorders. The consequences of disruption to the circadian system and sleep are profound and include myriad metabolic ramifications, some of which may be compounded by adverse effects on dietary choices. If not addressed, the deleterious effects of such disruption will continue to cause widespread health problems; therefore, implementation of the numerous behavioral and pharmaceutical interventions that can help restore circadian system alignment and enhance sleep will be important.
Collapse
Affiliation(s)
- Gregory D M Potter
- Division of Epidemiology and Biostatistics (G.D.M.P., L.J.H.), LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom; Chronobiology Section (D.J.S., J.A.), Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, United Kingdom; Nutritional Epidemiology Group (J.E.C.), School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, United Kingdom; and Division of Cardiovascular & Diabetes Research (P.J.G.), LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Debra J Skene
- Division of Epidemiology and Biostatistics (G.D.M.P., L.J.H.), LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom; Chronobiology Section (D.J.S., J.A.), Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, United Kingdom; Nutritional Epidemiology Group (J.E.C.), School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, United Kingdom; and Division of Cardiovascular & Diabetes Research (P.J.G.), LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Josephine Arendt
- Division of Epidemiology and Biostatistics (G.D.M.P., L.J.H.), LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom; Chronobiology Section (D.J.S., J.A.), Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, United Kingdom; Nutritional Epidemiology Group (J.E.C.), School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, United Kingdom; and Division of Cardiovascular & Diabetes Research (P.J.G.), LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Janet E Cade
- Division of Epidemiology and Biostatistics (G.D.M.P., L.J.H.), LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom; Chronobiology Section (D.J.S., J.A.), Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, United Kingdom; Nutritional Epidemiology Group (J.E.C.), School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, United Kingdom; and Division of Cardiovascular & Diabetes Research (P.J.G.), LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Peter J Grant
- Division of Epidemiology and Biostatistics (G.D.M.P., L.J.H.), LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom; Chronobiology Section (D.J.S., J.A.), Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, United Kingdom; Nutritional Epidemiology Group (J.E.C.), School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, United Kingdom; and Division of Cardiovascular & Diabetes Research (P.J.G.), LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Laura J Hardie
- Division of Epidemiology and Biostatistics (G.D.M.P., L.J.H.), LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom; Chronobiology Section (D.J.S., J.A.), Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, United Kingdom; Nutritional Epidemiology Group (J.E.C.), School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, United Kingdom; and Division of Cardiovascular & Diabetes Research (P.J.G.), LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom
| |
Collapse
|
234
|
Abstract
A majority of mammalian genes exhibit daily fluctuations in expression levels, making circadian expression rhythms the largest known regulatory network in normal physiology. Cell-autonomous circadian clocks interact with daily light-dark and feeding-fasting cycles to generate approximately 24-hour oscillations in the function of thousands of genes. Circadian expression of secreted molecules and signaling components transmits timing information between cells and tissues. Such intra- and intercellular daily rhythms optimize physiology both by managing energy use and by temporally segregating incompatible processes. Experimental animal models and epidemiological data indicate that chronic circadian rhythm disruption increases the risk of metabolic diseases. Conversely, time-restricted feeding, which imposes daily cycles of feeding and fasting without caloric reduction, sustains robust diurnal rhythms and can alleviate metabolic diseases. These findings highlight an integrative role of circadian rhythms in physiology and offer a new perspective for treating chronic diseases in which metabolic disruption is a hallmark.
Collapse
Affiliation(s)
- Satchidananda Panda
- Salk Institute of Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
235
|
Tyanova S, Temu T, Cox J. The MaxQuant computational platform for mass spectrometry-based shotgun proteomics. Nat Protoc 2016; 11:2301-2319. [PMID: 27809316 DOI: 10.1038/nprot.2016.136] [Citation(s) in RCA: 3144] [Impact Index Per Article: 349.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
MaxQuant is one of the most frequently used platforms for mass-spectrometry (MS)-based proteomics data analysis. Since its first release in 2008, it has grown substantially in functionality and can be used in conjunction with more MS platforms. Here we present an updated protocol covering the most important basic computational workflows, including those designed for quantitative label-free proteomics, MS1-level labeling and isobaric labeling techniques. This protocol presents a complete description of the parameters used in MaxQuant, as well as of the configuration options of its integrated search engine, Andromeda. This protocol update describes an adaptation of an existing protocol that substantially modifies the technique. Important concepts of shotgun proteomics and their implementation in MaxQuant are briefly reviewed, including different quantification strategies and the control of false-discovery rates (FDRs), as well as the analysis of post-translational modifications (PTMs). The MaxQuant output tables, which contain information about quantification of proteins and PTMs, are explained in detail. Furthermore, we provide a short version of the workflow that is applicable to data sets with simple and standard experimental designs. The MaxQuant algorithms are efficiently parallelized on multiple processors and scale well from desktop computers to servers with many cores. The software is written in C# and is freely available at http://www.maxquant.org.
Collapse
Affiliation(s)
- Stefka Tyanova
- Computational Systems Biochemistry, Max-Planck Institute for Biochemistry, Martinsried, Germany
| | - Tikira Temu
- Computational Systems Biochemistry, Max-Planck Institute for Biochemistry, Martinsried, Germany
| | - Juergen Cox
- Computational Systems Biochemistry, Max-Planck Institute for Biochemistry, Martinsried, Germany
| |
Collapse
|
236
|
Minard AY, Wong MKL, Chaudhuri R, Tan SX, Humphrey SJ, Parker BL, Yang JY, Laybutt DR, Cooney GJ, Coster ACF, Stöckli J, James DE. Hyperactivation of the Insulin Signaling Pathway Improves Intracellular Proteostasis by Coordinately Up-regulating the Proteostatic Machinery in Adipocytes. J Biol Chem 2016; 291:25629-25640. [PMID: 27738101 DOI: 10.1074/jbc.m116.741140] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 10/11/2016] [Indexed: 01/25/2023] Open
Abstract
Hyperinsulinemia, which is associated with aging and metabolic disease, may lead to defective protein homeostasis (proteostasis) due to hyperactivation of insulin-sensitive pathways such as protein synthesis. We investigated the effect of chronic hyperinsulinemia on proteostasis by generating a time-resolved map of insulin-regulated protein turnover in adipocytes using metabolic pulse-chase labeling and high resolution mass spectrometry. Hyperinsulinemia increased the synthesis of nearly half of all detected proteins and did not affect protein degradation despite suppressing autophagy. Unexpectedly, this marked elevation in protein synthesis was accompanied by enhanced protein stability and folding and not by markers of proteostasis stress such as protein carbonylation and aggregation. The improvement in proteostasis was attributed to a coordinate up-regulation of proteins in the global proteostasis network, including ribosomal, proteasomal, chaperone, and endoplasmic reticulum/mitochondrial unfolded protein response proteins. We conclude that defects associated with hyperactivation of the insulin signaling pathway are unlikely attributed to defective proteostasis because up-regulation of protein synthesis by insulin is accompanied by up-regulation of proteostatic machinery.
Collapse
Affiliation(s)
- Annabel Y Minard
- From The Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia.,Charles Perkins Centre, School of Life Environmental Sciences
| | - Martin K L Wong
- From The Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia.,Charles Perkins Centre, School of Life Environmental Sciences.,School of Physics
| | - Rima Chaudhuri
- Charles Perkins Centre, School of Life Environmental Sciences
| | - Shi-Xiong Tan
- From The Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia
| | - Sean J Humphrey
- Charles Perkins Centre, School of Life Environmental Sciences
| | | | | | - D Ross Laybutt
- From The Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia
| | | | - Adelle C F Coster
- Department of Applied Mathematics, School of Mathematics and Statistics, University of New South Wales, Sydney, New South Wales 2052, Australia
| | | | - David E James
- Charles Perkins Centre, School of Life Environmental Sciences, .,School of Medicine, University of Sydney, Sydney, New South Wales 2006, Australia, and
| |
Collapse
|
237
|
Rose CM, Isasa M, Ordureau A, Prado MA, Beausoleil SA, Jedrychowski MP, Finley DJ, Harper JW, Gygi SP. Highly Multiplexed Quantitative Mass Spectrometry Analysis of Ubiquitylomes. Cell Syst 2016; 3:395-403.e4. [PMID: 27667366 DOI: 10.1016/j.cels.2016.08.009] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 06/21/2016] [Accepted: 08/05/2016] [Indexed: 12/26/2022]
Abstract
System-wide quantitative analysis of ubiquitylomes has proven to be a valuable tool for elucidating targets and mechanisms of the ubiquitin-driven signaling systems, as well as gaining insights into neurodegenerative diseases and cancer. Current mass spectrometry methods for ubiquitylome detection require large amounts of starting material and rely on stochastic data collection to increase replicate analyses. We describe a method compatible with cell line and tissue samples for large-scale quantification of 5,000-9,000 ubiquitylation forms across ten samples simultaneously. Using this method, we reveal site-specific ubiquitylation in mammalian brain and liver tissues, as well as in cancer cells undergoing proteasome inhibition. To demonstrate the power of the approach for signal-dependent ubiquitylation, we examined protein and ubiquitylation dynamics for mitochondria undergoing PARKIN- and PINK1-dependent mitophagy. This analysis revealed the largest collection of PARKIN- and PINK1-dependent ubiquitylation targets to date in a single experiment, and it also revealed a subset of proteins recruited to the mitochondria during mitophagy.
Collapse
Affiliation(s)
- Christopher M Rose
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Marta Isasa
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Alban Ordureau
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Miguel A Prado
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | | | | | - Daniel J Finley
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - J Wade Harper
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
238
|
Circadian clock regulation of mRNA translation through eukaryotic elongation factor eEF-2. Proc Natl Acad Sci U S A 2016; 113:9605-10. [PMID: 27506798 DOI: 10.1073/pnas.1525268113] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The circadian clock has a profound effect on gene regulation, controlling rhythmic transcript accumulation for up to half of expressed genes in eukaryotes. Evidence also exists for clock control of mRNA translation, but the extent and mechanisms for this regulation are not known. In Neurospora crassa, the circadian clock generates daily rhythms in the activation of conserved mitogen-activated protein kinase (MAPK) pathways when cells are grown in constant conditions, including rhythmic activation of the well-characterized p38 osmosensing (OS) MAPK pathway. Rhythmic phosphorylation of the MAPK OS-2 (P-OS-2) leads to temporal control of downstream targets of OS-2. We show that osmotic stress in N. crassa induced the phosphorylation of a eukaryotic elongation factor-2 (eEF-2) kinase, radiation sensitivity complementing kinase-2 (RCK-2), and that RCK-2 is necessary for high-level phosphorylation of eEF-2, a key regulator of translation elongation. The levels of phosphorylated RCK-2 and phosphorylated eEF-2 cycle in abundance in wild-type cells but not in cells deleted for OS-2 or the core clock component FREQUENCY (FRQ). Translation extracts from cells grown in constant conditions show decreased translational activity in the late subjective morning, coincident with the peak in eEF-2 phosphorylation, and rhythmic translation of glutathione S-transferase (GST-3) from constitutive mRNA levels in vivo is dependent on circadian regulation of eEF-2 activity. In contrast, rhythms in phosphorylated eEF-2 levels are not necessary for rhythms in accumulation of the clock protein FRQ, indicating that clock control of eEF-2 activity promotes rhythmic translation of specific mRNAs.
Collapse
|
239
|
Heegaard NHH, Carlsen AL, Lilje B, Ng KL, Rønne ME, Jørgensen HL, Sennels H, Fahrenkrug J. Diurnal Variations of Human Circulating Cell-Free Micro-RNA. PLoS One 2016; 11:e0160577. [PMID: 27494182 PMCID: PMC4975411 DOI: 10.1371/journal.pone.0160577] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 07/21/2016] [Indexed: 12/26/2022] Open
Abstract
A 24-hour light and dark cycle-dependent rhythmicity pervades physiological processes in virtually all living organisms including humans. These regular oscillations are caused by external cues to endogenous, independent biological time-keeping systems (clocks). The rhythm is reflected by gene expression that varies in a circadian and specific fashion in different organs and tissues and is regulated largely by dynamic epigenetic and post-transcriptional mechanisms. This leads to well-documented oscillations of specific electrolytes, hormones, metabolites, and plasma proteins in blood samples. An emerging, important class of gene regulators is short single-stranded RNA (micro-RNA, miRNA) that interferes post-transcriptionally with gene expression and thus may play a role in the circadian variation of gene expression. MiRNAs are promising biomarkers by virtue of their disease-specific tissue expression and because of their presence as stable entities in the circulation. However, no studies have addressed the putative circadian rhythmicity of circulating, cell-free miRNAs. This question is important both for using miRNAs as biological markers and for clues to miRNA function in the regulation of circadian gene expression. Here, we investigate 92 miRNAs in plasma samples from 24 young male, healthy volunteers repeatedly sampled 9 times during a 24-hour stay in a regulated environment. We demonstrate that a third (26/79) of the measurable plasma miRNAs (using RT-qPCR on a microfluidic system) exhibit a rhythmic behavior and are distributed in two main phase patterns. Some of these miRNAs weakly target known clock genes and many have strong targets in intracellular MAPK signaling pathways. These novel findings highlight the importance of considering bio-oscillations in miRNA biomarker studies and suggest the further study of a set of specific circulating miRNAs in the regulation and functioning of biological clocks.
Collapse
Affiliation(s)
- Niels H. H. Heegaard
- Department of Autoimmunology and Biomarkers, Statens Serum Institut, Copenhagen, Denmark
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
- * E-mail:
| | - Anting Liu Carlsen
- Department of Autoimmunology and Biomarkers, Statens Serum Institut, Copenhagen, Denmark
- Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Berit Lilje
- Department of Microbiology and Infection Control, Statens Serum Institut, Copenhagen, Denmark
| | - Kim Lee Ng
- Department of Microbiology and Infection Control, Statens Serum Institut, Copenhagen, Denmark
| | - Mette E. Rønne
- Department of Autoimmunology and Biomarkers, Statens Serum Institut, Copenhagen, Denmark
| | - Henrik L. Jørgensen
- Department of Clinical Biochemistry, Faculty of Health Science, Bispebjerg Hospital, University of Copenhagen, Denmark
| | - Henriette Sennels
- Department of Clinical Biochemistry, Faculty of Health Science, Rigshospitalet - Glostrup, University of Copenhagen, Copenhagen, Denmark
| | - Jan Fahrenkrug
- Department of Clinical Biochemistry, Faculty of Health Science, Bispebjerg Hospital, University of Copenhagen, Denmark
| |
Collapse
|
240
|
Tyanova S, Temu T, Sinitcyn P, Carlson A, Hein MY, Geiger T, Mann M, Cox J. The Perseus computational platform for comprehensive analysis of (prote)omics data. Nat Methods 2016; 13:731-40. [DOI: 10.1038/nmeth.3901] [Citation(s) in RCA: 4028] [Impact Index Per Article: 447.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 05/10/2016] [Indexed: 02/06/2023]
|
241
|
Abstract
Most animals alternate periods of feeding with periods of fasting often coinciding with sleep. Upon >24 hr of fasting, humans, rodents, and other mammals enter alternative metabolic phases, which rely less on glucose and more on ketone body-like carbon sources. Both intermittent and periodic fasting result in benefits ranging from the prevention to the enhanced treatment of diseases. Similarly, time-restricted feeding (TRF), in which food consumption is restricted to certain hours of the day, allows the daily fasting period to last >12 hr, thus imparting pleiotropic benefits. Understanding the mechanistic link between nutrients and the fasting benefits is leading to the identification of fasting-mimicking diets (FMDs) that achieve changes similar to those caused by fasting. Given the pleiotropic and sustained benefits of TRF and FMDs, both basic science and translational research are warranted to develop fasting-associated interventions into feasible, effective, and inexpensive treatments with the potential to improve healthspan.
Collapse
|
242
|
Mass spectrometry-based absolute quantification reveals rhythmic variation of mouse circadian clock proteins. Proc Natl Acad Sci U S A 2016; 113:E3461-7. [PMID: 27247408 DOI: 10.1073/pnas.1603799113] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Absolute values of protein expression levels in cells are crucial information for understanding cellular biological systems. Precise quantification of proteins can be achieved by liquid chromatography (LC)-mass spectrometry (MS) analysis of enzymatic digests of proteins in the presence of isotope-labeled internal standards. Thus, development of a simple and easy way for the preparation of internal standards is advantageous for the analyses of multiple target proteins, which will allow systems-level studies. Here we describe a method, termed MS-based Quantification By isotope-labeled Cell-free products (MS-QBiC), which provides the simple and high-throughput preparation of internal standards by using a reconstituted cell-free protein synthesis system, and thereby facilitates both multiplexed and sensitive quantification of absolute amounts of target proteins. This method was applied to a systems-level dynamic analysis of mammalian circadian clock proteins, which consist of transcription factors and protein kinases that govern central and peripheral circadian clocks in mammals. Sixteen proteins from 20 selected circadian clock proteins were successfully quantified from mouse liver over a 24-h time series, and 14 proteins had circadian variations. Quantified values were applied to detect internal body time using a previously developed molecular timetable method. The analyses showed that single time-point data from wild-type mice can predict the endogenous state of the circadian clock, whereas data from clock mutant mice are not applicable because of the disappearance of circadian variation.
Collapse
|
243
|
Subramanian P, Jayapalan JJ, Abdul-Rahman PS, Arumugam M, Hashim OH. Temporal regulation of proteome profile in the fruit fly, Drosophila melanogaster. PeerJ 2016; 4:e2080. [PMID: 27257555 PMCID: PMC4888302 DOI: 10.7717/peerj.2080] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 05/03/2016] [Indexed: 12/11/2022] Open
Abstract
Background. Diurnal rhythms of protein synthesis controlled by the biological clock underlie the rhythmic physiology in the fruit fly, Drosophila melanogaster. In this study, we conducted a proteome-wide investigation of rhythmic protein accumulation in D. melanogaster. Materials and Methods. Total protein collected from fly samples harvested at 4 h intervals over the 24 h period were subjected to two-dimensional gel electrophoresis, trypsin digestion and MS/MS analysis. Protein spots/clusters were identified with MASCOT search engine and Swiss-Prot database. Expression of proteins was documented as percentage of volume contribution using the Image Master 2D Platinum software. Results. A total of 124 protein spots/clusters were identified using MS/MS analysis. Significant variation in the expression of 88 proteins over the 24-h period was observed. A relatively higher number of proteins was upregulated during the night compared to the daytime. The complexity of temporal regulation of the D. melanogaster proteome was further reflected from functional annotations of the differently expressed proteins, with those that were upregulated at night being restricted to the heat shock proteins and proteins involved in metabolism, muscle activity, protein synthesis/folding/degradation and apoptosis, whilst those that were overexpressed in the daytime were apparently involved in metabolism, muscle activity, ion-channel/cellular transport, protein synthesis/folding/degradation, redox homeostasis, development and transcription. Conclusion. Our data suggests that a wide range of proteins synthesized by the fruit fly, D. melanogaster, is under the regulation of the biological clock.
Collapse
Affiliation(s)
- Perumal Subramanian
- Department of Biochemistry and Biotechnology, Annamalai University , Chidambaram, Tamil Nadu , India
| | - Jaime J Jayapalan
- University of Malaya Centre for Proteomics Research (UMCPR), Faculty of Medicine, University of Malaya , Kuala Lumpur , Malaysia
| | - Puteri S Abdul-Rahman
- University of Malaya Centre for Proteomics Research (UMCPR), Department of Molecular Medicine, Faculty of Medicine, University of Malaya , Kuala Lumpur , Malaysia
| | - Manjula Arumugam
- Department of Biochemistry and Biotechnology, Annamalai University , Chidambaram, Tamil Nadu , India
| | - Onn H Hashim
- University of Malaya Centre for Proteomics Research (UMCPR), Department of Molecular Medicine, Faculty of Medicine, University of Malaya , Kuala Lumpur , Malaysia
| |
Collapse
|
244
|
Hoehenwarter W, Mönchgesang S, Neumann S, Majovsky P, Abel S, Müller J. Comparative expression profiling reveals a role of the root apoplast in local phosphate response. BMC PLANT BIOLOGY 2016; 16:106. [PMID: 27121119 PMCID: PMC4849097 DOI: 10.1186/s12870-016-0790-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 04/18/2016] [Indexed: 05/03/2023]
Abstract
BACKGROUND Plant adaptation to limited phosphate availability comprises a wide range of responses to conserve and remobilize internal phosphate sources and to enhance phosphate acquisition. Vigorous restructuring of root system architecture provides a developmental strategy for topsoil exploration and phosphate scavenging. Changes in external phosphate availability are locally sensed at root tips and adjust root growth by modulating cell expansion and cell division. The functionally interacting Arabidopsis genes, LOW PHOSPHATE RESPONSE 1 and 2 (LPR1/LPR2) and PHOSPHATE DEFICIENCY RESPONSE 2 (PDR2), are key components of root phosphate sensing. We recently demonstrated that the LOW PHOSPHATE RESPONSE 1 - PHOSPHATE DEFICIENCY RESPONSE 2 (LPR1-PDR2) module mediates apoplastic deposition of ferric iron (Fe(3+)) in the growing root tip during phosphate limitation. Iron deposition coincides with sites of reactive oxygen species generation and triggers cell wall thickening and callose accumulation, which interfere with cell-to-cell communication and inhibit root growth. RESULTS We took advantage of the opposite phosphate-conditional root phenotype of the phosphate deficiency response 2 mutant (hypersensitive) and low phosphate response 1 and 2 double mutant (insensitive) to investigate the phosphate dependent regulation of gene and protein expression in roots using genome-wide transcriptome and proteome analysis. We observed an overrepresentation of genes and proteins that are involved in the regulation of iron homeostasis, cell wall remodeling and reactive oxygen species formation, and we highlight a number of candidate genes with a potential function in root adaptation to limited phosphate availability. Our experiments reveal that FERRIC REDUCTASE DEFECTIVE 3 mediated, apoplastic iron redistribution, but not intracellular iron uptake and iron storage, triggers phosphate-dependent root growth modulation. We further highlight expressional changes of several cell wall-modifying enzymes and provide evidence for adjustment of the pectin network at sites of iron accumulation in the root. CONCLUSION Our study reveals new aspects of the elaborate interplay between phosphate starvation responses and changes in iron homeostasis. The results emphasize the importance of apoplastic iron redistribution to mediate phosphate-dependent root growth adjustment and suggest an important role for citrate in phosphate-dependent apoplastic iron transport. We further demonstrate that root growth modulation correlates with an altered expression of cell wall modifying enzymes and changes in the pectin network of the phosphate-deprived root tip, supporting the hypothesis that pectins are involved in iron binding and/or phosphate mobilization.
Collapse
Affiliation(s)
- Wolfgang Hoehenwarter
- Proteome Analytics Research Group, Leibniz Institute of Plant Biochemistry, D-06120, Halle (Saale), Germany
| | - Susann Mönchgesang
- Department of Stress and Developmental Biology, Leibniz Institute of Plant Biochemistry, D-06120, Halle (Saale), Germany
| | - Steffen Neumann
- Department of Stress and Developmental Biology, Leibniz Institute of Plant Biochemistry, D-06120, Halle (Saale), Germany
| | - Petra Majovsky
- Proteome Analytics Research Group, Leibniz Institute of Plant Biochemistry, D-06120, Halle (Saale), Germany
| | - Steffen Abel
- Department of Molecular Signal Processing, Leibniz Institute of Plant Biochemistry, D-06120, Halle (Saale), Germany
- Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, D-06120, Halle (Saale), Germany
- Department of Plant Sciences, University of California-Davis, Davis, CA, 95616, USA
| | - Jens Müller
- Department of Molecular Signal Processing, Leibniz Institute of Plant Biochemistry, D-06120, Halle (Saale), Germany.
| |
Collapse
|
245
|
Dallmann R, Okyar A, Lévi F. Dosing-Time Makes the Poison: Circadian Regulation and Pharmacotherapy. Trends Mol Med 2016; 22:430-445. [PMID: 27066876 DOI: 10.1016/j.molmed.2016.03.004] [Citation(s) in RCA: 178] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 03/17/2016] [Indexed: 12/14/2022]
Abstract
Daily rhythms in physiology significantly modulate drug pharmacokinetics and pharmacodynamics according to the time-of-day, a finding that has led to the concept of chronopharmacology. The importance of biological clocks for xenobiotic metabolism has gained increased attention with the discovery of the molecular circadian clockwork. Mechanistic understanding of the cell-autonomous molecular circadian oscillator and the circadian timing system as a whole has opened new conceptual and methodological lines of investigation to understand first, the clock's impact on a specific drug's daily variations or the effects/side effects of environmental substances, and second, how clock-controlled pathways are coordinated within a given tissue or organism. Today, there is an increased understanding of the circadian modulation of drug effects. Moreover, several molecular strategies are being developed to treat disease-dependent and drug-induced clock disruptions in humans.
Collapse
Affiliation(s)
- Robert Dallmann
- Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK; Warwick Systems Biology Centre, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK.
| | - Alper Okyar
- Department of Pharmacology, Faculty of Pharmacy, Istanbul University, Beyazit-Istanbul, Turkey
| | - Francis Lévi
- Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK; Warwick Systems Biology Centre, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| |
Collapse
|
246
|
Circadian clock, nutrient quality, and eating pattern tune diurnal rhythms in the mitochondrial proteome. Proc Natl Acad Sci U S A 2016; 113:3127-9. [PMID: 26979954 DOI: 10.1073/pnas.1601786113] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
247
|
Johnston JD, Ordovás JM, Scheer FA, Turek FW. Circadian Rhythms, Metabolism, and Chrononutrition in Rodents and Humans. Adv Nutr 2016; 7:399-406. [PMID: 26980824 PMCID: PMC4785478 DOI: 10.3945/an.115.010777] [Citation(s) in RCA: 168] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Chrononutrition is an emerging discipline that builds on the intimate relation between endogenous circadian (24-h) rhythms and metabolism. Circadian regulation of metabolic function can be observed from the level of intracellular biochemistry to whole-organism physiology and even postprandial responses. Recent work has elucidated the metabolic roles of circadian clocks in key metabolic tissues, including liver, pancreas, white adipose, and skeletal muscle. For example, tissue-specific clock disruption in a single peripheral organ can cause obesity or disruption of whole-organism glucose homeostasis. This review explains mechanistic insights gained from transgenic animal studies and how these data are being translated into the study of human genetics and physiology. The principles of chrononutrition have already been demonstrated to improve human weight loss and are likely to benefit the health of individuals with metabolic disease, as well as of the general population.
Collapse
Affiliation(s)
| | - José M Ordovás
- Tufts University, Boston, MA;,Madrid Institutes of Advanced Studies-Food, Madrid, Spain
| | - Frank A Scheer
- Brigham and Women's Hospital, Boston, MA;,Harvard Medical School, Boston, MA; and
| | | |
Collapse
|
248
|
Reinke H, Asher G. Circadian Clock Control of Liver Metabolic Functions. Gastroenterology 2016; 150:574-80. [PMID: 26657326 DOI: 10.1053/j.gastro.2015.11.043] [Citation(s) in RCA: 212] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 11/02/2015] [Accepted: 11/03/2015] [Indexed: 02/08/2023]
Abstract
The circadian clock is an endogenous biological timekeeping system that synchronizes physiology and behavior to day/night cycles. A wide variety of processes throughout the entire gastrointestinal tract and notably the liver appear to be under circadian control. These include various metabolic functions such as nutrient uptake, processing, and detoxification, which align organ function to cycle with nutrient supply and demand. Remarkably, genetic or environmental disruption of the circadian clock can cause metabolic diseases or exacerbate pathological states. In addition, modern lifestyles force more and more people worldwide into asynchrony between the external time and their circadian clock, resulting in a constant state of social jetlag. Recent evidence indicates that interactions between altered energy metabolism and disruptions in the circadian clock create a downward spiral that can lead to diabetes and other metabolic diseases. In this review, we provide an overview of rhythmic processes in the liver and highlight the functions of circadian clock genes under physiological and pathological conditions; we focus on their roles in regulation of hepatic glucose as well as lipid and bile acid metabolism and detoxification and their potential effects on the development of fatty liver and nonalcoholic steatohepatitis.
Collapse
Affiliation(s)
- Hans Reinke
- Medical Faculty, Institute of Clinical Chemistry and Laboratory Diagnostics, University of Düsseldorf, Düsseldorf, Germany; IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany.
| | - Gad Asher
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
249
|
Abstract
Our understanding of the molecular underpinnings of the mammalian circadian oscillator and its tight connection to physiology has progressed tremendously during the past decades. The liver is considered the prototypic experimental model tissue for circadian research in peripheral organs. Studies on liver clocks have been highly productive and yielded information about widely different aspects of circadian biology. The liver, as one of the largest organs in the body, has often been used for the identification of core clock and auxiliary clock components, for example, by biochemical purifications. Because the liver is also a major metabolic hub, studies addressing the interplay between circadian clocks and metabolism have been insightful. In addition, the use of liver-specific loss-of-function models for clock components highlighted not only specific physiological roles of the hepatic clock but also its interplay with systemic cues and oscillators in other organs. Recently, technological advances in omics approaches have been successfully applied on the liver, providing a comprehensive depiction of pervasive circadian control of gene expression and protein and metabolite accumulation. In this review, we chose to illuminate specific examples that demonstrate how different experimental approaches--namely, biochemical, metabolic, genetic, and omics methodologies--have advanced our knowledge regarding circadian liver biology and chronobiology in general.
Collapse
Affiliation(s)
- Ziv Zwighaft
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Hans Reinke
- University of Düsseldorf, Medical Faculty, Institute of Clinical Chemistry and Laboratory Diagnostics, Düsseldorf, Germany IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Gad Asher
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
250
|
Zhou D, Wang Y, Chen L, Jia L, Yuan J, Sun M, Zhang W, Wang P, Zuo J, Xu Z, Luan J. Evolving roles of circadian rhythms in liver homeostasis and pathology. Oncotarget 2016; 7:8625-39. [PMID: 26843619 PMCID: PMC4890992 DOI: 10.18632/oncotarget.7065] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 01/18/2016] [Indexed: 02/06/2023] Open
Abstract
Circadian clock in mammals is determined by a core oscillator in the suprachiasmatic nucleus (SCN) of the hypothalamus and synchronized peripheral clocks in other tissues. The coherent timing systems could sustain robust output of circadian rhythms in response to the entrainment controlled environmentally. Disparate approaches have discovered that clock genes and clock-controlled genes (CCGs) exist in nearly all mammalian cell types and are essential for establishing the mechanisms and complexity of internal time-keeping systems. Accumulating evidence demonstrates that the control of homeostasis and pathology in the liver involves intricate loops of transcriptional and post-translational regulation of clock genes expression. This review will focus on the recent advances with great importance concerning clock rhythms linking liver homeostasis and diseases. We particularly highlight what is currently known of the evolving insights into the mechanisms underlying circadian clock . Eventually , findings during recent years in the field might prompt new circadian-related chronotherapeutic strategies for the diagnosis and treatment of liver diseases by coupling these processes.
Collapse
Affiliation(s)
- Dexi Zhou
- Laboratory of Clinical Pharmacy of Wannan Medical College, Wuhu, Anhui Province, China
- Department of Pharmacy in Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Yaqin Wang
- Laboratory of Clinical Pharmacy of Wannan Medical College, Wuhu, Anhui Province, China
- Department of Pharmacy in Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Lu Chen
- Laboratory of Clinical Pharmacy of Wannan Medical College, Wuhu, Anhui Province, China
- Department of Pharmacy in Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Leijuan Jia
- Laboratory of Clinical Pharmacy of Wannan Medical College, Wuhu, Anhui Province, China
- Department of Pharmacy in Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Jie Yuan
- Laboratory of Clinical Pharmacy of Wannan Medical College, Wuhu, Anhui Province, China
- Department of Pharmacy in Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Mei Sun
- Laboratory of Clinical Pharmacy of Wannan Medical College, Wuhu, Anhui Province, China
- Department of Pharmacy in Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Wen Zhang
- Laboratory of Clinical Pharmacy of Wannan Medical College, Wuhu, Anhui Province, China
- Department of Pharmacy in Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Peipei Wang
- Laboratory of Clinical Pharmacy of Wannan Medical College, Wuhu, Anhui Province, China
- Department of Pharmacy in Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Jian Zuo
- Laboratory of Clinical Pharmacy of Wannan Medical College, Wuhu, Anhui Province, China
- Department of Pharmacy in Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Zhenyu Xu
- Laboratory of Clinical Pharmacy of Wannan Medical College, Wuhu, Anhui Province, China
- Department of Pharmacy in Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Jiajie Luan
- Laboratory of Clinical Pharmacy of Wannan Medical College, Wuhu, Anhui Province, China
- Department of Pharmacy in Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| |
Collapse
|