1
|
Duan J, Karri SS, Forouzesh K, Mortimer T, Plikus MV, Benitah SA, Takahashi JS, Andersen B. Designing and Evaluating Circadian Experiments on Mouse Skin. J Invest Dermatol 2025; 145:484-493. [PMID: 39891645 DOI: 10.1016/j.jid.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/27/2024] [Accepted: 01/08/2025] [Indexed: 02/03/2025]
Abstract
All skin layers and cutaneous appendages harbor a robust circadian clock, whose phase is under the influence of light through the central clock in the suprachiasmatic nucleus. The skin clock coordinates fundamental biological processes, including metabolism and stem cell activation. It also prominently modulates activity of skin-resident immune cells and the inflammatory response. Numerous diurnally regulated genes in the skin have been implicated in skin diseases in GWASs. Therefore, the mouse skin is a powerful model for understanding the diverse roles of circadian biology in maintaining tissue health and the initiation and propagation of disease states. When planning experiments to study the circadian biology of mouse skin, multiple technical and biological factors must be carefully considered. In this paper, we provide comprehensive guidance on the general circadian experimental design and associated housing for the mice. We highlight the importance of aligning sample collection with the desired hair cycle stage and animal age. We introduce methods to disrupt the clock in the skin, including altering light and feeding schedules as well as using transgenic mouse models. Finally, we discuss the use of transcriptomic data, both bulk and single cell, for circadian studies.
Collapse
Affiliation(s)
- Junyan Duan
- Center for Complex Biological Systems, University of California, Irvine, Irvine, California, USA; The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, California, USA
| | - Satya Swaroop Karri
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, California, USA; Division of Endocrinology, Department of Medicine, School of Medicine, University of California, Irvine, Irvine, California, USA
| | - Kiarash Forouzesh
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, California, USA; Division of Endocrinology, Department of Medicine, School of Medicine, University of California, Irvine, Irvine, California, USA
| | - Thomas Mortimer
- Institute for Research in Biomedicine, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Maksim V Plikus
- Center for Complex Biological Systems, University of California, Irvine, Irvine, California, USA; The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, California, USA; Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California, USA
| | - Salvador Aznar Benitah
- Institute for Research in Biomedicine, The Barcelona Institute of Science and Technology, Barcelona, Spain; Catalan Institution for Research and Advanced Studies, Barcelona, Spain
| | - Joseph S Takahashi
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Bogi Andersen
- Center for Complex Biological Systems, University of California, Irvine, Irvine, California, USA; Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, California, USA; Division of Endocrinology, Department of Medicine, School of Medicine, University of California, Irvine, Irvine, California, USA.
| |
Collapse
|
2
|
Mihut A, O'Neill JS, Partch CL, Crosby P. PERspectives on circadian cell biology. Philos Trans R Soc Lond B Biol Sci 2025; 380:20230483. [PMID: 39842483 PMCID: PMC11753889 DOI: 10.1098/rstb.2023.0483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/18/2024] [Accepted: 05/24/2024] [Indexed: 01/24/2025] Open
Abstract
Daily rhythms in the activities of PERIOD proteins are critical to the temporal regulation of mammalian physiology. While the molecular partners and genetic circuits that allow PERIOD to effect auto-repression and regulate transcriptional programmes are increasingly well understood, comprehension of the time-resolved mechanisms that allow PERIOD to conduct this daily dance is incomplete. Here, we consider the character and controversies of this central mammalian clock protein with a focus on its intrinsically disordered nature.This article is part of the Theo Murphy meeting issue 'Circadian rhythms in infection and immunity'.
Collapse
Affiliation(s)
- Andrei Mihut
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, CambridgeCB2 0QH, UK
| | - John S. O'Neill
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, CambridgeCB2 0QH, UK
| | - Carrie L. Partch
- Department of Chemistry and Biochemistry, University of California, 1156 High Street, Santa Cruz, CA95064, USA
| | - Priya Crosby
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, EdinburghEH9 3BF, UK
| |
Collapse
|
3
|
Colita CI, Hermann DM, Filfan M, Colita D, Doepnner TR, Tica O, Glavan D, Popa-Wagner A. Optimizing Chronotherapy in Psychiatric Care: The Impact of Circadian Rhythms on Medication Timing and Efficacy. Clocks Sleep 2024; 6:635-655. [PMID: 39584972 PMCID: PMC11586979 DOI: 10.3390/clockssleep6040043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/15/2024] [Accepted: 10/31/2024] [Indexed: 11/26/2024] Open
Abstract
In many medical settings, medications are typically administered in the morning or evening, aligning with patients' daily routines. This practice does not stem from chronotherapy, which involves scheduling drug administration to enhance its effectiveness, but rather from the way clinical operations are structured. The timing of drug administration can significantly affect a medication's effectiveness and side effects, with the impact varying by up to ten times based on circadian rhythms. Disorders such as major depression, bipolar disorder, and schizophrenia are linked to disruptions in these rhythms. Recent studies have found that circadian dysfunctions, including genetic and neurohumoral changes, underlie many psychiatric conditions. Issues such as an altered glucocorticoid rhythm due to impaired HPA axis function, disturbed melatonin balance, and sleep disturbances have been noted in psychotic disorders. Furthermore, mood disorders have been associated with changes in the expression of circadian rhythm genes such as Clock, Bmal1, and Per. Considering that the absorption, biodistribution, effects on target organs, half-life, metabolism, and elimination of drugs are all influenced by the body's circadian rhythms, this narrative review explores the optimal timing of medication administration to maximize efficacy and minimize side effects in the treatment of psychiatric disorders. By closely monitoring circadian variations in cortisol, melatonin, and key clock genes, as well as by deepening our understanding of the metabolisms and pharmacokinetics of antipsychotic medications, we propose a chronotherapy approach for psychiatric patients that could significantly enhance patient care.
Collapse
Affiliation(s)
- Cezar-Ivan Colita
- Doctoral School, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (C.-I.C.); (D.C.)
| | - Dirk M. Hermann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany;
- Department of Psychiatry, University of Medicine and Pharmacy, 200349 Craiova, Romania;
| | - Madalina Filfan
- Department of Psychiatry, University of Medicine and Pharmacy, 200349 Craiova, Romania;
| | - Daniela Colita
- Doctoral School, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (C.-I.C.); (D.C.)
| | - Thorsten R. Doepnner
- Department of Neurology, University Medical Center, Klinikstraße 33, 35392 Gießen, Germany;
| | - Oana Tica
- Department of Pharmacology, University of Medicine and Pharmacy, 200349 Craiova, Romania;
| | - Daniela Glavan
- Department of Psychiatry, University of Medicine and Pharmacy, 200349 Craiova, Romania;
| | - Aurel Popa-Wagner
- Doctoral School, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (C.-I.C.); (D.C.)
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany;
| |
Collapse
|
4
|
Jiao H, Kalsbeek A, Yi CX. Microglia, circadian rhythm and lifestyle factors. Neuropharmacology 2024; 257:110029. [PMID: 38852838 DOI: 10.1016/j.neuropharm.2024.110029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/11/2024]
Abstract
Microglia, a vital homeostasis-keeper of the central nervous system, perform critical functions such as synaptic pruning, clearance of cellular debris, and participation in neuroinflammatory processes. Recent research has shown that microglia exhibit strong circadian rhythms that not only actively regulate their own immune activity, but also affect neuronal function. Disruptions of the circadian clock have been linked to a higher risk of developing a variety of diseases. In this article we will provide an overview of how lifestyle factors impact microglial function, with a focus on disruptions caused by irregular sleep-wake patterns, reduced physical activity, and eating at the wrong time-of-day. We will also discuss the potential connection between these lifestyle factors, disrupted circadian rhythms, and the role of microglia in keeping brain health. This article is part of the Special Issue on "Microglia".
Collapse
Affiliation(s)
- Han Jiao
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, location AMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam, the Netherlands; Department of Clinical Chemistry, Laboratory of Endocrinology, Amsterdam University Medical Center, location AMC, Amsterdam, the Netherlands; Netherlands Institute for Neuroscience, Amsterdam, the Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, location AMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam, the Netherlands; Department of Clinical Chemistry, Laboratory of Endocrinology, Amsterdam University Medical Center, location AMC, Amsterdam, the Netherlands; Netherlands Institute for Neuroscience, Amsterdam, the Netherlands
| | - Chun-Xia Yi
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, location AMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam, the Netherlands; Department of Clinical Chemistry, Laboratory of Endocrinology, Amsterdam University Medical Center, location AMC, Amsterdam, the Netherlands; Netherlands Institute for Neuroscience, Amsterdam, the Netherlands.
| |
Collapse
|
5
|
Ma C, Li H, Li W, Yang G, Chen L. Adaptive Differences in Cellular and Behavioral Responses to Circadian Disruption between C57BL/6 and BALB/c Strains. Int J Mol Sci 2024; 25:10404. [PMID: 39408733 PMCID: PMC11476807 DOI: 10.3390/ijms251910404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
The regulation of the mammalian circadian clock is largely dependent on heredity. In model animals for circadian rhythm studies, C57BL/6 and BALB/c mice exhibit considerable differences in their adaptation to circadian disruption, yet deeper comparisons remain unexplored. Here, we have established embryonic fibroblast cells derived from C57BL/6 mice (MEF) and BALB/c (BALB/3T3) mice, which have been transfected with the Bmal1 promoter-driven luciferase (Bmal1-Luc) reporter gene. Next, dexamethasone was applied for various cyclic stimulations, which revealed that Bmal1 bioluminescence of MEF cells was entrained to 24 to 26 h cycles, whereas BALB/3T3 cells have a wider range (22 to 28 h) with lower amplitudes. Behaviorally, BALB/c mice swiftly adapted to a 6-h advance light/dark cycle, unlike C57BL/6 mice. Furthermore, we found the expression of the circadian rhythm gene Npas2 in BALB/c mice is significantly lower than that in C57BL/6 mice. This observation is consistent with the differentially expressed genes (DEGs) in the intestine and lung tissues of C57BL/6 and BALB/c mice, based on the RNA-seq datasets downloaded from the Gene Expression Omnibus (GEO). In summary, our study uncovers that BALB/c mice possess greater resilience in circadian rhythm than C57BL/6 mice, both cellular and behaviorally, identifying potential genes underlying this difference.
Collapse
Affiliation(s)
- Changxiao Ma
- School of Bioengineering, Dalian University of Technology, Dalian 116024, China; (C.M.); (H.L.)
- Health Science Center, East China Normal University, Shanghai 200241, China;
| | - Haonan Li
- School of Bioengineering, Dalian University of Technology, Dalian 116024, China; (C.M.); (H.L.)
| | - Wenyu Li
- Health Science Center, East China Normal University, Shanghai 200241, China;
| | - Guangrui Yang
- School of Clinical Medicine, Shanghai University of Medicine & Health Sciences, Shanghai 201318, China
| | - Lihong Chen
- Health Science Center, East China Normal University, Shanghai 200241, China;
| |
Collapse
|
6
|
Sao K, Risbud MV. Sdc4 deletion perturbs intervertebral disc matrix homeostasis and promotes early osteopenia in the aging mouse spine. Matrix Biol 2024; 131:46-61. [PMID: 38806135 DOI: 10.1016/j.matbio.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/07/2024] [Accepted: 05/22/2024] [Indexed: 05/30/2024]
Abstract
Syndecan 4 (SDC4), a cell surface heparan sulfate proteoglycan, is known to regulate matrix catabolism by nucleus pulposus cells in an inflammatory milieu. However, the role of SDC4 in the aging spine has never been explored. Here we analyzed the spinal phenotype of Sdc4 global knockout (KO) mice as a function of age. Micro-computed tomography showed that Sdc4 deletion severely reduced vertebral trabecular and cortical bone mass, and biomechanical properties of vertebrae were significantly altered in Sdc4 KO mice. These changes in vertebral bone were likely due to elevated osteoclastic activity. The histological assessment showed subtle phenotypic changes in the intervertebral disc. Imaging-Fourier transform-infrared analyses showed a reduced relative ratio of mature collagen crosslinks in young adult nucleus pulposus (NP) and annulus fibrosus (AF) of KO compared to wildtype discs. Additionally, relative chondroitin sulfate levels increased in the NP compartment of the KO mice. Transcriptomic analysis of NP tissue using CompBio, an AI-based tool showed biological themes associated with prominent dysregulation of heparan sulfate GAG degradation, mitochondria metabolism, autophagy, endoplasmic reticulum (ER)-associated misfolded protein processes and ER to Golgi protein processing. Overall, this study highlights the important role of SDC4 in fine-tuning vertebral bone homeostasis and extracellular matrix homeostasis in the mouse intervertebral disc.
Collapse
Affiliation(s)
- Kimheak Sao
- Graduate Program in Cell Biology and Regenerative Medicine, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, United States; Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, 1025 Walnut Street, Suite 501 College Bldg., Philadelphia, PA 19107, United States
| | - Makarand V Risbud
- Graduate Program in Cell Biology and Regenerative Medicine, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, United States; Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, 1025 Walnut Street, Suite 501 College Bldg., Philadelphia, PA 19107, United States.
| |
Collapse
|
7
|
Xia H, Zhan Y, Wang L, Wang X. Exploring the interplay between circadian rhythms and prostate cancer: insights into androgen receptor signaling and therapeutic opportunities. Front Cell Dev Biol 2024; 12:1421204. [PMID: 39011396 PMCID: PMC11246886 DOI: 10.3389/fcell.2024.1421204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 06/11/2024] [Indexed: 07/17/2024] Open
Abstract
Circadian rhythm disruption is closely related to increased incidence of prostate cancer. Incorporating circadian rhythms into the study of prostate cancer pathogenesis can provide a more comprehensive understanding of the causes of cancer and offer new options for precise treatment. Therefore, this article comprehensively summarizes the epidemiology of prostate cancer, expounds the contradictory relationship between circadian rhythm disorders and prostate cancer risk, and elucidates the relationship between circadian rhythm regulators and the incidence of prostate cancer. Importantly, this article also focuses on the correlation between circadian rhythms and androgen receptor signaling pathways, as well as the applicability of time therapy in prostate cancer. This may prove significant in enhancing the clinical treatment of prostate cancer.
Collapse
Affiliation(s)
- Hongyan Xia
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Yang Zhan
- National Engineering Laboratory for AIDS Vaccine, College of Life Sciences, Jilin University, Changchun, China
| | - Li Wang
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| | - Xiaohui Wang
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
8
|
Sharma D, Partch CL. PAS Dimerization at the Nexus of the Mammalian Circadian Clock. J Mol Biol 2024; 436:168341. [PMID: 37924861 PMCID: PMC11729053 DOI: 10.1016/j.jmb.2023.168341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/23/2023] [Accepted: 10/29/2023] [Indexed: 11/06/2023]
Abstract
Circadian rhythms are genetically encoded molecular clocks for internal biological timekeeping. Organisms from single-cell bacteria to humans use these clocks to adapt to the external environment and synchronize their physiology and behavior to solar light/dark cycles. Although the proteins that constitute the molecular 'cogs' and give rise to circadian rhythms are now known, we still lack a detailed understanding of how these proteins interact to generate and sustain the ∼24-hour circadian clock. Structural studies have helped to expand the architecture of clock proteins and have revealed the abundance of the only well-defined structured regions in the mammalian clock called Per-ARNT-Sim (PAS) domains. PAS domains are modular, evolutionarily conserved sensory and signaling domains that typically mediate protein-protein interactions. In the mammalian circadian clock, PAS domains modulate homo and heterodimerization of several core clock proteins that assemble into transcription factors or repressors. This review will focus on the functional importance of the PAS domains in the circadian clock from a biophysical and biochemical standpoint and describe their roles in clock protein interactions and circadian timekeeping.
Collapse
Affiliation(s)
- Diksha Sharma
- Department of Chemistry and Biochemistry, University of California Santa Cruz, CA, United States
| | - Carrie L Partch
- Department of Chemistry and Biochemistry, University of California Santa Cruz, CA, United States; Center for Circadian Biology, University of California San Diego, CA, United States.
| |
Collapse
|
9
|
Hu Y, Li X, Zhang J, Liu D, Lu R, Li JD. A genome-wide CRISPR screen identifies USP1 as a novel regulator of the mammalian circadian clock. FEBS J 2024; 291:445-457. [PMID: 37909373 DOI: 10.1111/febs.16990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 09/07/2023] [Accepted: 10/27/2023] [Indexed: 11/03/2023]
Abstract
The circadian clock is generated by a molecular timekeeping mechanism coordinating daily oscillations of physiology and behaviors in mammals. In the mammalian circadian clockwork, basic helix-loop-helix ARNT-like protein 1 (BMAL1) is a core circadian component whose defects lead to circadian disruption and elicit behavioral arrhythmicity. To identify previously unknown regulators for circadian clocks, we searched for genes influencing BMAL1 protein level by using a CRISPR/Cas9-based genome-wide knockout library. As a result, we found that the deubiquitinase ubiquitin carboxyl-terminal hydrolase 1 (USP1) positively affects BMAL1 protein abundance. Overexpression of wild-type USP1, but not a deubiquitinase-inactive mutant USP1, upregulated BMAL1 protein level, whereas genetic ablation of USP1 downregulated BMAL1 protein level in U2OS cells. Furthermore, treatment with USP1 inhibitors led to significant downregulation of BMAL1 protein in U2OS cells as well as mouse tissues. Subsequently, genetic ablation or pharmacological inhibition of USP1 resulted in reduced mRNA levels of a panel of clock genes and disrupted circadian rhythms in U2OS cells. Mechanistically, USP1 was able to de-ubiquitinate BMAL1 and inhibit the proteasomal degradation of BMAL1. Interestingly, the expression of Usp1 was much higher than the other two deubiquitinases of BMAL1 (Usp2 and Usp9X) in the mouse heart, implying a tissue-specific function of USP1 in the regulation of BMAL1 stability. Our work thus identifies deubiquitinase USP1 as a previously unknown regulator of the mammalian circadian clock and highlights the potential of genome-wide CRISPR screens in the identification of regulators for the circadian clock.
Collapse
Affiliation(s)
- Ying Hu
- Furong Laboratory, Department of Anaesthesiology, Xiangya Hospital, Central South University, Changsha, China
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
- MOE Key Laboratory of Rare Pediatric Diseases, Changsha, China
| | - Xin Li
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
- MOE Key Laboratory of Rare Pediatric Diseases, Changsha, China
| | - Jing Zhang
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Dengfeng Liu
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Renbin Lu
- Furong Laboratory, Department of Anaesthesiology, Xiangya Hospital, Central South University, Changsha, China
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
- Department of Basic Medical Sciences, Changsha Medical University, Changsha, China
- National Clinical Research Center for Geratric Disorder, Xiangya Hospital, Central South University, Changsha, China
| | - Jia-Da Li
- Furong Laboratory, Department of Anaesthesiology, Xiangya Hospital, Central South University, Changsha, China
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
- MOE Key Laboratory of Rare Pediatric Diseases, Changsha, China
- Hunan Key Laboratory of Animal Models for Human Diseases, Changsha, China
- National Clinical Research Center for Geratric Disorder, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
10
|
Earnhardt-San AL, Baker EC, Riley DG, Ghaffari N, Long CR, Cardoso RC, Randel RD, Welsh TH. Differential Expression of Circadian Clock Genes in the Bovine Neuroendocrine Adrenal System. Genes (Basel) 2023; 14:2082. [PMID: 38003025 PMCID: PMC10670998 DOI: 10.3390/genes14112082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 11/08/2023] [Accepted: 11/11/2023] [Indexed: 11/26/2023] Open
Abstract
Knowledge of circadian rhythm clock gene expression outside the suprachiasmatic nucleus is increasing. The purpose of this study was to determine whether expression of circadian clock genes differed within or among the bovine stress axis tissues (e.g., amygdala, hypothalamus, pituitary, adrenal cortex, and adrenal medulla). Tissues were obtained at an abattoir from eight mature nonpregnant Brahman cows that had been maintained in the same pasture and nutritional conditions. Sample tissues were stored in RNase-free sterile cryovials at -80 °C until the total RNA was extracted, quantified, assessed, and sequenced (NovaSeq 6000 system; paired-end 150 bp cycles). The trimmed reads were then mapped to a Bos taurus (B. taurus) reference genome (Umd3.1). Further analysis used the edgeR package. Raw gene count tables were read into RStudio, and low-expression genes were filtered out using the criteria of three minimum reads per gene in at least five samples. Normalization factors were then calculated using the trimmed mean of M values method to produce normalized gene counts within each sample tissue. The normalized gene counts important for a circadian rhythm were analyzed within and between each tissue of the stress axis using the GLM and CORR procedures of the Statistical Analysis System (SAS). The relative expression profiles of circadian clock genes differed (p < 0.01) within each tissue, with neuronal PAS domain protein 2 (NPAS2) having greater expression in the amygdala (p < 0.01) and period circadian regulator (PER1) having greater expression in all other tissues (p < 0.01). The expression among tissues also differed (p < 0.01) for individual circadian clock genes, with circadian locomotor output cycles protein kaput (CLOCK) expression being greater within the adrenal tissues and nuclear receptor subfamily 1 group D member 1 (NR1D1) expression being greater within the other tissues (p < 0.01). Overall, the results indicate that within each tissue, the various circadian clock genes were differentially expressed, in addition to being differentially expressed among the stress tissues of mature Brahman cows. Future use of these findings may assist in improving livestock husbandry and welfare by understanding interactions of the environment, stress responsiveness, and peripheral circadian rhythms.
Collapse
Affiliation(s)
- Audrey L. Earnhardt-San
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (D.G.R.); (R.C.C.)
- Texas A&M AgriLife Research Center, Overton, TX 75684, USA; (C.R.L.); (R.D.R.)
| | - Emilie C. Baker
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (D.G.R.); (R.C.C.)
| | - David G. Riley
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (D.G.R.); (R.C.C.)
| | - Noushin Ghaffari
- Department of Computer Science, Prairie View A&M University, Prairie View, TX 77070, USA;
| | - Charles R. Long
- Texas A&M AgriLife Research Center, Overton, TX 75684, USA; (C.R.L.); (R.D.R.)
| | - Rodolfo C. Cardoso
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (D.G.R.); (R.C.C.)
| | - Ronald D. Randel
- Texas A&M AgriLife Research Center, Overton, TX 75684, USA; (C.R.L.); (R.D.R.)
| | - Thomas H. Welsh
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (D.G.R.); (R.C.C.)
| |
Collapse
|
11
|
Murgo E, Colangelo T, Bellet MM, Malatesta F, Mazzoccoli G. Role of the Circadian Gas-Responsive Hemeprotein NPAS2 in Physiology and Pathology. BIOLOGY 2023; 12:1354. [PMID: 37887064 PMCID: PMC10603908 DOI: 10.3390/biology12101354] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/14/2023] [Accepted: 10/20/2023] [Indexed: 10/28/2023]
Abstract
Neuronal PAS domain protein 2 (NPAS2) is a hemeprotein comprising a basic helix-loop-helix domain (bHLH) and two heme-binding sites, the PAS-A and PAS-B domains. This protein acts as a pyridine nucleotide-dependent and gas-responsive CO-dependent transcription factor and is encoded by a gene whose expression fluctuates with circadian rhythmicity. NPAS2 is a core cog of the molecular clockwork and plays a regulatory role on metabolic pathways, is important for the function of the central nervous system in mammals, and is involved in carcinogenesis as well as in normal biological functions and processes, such as cardiovascular function and wound healing. We reviewed the scientific literature addressing the various facets of NPAS2 and framing this gene/protein in several and very different research and clinical fields.
Collapse
Affiliation(s)
- Emanuele Murgo
- Department of Medical Sciences, Division of Internal Medicine and Chronobiology Laboratory, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy;
| | - Tommaso Colangelo
- Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto 1, 71100 Foggia, Italy;
- Cancer Cell Signaling Unit, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy
| | - Maria Marina Bellet
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, 06132 Perugia, Italy;
| | - Francesco Malatesta
- Department of Biochemical Sciences “Alessandro Rossi Fanelli”, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Gianluigi Mazzoccoli
- Department of Medical Sciences, Division of Internal Medicine and Chronobiology Laboratory, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy;
| |
Collapse
|
12
|
Wei H, Adelsheim Z, Fischer R, McCarthy MJ. Serum from Myalgic encephalomyelitis/chronic fatigue syndrome patients causes loss of coherence in cellular circadian rhythms. J Neuroimmunol 2023; 381:578142. [PMID: 37393850 PMCID: PMC10527922 DOI: 10.1016/j.jneuroim.2023.578142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/12/2023] [Accepted: 06/23/2023] [Indexed: 07/04/2023]
Abstract
Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is a disabling disorder characterized by disrupted daily patterns of activity, sleep, and physiology. Past studies in ME/CFS patients have examined circadian rhythms, suggested that desynchronization between central and peripheral rhythms may be an important pathological feature, and identified associated changes in post-inflammatory cytokines such as transforming growth factor beta (TGFB). However, no previous studies have examined circadian rhythms in ME/CFS using cellular models or studied the role of cytokines on circadian rhythms. In this study, we used serum samples previously collected from ME/CFS patients (n = 20) selected for the presence of insomnia symptoms and matched controls (n = 20) to determine the effects of serum factors and TGFB on circadian rhythms in NIH3T3 mouse immortalized fibroblasts stably transfected with the Per2-luc bioluminescent circadian reporter. Compared to control serum, ME/CFS serum caused a significant loss of rhythm robustness (decreased goodness of fit) and nominally increased the rate of damping of cellular rhythms. Damping rate was associated with insomnia severity in ME/CFS patients using the Pittsburgh Sleep Quality Index (PSQI). Recombinant TGFB1 peptide applied to cells reduced rhythm amplitude, caused phase delay and decreased robustness of rhythms. However, there was no difference in TGFB1 levels between ME/CFS and control serum indicating the effects of serum on cellular rhythms cannot be explained by levels of this cytokine. Future studies will be required to identify additional serum factors in ME/CFS patients that alter circadian rhythms in cells.
Collapse
Affiliation(s)
- Heather Wei
- VA San Diego Healthcare System, San Diego,CA, USA
| | - Zoe Adelsheim
- Department of Psychiatry and Center for Circadian Biology, University of California San Diego, La Jolla, CA, USA
| | - Rita Fischer
- Department of Psychiatry and Center for Circadian Biology, University of California San Diego, La Jolla, CA, USA
| | - Michael J McCarthy
- VA San Diego Healthcare System, San Diego,CA, USA; Department of Psychiatry and Center for Circadian Biology, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
13
|
Marri D, Filipovic D, Kana O, Tischkau S, Bhattacharya S. Prediction of mammalian tissue-specific CLOCK-BMAL1 binding to E-box DNA motifs. Sci Rep 2023; 13:7742. [PMID: 37173345 PMCID: PMC10182026 DOI: 10.1038/s41598-023-34115-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
The Brain and Muscle ARNTL-Like 1 protein (BMAL1) forms a heterodimer with either Circadian Locomotor Output Cycles Kaput (CLOCK) or Neuronal PAS domain protein 2 (NPAS2) to act as a master regulator of the mammalian circadian clock gene network. The dimer binds to E-box gene regulatory elements on DNA, activating downstream transcription of clock genes. Identification of transcription factor binding sites and genomic features that correlate to DNA binding by BMAL1 is a challenging problem, given that CLOCK-BMAL1 or NPAS2-BMAL1 bind to several distinct binding motifs (CANNTG) on DNA. Using three different types of tissue-specific machine learning models with features based on (1) DNA sequence, (2) DNA sequence plus DNA shape, and (3) DNA sequence and shape plus histone modifications, we developed an interpretable predictive model of genome-wide BMAL1 binding to E-box motifs and dissected the mechanisms underlying BMAL1-DNA binding. Our results indicated that histone modifications, the local shape of the DNA, and the flanking sequence of the E-box motif are sufficient predictive features for BMAL1-DNA binding. Our models also provide mechanistic insights into tissue specificity of DNA binding by BMAL1.
Collapse
Affiliation(s)
- Daniel Marri
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - David Filipovic
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
- Department of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Omar Kana
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
| | - Shelley Tischkau
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Sudin Bhattacharya
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA.
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA.
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA.
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
14
|
Wollmuth EM, Angert ER. Microbial circadian clocks: host-microbe interplay in diel cycles. BMC Microbiol 2023; 23:124. [PMID: 37161348 PMCID: PMC10173096 DOI: 10.1186/s12866-023-02839-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 03/28/2023] [Indexed: 05/11/2023] Open
Abstract
BACKGROUND Circadian rhythms, observed across all domains of life, enable organisms to anticipate and prepare for diel changes in environmental conditions. In bacteria, a circadian clock mechanism has only been characterized in cyanobacteria to date. These clocks regulate cyclical patterns of gene expression and metabolism which contribute to the success of cyanobacteria in their natural environments. The potential impact of self-generated circadian rhythms in other bacterial and microbial populations has motivated extensive research to identify novel circadian clocks. MAIN TEXT Daily oscillations in microbial community composition and function have been observed in ocean ecosystems and in symbioses. These oscillations are influenced by abiotic factors such as light and the availability of nutrients. In the ocean ecosystems and in some marine symbioses, oscillations are largely controlled by light-dark cycles. In gut systems, the influx of nutrients after host feeding drastically alters the composition and function of the gut microbiota. Conversely, the gut microbiota can influence the host circadian rhythm by a variety of mechanisms including through interacting with the host immune system. The intricate and complex relationship between the microbiota and their host makes it challenging to disentangle host behaviors from bacterial circadian rhythms and clock mechanisms that might govern the daily oscillations observed in these microbial populations. CONCLUSIONS While the ability to anticipate the cyclical behaviors of their host would likely be enhanced by a self-sustained circadian rhythm, more evidence and further studies are needed to confirm whether host-associated heterotrophic bacteria possess such systems. In addition, the mechanisms by which heterotrophic bacteria might respond to diel cycles in environmental conditions has yet to be uncovered.
Collapse
Affiliation(s)
- Emily M Wollmuth
- Department of Microbiology, Cornell University, 123 Wing Drive, Ithaca, NY, 14853, USA
| | - Esther R Angert
- Department of Microbiology, Cornell University, 123 Wing Drive, Ithaca, NY, 14853, USA.
| |
Collapse
|
15
|
Brécier A, Li VW, Smith CS, Halievski K, Ghasemlou N. Circadian rhythms and glial cells of the central nervous system. Biol Rev Camb Philos Soc 2023; 98:520-539. [PMID: 36352529 DOI: 10.1111/brv.12917] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 10/17/2022] [Accepted: 10/25/2022] [Indexed: 11/12/2022]
Abstract
Glial cells are the most abundant cells in the central nervous system and play crucial roles in neural development, homeostasis, immunity, and conductivity. Over the past few decades, glial cell activity in mammals has been linked to circadian rhythms, the 24-h chronobiological clocks that regulate many physiological processes. Indeed, glial cells rhythmically express clock genes that cell-autonomously regulate glial function. In addition, recent findings in rodents have revealed that disruption of the glial molecular clock could impact the entire organism. In this review, we discuss the impact of circadian rhythms on the function of the three major glial cell types - astrocytes, microglia, and oligodendrocytes - across different locations within the central nervous system. We also review recent evidence uncovering the impact of glial cells on the body's circadian rhythm. Together, this sheds new light on the involvement of glial clock machinery in various diseases.
Collapse
Affiliation(s)
- Aurélie Brécier
- Pain Chronobiology & Neuroimmunology Laboratory, Queen's University, Botterell Hall, room 754, Kingston, ON, K7L 3N6, Canada
- Department of Biomedical & Molecular Sciences, 18 Stuart Street, Kingston, ON, K7L 3N6, Canada
| | - Vina W Li
- Pain Chronobiology & Neuroimmunology Laboratory, Queen's University, Botterell Hall, room 754, Kingston, ON, K7L 3N6, Canada
- Department of Biomedical & Molecular Sciences, 18 Stuart Street, Kingston, ON, K7L 3N6, Canada
| | - Chloé S Smith
- Pain Chronobiology & Neuroimmunology Laboratory, Queen's University, Botterell Hall, room 754, Kingston, ON, K7L 3N6, Canada
- Department of Biomedical & Molecular Sciences, 18 Stuart Street, Kingston, ON, K7L 3N6, Canada
| | - Katherine Halievski
- Pain Chronobiology & Neuroimmunology Laboratory, Queen's University, Botterell Hall, room 754, Kingston, ON, K7L 3N6, Canada
| | - Nader Ghasemlou
- Pain Chronobiology & Neuroimmunology Laboratory, Queen's University, Botterell Hall, room 754, Kingston, ON, K7L 3N6, Canada
- Department of Biomedical & Molecular Sciences, 18 Stuart Street, Kingston, ON, K7L 3N6, Canada
- Department of Anesthesiology & Perioperative Medicine, 76 Stuart Street, Kingston, ON, K7L 2V7, Canada
- Centre for Neuroscience Studies, Queen's University, 18 Stuart Street, Kingston, ON, K7L 3N6, Canada
| |
Collapse
|
16
|
Kim JY, Kim W, Lee KH. The role of microRNAs in the molecular link between circadian rhythm and autism spectrum disorder. Anim Cells Syst (Seoul) 2023; 27:38-52. [PMID: 36860270 PMCID: PMC9970207 DOI: 10.1080/19768354.2023.2180535] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023] Open
Abstract
Circadian rhythm regulates physiological cycles of awareness and sleepiness. Melatonin production is primarily regulated by circadian regulation of gene expression and is involved in sleep homeostasis. If the circadian rhythm is abnormal, sleep disorders, such as insomnia and several other diseases, can occur. The term 'autism spectrum disorder (ASD)' is used to characterize people who exhibit a certain set of repetitive behaviors, severely constrained interests, social deficits, and/or sensory behaviors that start very early in life. Because many patients with ASD suffer from sleep disorders, sleep disorders and melatonin dysregulation are attracting attention for their potential roles in ASD. ASD is caused by abnormalities during the neurodevelopmental processes owing to various genetic or environmental factors. Recently, the role of microRNAs (miRNAs) in circadian rhythm and ASD have gained attraction. We hypothesized that the relationship between circadian rhythm and ASD could be explained by miRNAs that can regulate or be regulated by either or both. In this study, we introduced a possible molecular link between circadian rhythm and ASD. We performed a thorough literature review to understand their complexity.
Collapse
Affiliation(s)
- Ji Young Kim
- Department of Molecular Biology, Pusan National University, Busan, Republic of Korea
| | - Wanil Kim
- Department of Biochemistry, College of Medicine, Gyeongsang National University, Jinju-si, Republic of Korea, Wanil Kim Department of Biochemistry, College of Medicine, Gyeongsang National University, Jinju-si, Gyeongsangnam-do52727, Republic of Korea; Kyung-Ha Lee Department of Molecular Biology, Pusan National University, 2, Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan46241, Republic of Korea
| | - Kyung-Ha Lee
- Department of Molecular Biology, Pusan National University, Busan, Republic of Korea, Wanil Kim Department of Biochemistry, College of Medicine, Gyeongsang National University, Jinju-si, Gyeongsangnam-do52727, Republic of Korea; Kyung-Ha Lee Department of Molecular Biology, Pusan National University, 2, Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan46241, Republic of Korea
| |
Collapse
|
17
|
Abstract
Driven by autonomous molecular clocks that are synchronized by a master pacemaker in the suprachiasmatic nucleus, cardiac physiology fluctuates in diurnal rhythms that can be partly or entirely circadian. Cardiac contractility, metabolism, and electrophysiology, all have diurnal rhythms, as does the neurohumoral control of cardiac and kidney function. In this review, we discuss the evidence that circadian biology regulates cardiac function, how molecular clocks may relate to the pathogenesis of heart failure, and how chronotherapeutics might be applied in heart failure. Disrupting molecular clocks can lead to heart failure in animal models, and the myocardial response to injury seems to be conditioned by the time of day. Human studies are consistent with these findings, and they implicate the clock and circadian rhythms in the pathogenesis of heart failure. Certain circadian rhythms are maintained in patients with heart failure, a factor that can guide optimal timing of therapy. Pharmacologic and nonpharmacologic manipulation of circadian rhythms and molecular clocks show promise in the prevention and treatment of heart failure.
Collapse
Affiliation(s)
- Nadim El Jamal
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ronan Lordan
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sarah L. Teegarden
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Tilo Grosser
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Translational Pharmacology, Bielefeld University, Bielefeld, Germany
| | - Garret FitzGerald
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
18
|
Giri A, Rahman I, Sundar IK. Circadian clock-based therapeutics in chronic pulmonary diseases. Trends Pharmacol Sci 2022; 43:1014-1029. [PMID: 36302705 PMCID: PMC9756397 DOI: 10.1016/j.tips.2022.09.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/16/2022] [Accepted: 09/20/2022] [Indexed: 11/11/2022]
Abstract
The circadian clock is the biochemical oscillator that orchestrates the observable circadian rhythms in physiology and behavior. Disruption of the circadian clock in the lungs during chronic pulmonary diseases is considered one of the key etiological risk factors that drive pathobiology. Preclinical studies support that pharmacological manipulation of the circadian clock is a conceivable approach for the development of novel clock-based therapeutics. Despite recent advances, no effort has been undertaken to integrate novel findings for the treatment and management of chronic lung diseases. We, therefore, recognize the need to discuss the candidate clock genes that can be potentially targeted for therapeutic intervention. Here, we aim to create the first roadmap that will advance the development of circadian- clock-based therapeutics that may provide better outcomes in treating chronic pulmonary diseases.
Collapse
Affiliation(s)
- Allan Giri
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Irfan Rahman
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester Medical Center, Rochester, New York, USA
| | - Isaac Kirubakaran Sundar
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA.
| |
Collapse
|
19
|
Du X, Cui Z, Ning Z, Deng X, Amevor FK, Shu G, Wang X, Zhang Z, Tian Y, Zhu Q, Wang Y, Li D, Zhang Y, Zhao X. Circadian miR-218-5p targets gene CA2 to regulate uterine carbonic anhydrase activity during egg shell calcification. Poult Sci 2022; 101:102158. [PMID: 36167021 PMCID: PMC9513254 DOI: 10.1016/j.psj.2022.102158] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 06/29/2022] [Accepted: 08/24/2022] [Indexed: 11/06/2022] Open
Abstract
MicroRNAs (miRNAs) are involved in regulating the circadian clock. In our previous work, miR-218-5p was found to be a circadian miRNA in the chicken uterus, but its role in the eggshell formation process was not clear. In the present study, we found that the expression levels of miR-218-5p and two 2 predicted target genes carbonic anhydrase 2 (CA2) and neuronal PAS domain protein 2 (NPAS2) were oscillated in the chicken uterus. The results of dual-luciferase reporter gene assays in the present study demonstrated that miR-218-5p directly targeted the 3' untranslated regions of CA2 and NPAS2. miR-218-5p showed an opposite expression profile to CA2 within a 24 h cycle in the chicken uterus. Moreover, over-expression of miR-218-5p reduced the mRNA and protein expression of CA2, while miR-218-5p knockdown increased CA2 mRNA and protein expression. Overexpression of CA2 also significantly increased the activity of carbonic anhydrase Ⅱ (P < 0.05), whereas knockdown of CA2 decreased the activity of carbonic anhydrase Ⅱ. miR-218-5p influenced carbonic anhydrase activity via regulating the expression of CA2. These results demonstrated that clock-controlled miR-218-5p regulates carbonic anhydrase activity in the chicken uterus by targeting CA2 during eggshell formation.
Collapse
Affiliation(s)
- Xiaxia Du
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan Province, P. R. China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agricultural and Rural Affairs, College of Animal and Technology (Institute of Animal Genetics and Breeding), Sichuan Agricultural University, P. R., Chengdu, China
| | - Zhifu Cui
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan Province, P. R. China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agricultural and Rural Affairs, College of Animal and Technology (Institute of Animal Genetics and Breeding), Sichuan Agricultural University, P. R., Chengdu, China
| | - Zifan Ning
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan Province, P. R. China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agricultural and Rural Affairs, College of Animal and Technology (Institute of Animal Genetics and Breeding), Sichuan Agricultural University, P. R., Chengdu, China
| | - Xun Deng
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan Province, P. R. China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agricultural and Rural Affairs, College of Animal and Technology (Institute of Animal Genetics and Breeding), Sichuan Agricultural University, P. R., Chengdu, China
| | - Felix Kwame Amevor
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan Province, P. R. China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agricultural and Rural Affairs, College of Animal and Technology (Institute of Animal Genetics and Breeding), Sichuan Agricultural University, P. R., Chengdu, China
| | - Gang Shu
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan Province, P. R. China
| | - Xiaoqi Wang
- Agriculture and Animal Husbandry Comprehensive Service Center, Tibet Autonomous Region, P. R. China
| | - Zhichao Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan Province, P. R. China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agricultural and Rural Affairs, College of Animal and Technology (Institute of Animal Genetics and Breeding), Sichuan Agricultural University, P. R., Chengdu, China
| | - Yaofu Tian
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan Province, P. R. China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agricultural and Rural Affairs, College of Animal and Technology (Institute of Animal Genetics and Breeding), Sichuan Agricultural University, P. R., Chengdu, China
| | - Qing Zhu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan Province, P. R. China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agricultural and Rural Affairs, College of Animal and Technology (Institute of Animal Genetics and Breeding), Sichuan Agricultural University, P. R., Chengdu, China
| | - Yan Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan Province, P. R. China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agricultural and Rural Affairs, College of Animal and Technology (Institute of Animal Genetics and Breeding), Sichuan Agricultural University, P. R., Chengdu, China
| | - Diyan Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan Province, P. R. China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agricultural and Rural Affairs, College of Animal and Technology (Institute of Animal Genetics and Breeding), Sichuan Agricultural University, P. R., Chengdu, China
| | - Yao Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan Province, P. R. China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agricultural and Rural Affairs, College of Animal and Technology (Institute of Animal Genetics and Breeding), Sichuan Agricultural University, P. R., Chengdu, China
| | - Xiaoling Zhao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan Province, P. R. China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agricultural and Rural Affairs, College of Animal and Technology (Institute of Animal Genetics and Breeding), Sichuan Agricultural University, P. R., Chengdu, China.
| |
Collapse
|
20
|
He Y, Cen H, Guo L, Zhang T, Yang Y, Dong D, Wu B. Circadian Oscillator NPAS2 Regulates Diurnal Expression and Activity of CYP1A2 in Mouse Liver. Biochem Pharmacol 2022; 206:115345. [DOI: 10.1016/j.bcp.2022.115345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 11/14/2022]
|
21
|
Smies CW, Bodinayake KK, Kwapis JL. Time to learn: The role of the molecular circadian clock in learning and memory. Neurobiol Learn Mem 2022; 193:107651. [PMID: 35697314 PMCID: PMC9903177 DOI: 10.1016/j.nlm.2022.107651] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/18/2022] [Accepted: 06/07/2022] [Indexed: 12/27/2022]
Abstract
The circadian system plays an important role in aligning biological processes with the external time of day. A range of physiological functions are governed by the circadian cycle, including memory processes, yet little is understood about how the clock interfaces with memory at a molecular level. The molecular circadian clock consists of four key genes/gene families, Period, Clock, Cryptochrome, and Bmal1, that rhythmically cycle in an ongoing transcription-translation negative feedback loop that maintains an approximately 24-hour cycle within cells of the brain and body. In addition to their roles in generating the circadian rhythm within the brain's master pacemaker (the suprachiasmatic nucleus), recent research has suggested that these clock genes may function locally within memory-relevant brain regions to modulate memory across the day/night cycle. This review will discuss how these clock genes function both within the brain's central clock and within memory-relevant brain regions to exert circadian control over memory processes. For each core clock gene, we describe the current research that demonstrates a potential role in memory and outline how these clock genes might interface with cascades known to support long-term memory formation. Together, the research suggests that clock genes function locally within satellite clocks across the brain to exert circadian control over long-term memory formation and possibly other biological processes. Understanding how clock genes might interface with local molecular cascades in the hippocampus and other brain regions is a critical step toward developing treatments for the myriad disorders marked by dysfunction of both the circadian system and cognitive processes.
Collapse
Affiliation(s)
- Chad W Smies
- Department of Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Kasuni K Bodinayake
- Department of Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Janine L Kwapis
- Department of Biology, Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|
22
|
Puig S, Shelton MA, Barko K, Seney ML, Logan RW. Sex-specific role of the circadian transcription factor NPAS2 in opioid tolerance, withdrawal and analgesia. GENES, BRAIN, AND BEHAVIOR 2022; 21:e12829. [PMID: 36053258 PMCID: PMC9744556 DOI: 10.1111/gbb.12829] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 08/09/2022] [Accepted: 08/09/2022] [Indexed: 02/05/2023]
Abstract
Opioids like fentanyl remain the mainstay treatment for chronic pain. Unfortunately, opioid's high dependence liability has led to the current opioid crisis, in part, because of side-effects that develop during long-term use, including analgesic tolerance and physical dependence. Both tolerance and dependence to opioids may lead to escalation of required doses to achieve previous therapeutic efficacy. Additionally, altered sleep and circadian rhythms are common in people on opioid therapy. Opioids impact sleep and circadian rhythms, while disruptions to sleep and circadian rhythms likely mediate the effects of opioids. However, the mechanisms underlying these bidirectional relationships between circadian rhythms and opioids remain largely unknown. The circadian protein, neuronal PAS domain protein 2 (NPAS2), regulates circadian-dependent gene transcription in structure of the central nervous system that modulate opioids and pain. Here, male and female wild-type and NPAS2-deficient (NPAS2-/-) mice were used to investigate the role of NPAS2 in fentanyl analgesia, tolerance, hyperalgesia and physical dependence. Overall, thermal pain thresholds, acute analgesia and tolerance to a fixed dose of fentanyl were largely similar between wild-type and NPAS2-/- mice. However, female NPAS2-/- exhibited augmented analgesic tolerance and significantly more behavioral symptoms of physical dependence to fentanyl. Only male NPAS2-/- mice had increased fentanyl-induced hypersensitivity, when compared with wild-type males. Together, our findings suggest sex-specific effects of NPAS2 signaling in the regulation of fentanyl-induced tolerance, hyperalgesia and dependence.
Collapse
Affiliation(s)
- Stephanie Puig
- Department of Pharmacology and Experimental TherapeuticsBoston University School of MedicineBostonMassachusettsUSA
- Translational Neuroscience Program, Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Micah A. Shelton
- Translational Neuroscience Program, Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Kelly Barko
- Translational Neuroscience Program, Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Marianne L. Seney
- Translational Neuroscience Program, Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Ryan W. Logan
- Department of Pharmacology and Experimental TherapeuticsBoston University School of MedicineBostonMassachusettsUSA
- Translational Neuroscience Program, Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Center for Systems NeuroscienceBoston UniversityBostonMassachusettsUSA
| |
Collapse
|
23
|
Becker-Krail DD, Walker WH, Nelson RJ. The Ventral Tegmental Area and Nucleus Accumbens as Circadian Oscillators: Implications for Drug Abuse and Substance Use Disorders. Front Physiol 2022; 13:886704. [PMID: 35574492 PMCID: PMC9094703 DOI: 10.3389/fphys.2022.886704] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/04/2022] [Indexed: 12/15/2022] Open
Abstract
Circadian rhythms convergently evolved to allow for optimal synchronization of individuals’ physiological and behavioral processes with the Earth’s 24-h periodic cycling of environmental light and temperature. Whereas the suprachiasmatic nucleus (SCN) is considered the primary pacemaker of the mammalian circadian system, many extra-SCN oscillatory brain regions have been identified to not only exhibit sustainable rhythms in circadian molecular clock function, but also rhythms in overall region activity/function and mediated behaviors. In this review, we present the most recent evidence for the ventral tegmental area (VTA) and nucleus accumbens (NAc) to serve as extra-SCN oscillators and highlight studies that illustrate the functional significance of the VTA’s and NAc’s inherent circadian properties as they relate to reward-processing, drug abuse, and vulnerability to develop substance use disorders (SUDs).
Collapse
Affiliation(s)
- Darius D Becker-Krail
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | - William H Walker
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | - Randy J Nelson
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
24
|
Murray A, Tharmalingam S, Khurana S, Lalonde C, Nguyen P, Tai TC. Effect of Prenatal Glucocorticoid Exposure on Circadian Rhythm Gene Expression in the Brains of Adult Rat Offspring. Cells 2022; 11:cells11101613. [PMID: 35626652 PMCID: PMC9139626 DOI: 10.3390/cells11101613] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/03/2022] [Accepted: 05/10/2022] [Indexed: 11/16/2022] Open
Abstract
Circadian clocks control many vital aspects of physiology from the sleep-wake cycle to metabolism. The circadian clock operates through transcriptional-translational feedback loops. The normal circadian signaling relies on a ‘master clock’, located in the suprachiasmatic nucleus (SCN), which synchronizes peripheral oscillators. Glucocorticoid receptor (GR) signaling has the ability to reset the phase of peripheral clocks. It has been shown that maternal exposure to glucocorticoids (GCs) can lead to modification of hypothalamic-pituitary-adrenal (HPA) function, impact stress-related behaviors, and result in a hypertensive state via GR activation. We previously demonstrated altered circadian rhythm signaling in the adrenal glands of offspring exposed to the synthetic GC, dexamethasone (Dex). Results from the current study show that prenatal exposure to Dex affects circadian rhythm gene expression in a brain region-specific and a sex-specific manner within molecular oscillators of the amygdala, hippocampus, paraventricular nucleus, and prefrontal cortex, as well as the main oscillator in the SCN. Results also show that spontaneously hypertensive rats (SHR) exhibited dysregulated circadian rhythm gene expression in these same brain regions compared with normotensive Wistar-Kyoto rats (WKY), although the pattern of dysregulation was markedly different from that seen in adult offspring prenatally exposed to GCs.
Collapse
Affiliation(s)
- Alyssa Murray
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada; (A.M.); (S.T.); (S.K.); (C.L.)
- Department of Biology, Laurentian University, Sudbury, ON P3E 2C6, Canada;
| | - Sujeenthar Tharmalingam
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada; (A.M.); (S.T.); (S.K.); (C.L.)
- Department of Biology, Laurentian University, Sudbury, ON P3E 2C6, Canada;
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, ON P3E 2C6, Canada
- Biomolecular Sciences Program, Laurentian University, Sudbury, ON P3E 2C6, Canada
| | - Sandhya Khurana
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada; (A.M.); (S.T.); (S.K.); (C.L.)
| | - Christine Lalonde
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada; (A.M.); (S.T.); (S.K.); (C.L.)
- Biomolecular Sciences Program, Laurentian University, Sudbury, ON P3E 2C6, Canada
| | - Phong Nguyen
- Department of Biology, Laurentian University, Sudbury, ON P3E 2C6, Canada;
| | - T. C. Tai
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada; (A.M.); (S.T.); (S.K.); (C.L.)
- Department of Biology, Laurentian University, Sudbury, ON P3E 2C6, Canada;
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, ON P3E 2C6, Canada
- Biomolecular Sciences Program, Laurentian University, Sudbury, ON P3E 2C6, Canada
- Correspondence: ; Tel.: +1-705-662-7239
| |
Collapse
|
25
|
Egstrand S, Mace ML, Morevati M, Nordholm A, Engelholm LH, Thomsen JS, Brüel A, Naveh-Many T, Guo Y, Olgaard K, Lewin E. Hypomorphic expression of parathyroid Bmal1 disrupts the internal parathyroid circadian clock and increases parathyroid cell proliferation in response to uremia. Kidney Int 2022; 101:1232-1250. [PMID: 35276205 DOI: 10.1016/j.kint.2022.02.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 01/24/2022] [Accepted: 02/08/2022] [Indexed: 01/22/2023]
Abstract
The molecular circadian clock is an evolutionary adaptation to anticipate recurring changes in the environment and to coordinate variations in activity, metabolism and hormone secretion. Parathyroid hyperplasia in uremia is a significant clinical challenge. Here, we examined changes in the transcriptome of the murine parathyroid gland over 24 hours and found a rhythmic expression of parathyroid signature genes, such as Casr, Vdr, Fgfr1 and Gcm2. Overall, 1455 genes corresponding to 6.9% of all expressed genes had significant circadian rhythmicity. Biological pathway analysis indicated that the circadian clock system is essential for the regulation of parathyroid cell function. To study this, a novel mouse strain with parathyroid gland-specific knockdown of the core clock gene Bmal1 (PTHcre;Bmal1flox/flox) was created. Dampening of the parathyroid circadian clock rhythmicity was found in these knockdown mice, resulting in abrogated rhythmicity of regulators of parathyroid cell proliferation such as Sp1, Mafb, Gcm2 and Gata3, indicating circadian clock regulation of these genes. Furthermore, the knockdown resulted in downregulation of genes involved in mitochondrial function and synthesis of ATP. When superimposed by uremia, these PTHcre;Bmal1flox/flox mice had an increased parathyroid cell proliferative response, compared to wild type mice. Thus, our findings indicate a role of the internal parathyroid circadian clock in the development of parathyroid gland hyperplasia in uremia.
Collapse
Affiliation(s)
- Søren Egstrand
- Nephrological Department B, Herlev Hospital, University of Copenhagen, Denmark; Nephrological Department P, Rigshospitalet, University of Copenhagen, Denmark
| | - Maria Lerche Mace
- Nephrological Department P, Rigshospitalet, University of Copenhagen, Denmark
| | - Marya Morevati
- Nephrological Department P, Rigshospitalet, University of Copenhagen, Denmark
| | - Anders Nordholm
- Nephrological Department B, Herlev Hospital, University of Copenhagen, Denmark; Nephrological Department P, Rigshospitalet, University of Copenhagen, Denmark
| | - Lars Henning Engelholm
- Biotech Research and Innovation Centre, University of Copenhagen, Denmark; Finsen Laboratory, University of Copenhagen, Denmark
| | | | - Annemarie Brüel
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Tally Naveh-Many
- Minerva Center for Calcium and Bone Metabolism, Nephrology Services, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Yuliu Guo
- Department of Genomic Medicine, Rigshospitalet, Centre of Diagnostics, Copenhagen, Denmark
| | - Klaus Olgaard
- Nephrological Department P, Rigshospitalet, University of Copenhagen, Denmark
| | - Ewa Lewin
- Nephrological Department B, Herlev Hospital, University of Copenhagen, Denmark; Nephrological Department P, Rigshospitalet, University of Copenhagen, Denmark.
| |
Collapse
|
26
|
Becker-Krail DD, Parekh PK, Ketchesin KD, Yamaguchi S, Yoshino J, Hildebrand MA, Dunham B, Ganapathiraiu MK, Logan RW, McClung CA. Circadian transcription factor NPAS2 and the NAD + -dependent deacetylase SIRT1 interact in the mouse nucleus accumbens and regulate reward. Eur J Neurosci 2022; 55:675-693. [PMID: 35001440 PMCID: PMC9355311 DOI: 10.1111/ejn.15596] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 12/14/2021] [Accepted: 01/06/2022] [Indexed: 02/03/2023]
Abstract
Substance use disorders are associated with disruptions to both circadian rhythms and cellular metabolic state. At the molecular level, the circadian molecular clock and cellular metabolic state may be interconnected through interactions with the nicotinamide adenine dinucleotide (NAD+ )-dependent deacetylase, sirtuin 1 (SIRT1). In the nucleus accumbens (NAc), a region important for reward, both SIRT1 and the circadian transcription factor neuronal PAS domain protein 2 (NPAS2) are highly enriched, and both are regulated by the metabolic cofactor NAD+ . Substances of abuse, like cocaine, greatly disrupt cellular metabolism and promote oxidative stress; however, their effects on NAD+ in the brain remain unclear. Interestingly, cocaine also induces NAc expression of both NPAS2 and SIRT1, and both have independently been shown to regulate cocaine reward in mice. However, whether NPAS2 and SIRT1 interact in the NAc and/or whether together they regulate reward is unknown. Here, we demonstrate diurnal expression of Npas2, Sirt1 and NAD+ in the NAc, which is altered by cocaine-induced upregulation. Additionally, co-immunoprecipitation reveals NPAS2 and SIRT1 interact in the NAc, and cross-analysis of NPAS2 and SIRT1 chromatin immunoprecipitation sequencing reveals several reward-relevant and metabolic-related pathways enriched among shared gene targets. Notably, NAc-specific Npas2 knock-down or a functional Npas2 mutation in mice attenuates SIRT1-mediated increases in cocaine preference. Together, our data reveal an interaction between NPAS2 and SIRT1 in the NAc, which may serve to integrate cocaine's effects on circadian and metabolic factors, leading to regulation of drug reward.
Collapse
Affiliation(s)
- Darius D. Becker-Krail
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA,Center for Neuroscience, University of Pittsburgh, PA, USA
| | - Puja K. Parekh
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA,Center for Neuroscience, University of Pittsburgh, PA, USA
| | - Kyle D. Ketchesin
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA,Center for Neuroscience, University of Pittsburgh, PA, USA
| | - Shintaro Yamaguchi
- Center for Human Nutrition, Division of Geriatrics and Nutritional Science, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Jun Yoshino
- Center for Human Nutrition, Division of Geriatrics and Nutritional Science, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Mariah A. Hildebrand
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA,Center for Neuroscience, University of Pittsburgh, PA, USA
| | - Brandon Dunham
- Department of Biomedical Informatics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Madhavi K. Ganapathiraiu
- Department of Biomedical Informatics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ryan W. Logan
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Colleen A. McClung
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA,Center for Neuroscience, University of Pittsburgh, PA, USA,Correspondence: Colleen A. McClung,
| |
Collapse
|
27
|
Cui Z, Zhang Z, Amevor FK, Du X, Li L, Tian Y, Kang X, Shu G, Zhu Q, Wang Y, Li D, Zhang Y, Zhao X. Circadian miR-449c-5p regulates uterine Ca 2+ transport during eggshell calcification in chickens. BMC Genomics 2021; 22:764. [PMID: 34702171 PMCID: PMC8547053 DOI: 10.1186/s12864-021-08074-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 10/06/2021] [Indexed: 12/27/2022] Open
Abstract
Background miRNAs regulate circadian patterns by modulating the biological clocks of animals. In our previous study, we found that the clock gene exhibited a cosine expression pattern in the fallopian tube of chicken uterus. Clock-controlled miRNAs are present in mammals and Drosophila; however, whether there are clock-controlled miRNAs in the chicken uterus and, if so, how they regulate egg-laying rhythms is unclear. In this study, we selected 18 layer hens with similar ovipositional rhythmicity (each of three birds were sacrificed for study per 4 h throughout 24 h); their transcriptomes were scanned to identify the circadian miRNAs and to explore regulatory mechanisms within the uterus of chickens. Results We identified six circadian miRNAs that are mainly associated with several biological processes including ion trans-membrane transportation, response to calcium ion, and enrichment of calcium signaling pathways. Verification of the experimental results revealed that miR-449c-5p exhibited a cosine expression pattern in the chicken uterus. Ca2+-transporting ATPase 4 (ATP2B4) in the plasma membrane is the predicted target gene of circadian miR-449c-5p and is highly enriched in the calcium signaling pathway. We speculated that clock-controlled miR-449c-5p regulated Ca2+ transportation during eggshell calcification in the chicken uterus by targeting ATP2B4. ATP2B4 mRNA and protein were rhythmically expressed in the chicken uterus, and dual-luciferase reporter gene assays confirmed that ATP2B4 was directly targeted by miR-449c-5p. The expression of miR-449c-5p showed an opposite trend to that of ATP2B4 within a 24 h cycle in the chicken uterus; it inhibited mRNA and protein expression of ATP2B4 in the uterine tubular gland cells. In addition, overexpression of ATP2B4 significantly decreased intracellular Ca2+ concentration (P < 0.05), while knockdown of ATP2B4 accelerated intracellular Ca2+ concentrations. We found similar results after ATP2B4 knockdown by miR-449c-5p. Taken together, these results indicate that ATP2B4 promotes uterine Ca2+ trans-epithelial transport. Conclusions Clock-controlled miR-449c-5p regulates Ca2+ transport in the chicken uterus by targeting ATP2B4 during eggshell calcification. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-08074-3.
Collapse
Affiliation(s)
- Zhifu Cui
- Department of Animal Science, Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Apt 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan Province, People's Republic of China
| | - Zhichao Zhang
- Department of Animal Science, Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Apt 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan Province, People's Republic of China
| | - Felix Kwame Amevor
- Department of Animal Science, Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Apt 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan Province, People's Republic of China
| | - Xiaxia Du
- Department of Animal Science, Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Apt 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan Province, People's Republic of China
| | - Liang Li
- Department of Animal Science, Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Apt 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan Province, People's Republic of China
| | - Yaofu Tian
- Department of Animal Science, Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Apt 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan Province, People's Republic of China
| | - Xincheng Kang
- Department of Animal Science, Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Apt 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan Province, People's Republic of China
| | - Gang Shu
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan Province, People's Republic of China
| | - Qing Zhu
- Department of Animal Science, Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Apt 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan Province, People's Republic of China
| | - Yan Wang
- Department of Animal Science, Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Apt 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan Province, People's Republic of China
| | - Diyan Li
- Department of Animal Science, Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Apt 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan Province, People's Republic of China
| | - Yao Zhang
- Department of Animal Science, Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Apt 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan Province, People's Republic of China
| | - Xiaoling Zhao
- Department of Animal Science, Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Apt 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan Province, People's Republic of China.
| |
Collapse
|
28
|
Peng LU, Bai G, Pang Y. Roles of NPAS2 in circadian rhythm and disease. Acta Biochim Biophys Sin (Shanghai) 2021; 53:1257-1265. [PMID: 34415290 DOI: 10.1093/abbs/gmab105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Indexed: 11/14/2022] Open
Abstract
NPAS2, a circadian rhythm gene encoding the neuronal PAS domain protein 2 (NPAS2), has received widespread attention because of its complex functions in cells and diverse roles in disease progression, especially tumorigenesis. NPAS2 binds with DNA at E-box sequences and forms heterodimers with another circadian protein, brain and muscle ARNT-like protein 1 (BMAL1). Nucleotide variations of the NPAS2 gene have been shown to influence the overall survival and risk of death of cancer patients, and differential expression of NPAS2 has been linked to patient outcomes in breast cancer, lung cancer, non-Hodgkin's lymphoma, and other diseases. Here, we review the latest advances in our understanding of NPAS2 with the aim of drawing attention to its potential clinical applications and prospects.
Collapse
Affiliation(s)
- L u Peng
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Gaigai Bai
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Yingxin Pang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan 250012, China
| |
Collapse
|
29
|
Quintela T, Furtado A, Duarte AC, Gonçalves I, Myung J, Santos CRA. The role of circadian rhythm in choroid plexus functions. Prog Neurobiol 2021; 205:102129. [PMID: 34343629 DOI: 10.1016/j.pneurobio.2021.102129] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 07/22/2021] [Accepted: 07/28/2021] [Indexed: 12/17/2022]
Abstract
For several years, a great effort has been devoted to understand how circadian oscillations in physiological processes are determined by the circadian clock system. This system is composed by the master clock at the suprachiasmatic nucleus which sets the pace and tunes peripheral clocks in several organs. It was recently demonstrated that the choroid plexus epithelial cells that compose the blood-cerebrospinal fluid barrier hold a circadian clock which might control their multiple functions with implications for the maintenance of brain homeostasis. However, the choroid plexus activities regulated by its inner clock are still largely unknown. In this review, we propose that several choroid plexus functions might be regulated by the circadian clock, alike in other tissues. We provide evidences that the timing of cerebrospinal fluid secretion, clearance of amyloid-beta peptides and xenobiotics, and the barrier function of the blood-cerebrospinal fluid barrier are regulated by the circadian clock. These data, highlight that the circadian regulation of the blood-cerebrospinal fluid barrier must be taken into consideration for enhancing drug delivery to central nervous system disorders.
Collapse
Affiliation(s)
- Telma Quintela
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal.
| | - André Furtado
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Ana C Duarte
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Isabel Gonçalves
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Jihwan Myung
- Graduate Institute of Mind, Brain, and Consciousness, Taipei Medical University, No. 172-1 Sec. 2 Keelung Road, Da'an District, Taipei 106, Taiwan; Brain and Consciousness Research Centre, Shuang Ho Hospital, Ministry of Health and Welfare, No. 291 Zhongzheng Road, Zhonghe District, New Taipei City 235, Taiwan
| | - Cecília R A Santos
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| |
Collapse
|
30
|
Regulation of sinus node pacemaking and atrioventricular node conduction by HCN channels in health and disease. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021; 166:61-85. [PMID: 34197836 DOI: 10.1016/j.pbiomolbio.2021.06.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 06/02/2021] [Accepted: 06/14/2021] [Indexed: 12/19/2022]
Abstract
The funny current, If, was first recorded in the heart 40 or more years ago by Dario DiFrancesco and others. Since then, we have learnt that If plays an important role in pacemaking in the sinus node, the innate pacemaker of the heart, and more recently evidence has accumulated to show that If may play an important role in action potential conduction through the atrioventricular (AV) node. Evidence has also accumulated to show that regulation of the transcription and translation of the underlying Hcn genes plays an important role in the regulation of sinus node pacemaking and AV node conduction under normal physiological conditions - in athletes, during the circadian rhythm, in pregnancy, and during postnatal development - as well as pathological states - ageing, heart failure, pulmonary hypertension, diabetes and atrial fibrillation. There may be yet more pathological conditions involving changes in the expression of the Hcn genes. Here, we review the role of If and the underlying HCN channels in physiological and pathological changes of the sinus and AV nodes and we begin to explore the signalling pathways (microRNAs, transcription factors, GIRK4, the autonomic nervous system and inflammation) involved in this regulation. This review is dedicated to Dario DiFrancesco on his retirement.
Collapse
|
31
|
Putker M, Wong DCS, Seinkmane E, Rzechorzek NM, Zeng A, Hoyle NP, Chesham JE, Edwards MD, Feeney KA, Fischer R, Peschel N, Chen K, Vanden Oever M, Edgar RS, Selby CP, Sancar A, O’Neill JS. CRYPTOCHROMES confer robustness, not rhythmicity, to circadian timekeeping. EMBO J 2021; 40:e106745. [PMID: 33491228 PMCID: PMC8013833 DOI: 10.15252/embj.2020106745] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/08/2020] [Accepted: 12/18/2020] [Indexed: 12/22/2022] Open
Abstract
Circadian rhythms are a pervasive property of mammalian cells, tissues and behaviour, ensuring physiological adaptation to solar time. Models of cellular timekeeping revolve around transcriptional feedback repression, whereby CLOCK and BMAL1 activate the expression of PERIOD (PER) and CRYPTOCHROME (CRY), which in turn repress CLOCK/BMAL1 activity. CRY proteins are therefore considered essential components of the cellular clock mechanism, supported by behavioural arrhythmicity of CRY-deficient (CKO) mice under constant conditions. Challenging this interpretation, we find locomotor rhythms in adult CKO mice under specific environmental conditions and circadian rhythms in cellular PER2 levels when CRY is absent. CRY-less oscillations are variable in their expression and have shorter periods than wild-type controls. Importantly, we find classic circadian hallmarks such as temperature compensation and period determination by CK1δ/ε activity to be maintained. In the absence of CRY-mediated feedback repression and rhythmic Per2 transcription, PER2 protein rhythms are sustained for several cycles, accompanied by circadian variation in protein stability. We suggest that, whereas circadian transcriptional feedback imparts robustness and functionality onto biological clocks, the core timekeeping mechanism is post-translational.
Collapse
Affiliation(s)
| | | | | | | | - Aiwei Zeng
- MRC Laboratory of Molecular BiologyCambridgeUK
| | | | | | - Mathew D Edwards
- MRC Laboratory of Molecular BiologyCambridgeUK
- Present address:
UCL Sainsbury Wellcome Centre for Neural Circuits and BehaviourLondonUK
| | | | | | | | - Ko‐Fan Chen
- Institute of NeurologyUniversity College LondonLondonUK
- Present address:
Department of Genetics and Genome BiologyUniversity of LeicesterLeicesterUK
| | | | | | - Christopher P Selby
- Department of Biochemistry and BiophysicsUniversity of North Carolina School of MedicineChapel HillNCUSA
| | - Aziz Sancar
- Department of Biochemistry and BiophysicsUniversity of North Carolina School of MedicineChapel HillNCUSA
| | | |
Collapse
|
32
|
Abo SMC, Layton AT. Modeling the circadian regulation of the immune system: Sexually dimorphic effects of shift work. PLoS Comput Biol 2021; 17:e1008514. [PMID: 33788832 PMCID: PMC8041207 DOI: 10.1371/journal.pcbi.1008514] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 04/12/2021] [Accepted: 03/06/2021] [Indexed: 12/31/2022] Open
Abstract
The circadian clock exerts significance influence on the immune system and disruption of circadian rhythms has been linked to inflammatory pathologies. Shift workers often experience circadian misalignment as their irregular work schedules disrupt the natural light-dark cycle, which in turn can cause serious health problems associated with alterations in genetic expressions of clock genes. In particular, shift work is associated with impairment in immune function, and those alterations are sex-specific. The goal of this study is to better understand the mechanisms that explain the weakened immune system in shift workers. To achieve that goal, we have constructed a mathematical model of the mammalian pulmonary circadian clock coupled to an acute inflammation model in the male and female rats. Shift work was simulated by an 8h-phase advance of the circadian system with sex-specific modulation of clock genes. The model reproduces the clock gene expression in the lung and the immune response to various doses of lipopolysaccharide (LPS). Under normal conditions, our model predicts that a host is more sensitive to LPS at circadian time (CT) CT12 versus CT0 due to a dynamic change of Interleukin 10 (IL-10), an anti-inflammatory cytokine. We identify REV-ERB as a key modulator of IL-10 activity throughout the circadian day. The model also predicts a reversal of the times of lowest and highest sensitivity to LPS, with males and females exhibiting an exaggerated response to LPS at CT0, which is countered by a blunted immune response at CT12. Overall, females produce fewer pro-inflammatory cytokines than males, but the extent of sequelae experienced by males and females varies across the circadian day. This model can serve as an essential component in an integrative model that will yield mechanistic understanding of how shift work-mediated circadian disruptions affect the inflammatory and other physiological responses.
Collapse
Affiliation(s)
- Stéphanie M. C. Abo
- Department of Applied Mathematics, University of Waterloo, Waterloo, Ontario, Canada
| | - Anita T. Layton
- Department of Applied Mathematics, University of Waterloo, Waterloo, Ontario, Canada
- Department of Biology, Cheriton School of Computer Science, and School of Pharmacology, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
33
|
Kim SM, Vadnie CA, Philip VM, Gagnon LH, Chowdari KV, Chesler EJ, McClung CA, Logan RW. High-throughput measurement of fibroblast rhythms reveals genetic heritability of circadian phenotypes in diversity outbred mice and their founder strains. Sci Rep 2021; 11:2573. [PMID: 33510298 PMCID: PMC7843998 DOI: 10.1038/s41598-021-82069-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 01/14/2021] [Indexed: 01/21/2023] Open
Abstract
Circadian variability is driven by genetics and Diversity Outbred (DO) mice is a powerful tool for examining the genetics of complex traits because their high genetic and phenotypic diversity compared to conventional mouse crosses. The DO population combines the genetic diversity of eight founder strains including five common inbred and three wild-derived strains. In DO mice and their founders, we established a high-throughput system to measure cellular rhythms using in vitro preparations of skin fibroblasts. Among the founders, we observed strong heritability for rhythm period, robustness, phase and amplitude. We also found significant sex and strain differences for these rhythms. Extreme differences in period for molecular and behavioral rhythms were found between the inbred A/J strain and the wild-derived CAST/EiJ strain, where A/J had the longest period and CAST/EiJ had the shortest. In addition, we measured cellular rhythms in 329 DO mice, which displayed far greater phenotypic variability than the founders—80% of founders compared to only 25% of DO mice had periods of ~ 24 h. Collectively, our findings demonstrate that genetic diversity contributes to phenotypic variability in circadian rhythms, and high-throughput characterization of fibroblast rhythms in DO mice is a tractable system for examining the genetics of circadian traits.
Collapse
Affiliation(s)
- Sam-Moon Kim
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, 450 Technology Drive, Pittsburgh, PA, 15219, USA.,Center for Systems Neurogenetics of Addiction, The Jackson Laboratory, 600 Main Street, Bar Harbor, 04609, ME, USA
| | - Chelsea A Vadnie
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, 450 Technology Drive, Pittsburgh, PA, 15219, USA
| | - Vivek M Philip
- Center for Systems Neurogenetics of Addiction, The Jackson Laboratory, 600 Main Street, Bar Harbor, 04609, ME, USA
| | - Leona H Gagnon
- Center for Systems Neurogenetics of Addiction, The Jackson Laboratory, 600 Main Street, Bar Harbor, 04609, ME, USA
| | - Kodavali V Chowdari
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, 450 Technology Drive, Pittsburgh, PA, 15219, USA
| | - Elissa J Chesler
- Center for Systems Neurogenetics of Addiction, The Jackson Laboratory, 600 Main Street, Bar Harbor, 04609, ME, USA
| | - Colleen A McClung
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, 450 Technology Drive, Pittsburgh, PA, 15219, USA. .,Center for Systems Neurogenetics of Addiction, The Jackson Laboratory, 600 Main Street, Bar Harbor, 04609, ME, USA.
| | - Ryan W Logan
- Center for Systems Neurogenetics of Addiction, The Jackson Laboratory, 600 Main Street, Bar Harbor, 04609, ME, USA. .,Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, 700 Albany Street, Boston, 02118, MA, USA.
| |
Collapse
|
34
|
Clock knockdown attenuated reactive oxygen species-mediated senescence of chondrocytes through restoring autophagic flux. Life Sci 2021; 269:119036. [PMID: 33450259 DOI: 10.1016/j.lfs.2021.119036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/03/2021] [Accepted: 01/04/2021] [Indexed: 11/20/2022]
Abstract
AIMS Articular cartilage degeneration has been recognized as the primary pathological change in osteoarthritis (OA). Mechanisms that govern the shift from cartilage homeostasis to OA remain unknown. Previous studies have reported that intrinsic circadian clock in chondrocytes could function to optimize cartilage repair/remodeling to optimum times of day, but little is known about its molecular mechanisms. This study attempted to investigate the potential role and mechanism of circadian gene Clock in OA pathology. MATERIALS AND METHODS The expression of Clock in OA chondrocytes and cartilage was detected by qRT-PCR, western blot and immunohistochemistry. Temporal gene expression changes were analyzed using qRT-PCR in chondrocytes transfected with siClock following dexamethasone synchronization. In addition, the effect of Clock knockdown on senescent phenotypes and autophagic flux was evaluated in chondrocytes treated with siClock or siCntrl. KEY FINDINGS The expression of Clock was up-regulated in OA cartilage from humans and mouse models. Clock knockdown had no influence on rhythmic expression of the downstream genes in primary chondrocytes. We also found that Clock knockdown elevated antioxidant enzyme activities, diminished reactive oxygen species (ROS) production and attenuated senescence of chondrocytes via restoring autophagic flux. SIGNIFICANCE Clock knockdown can attenuate ROS-mediated senescence of chondrocytes through restoring autophagic flux in non-circadian manner, providing a potential therapeutic target for OA.
Collapse
|
35
|
Negri M, Pivonello C, Simeoli C, Di Gennaro G, Venneri MA, Sciarra F, Ferrigno R, de Angelis C, Sbardella E, De Martino MC, Colao A, Isidori AM, Pivonello R. Cortisol Circadian Rhythm and Insulin Resistance in Muscle: Effect of Dosing and Timing of Hydrocortisone Exposure on Insulin Sensitivity in Synchronized Muscle Cells. Neuroendocrinology 2021; 111:1005-1028. [PMID: 33130679 DOI: 10.1159/000512685] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/29/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION/AIM Circadian clock disruption is emerging as a risk factor for metabolic disorders, and particularly, alterations in clock genes circadian expression have been shown to influence insulin sensitivity. Recently, the reciprocal interplay between the circadian clock machinery and hypothal-amus-pituitary-adrenal axis has been largely demonstrated: the circadian clock may control the physiological circadian endogenous glucocorticoid (GC) secretion and action; GCs, in turn, are potent regulators of the circadian clock and their inappropriate replacement has been associated with metabolic impairment. The aim of the current study was to investigate in vitro the interaction between the timing-of-the-day exposure to different hydrocortisone (HC) concentrations and muscle insulin sensitivity. METHODS Serum-shock synchronized mouse skeletal muscle C2C12 cells were exposed to different HC concentrations resembling the circulating daily physiological cortisol profile (standard cortisol profile) and the circulating daily cortisol profile that reached in adrenal insufficient (AI) patients treated with once-daily modified-release HC (flat cortisol profile) and treated with thrice-daily conventional immediate-release HC (steep cortisol profile). The 24 h spontaneous oscillation of the clock genes in synchronized C2C12 cells was used to align the timing for in vitro HC exposure (Bmal1 acrophase, midphase, and bathyphase) with the reference times of cortisol peaks in AI patients treated with IR-HC (8 a.m., 1 p.m., and 6 p.m.). A panel of 84 insulin sensitivity-related genes and intracellular insulin signaling proteins were analyzed by RT-qPCR and Western blot, respectively. RESULTS The steep profile, characterized by a higher HC exposure during Bmal1bathyphase, produced significant downregulation in 21 insulin sensitivity-related genes including Insr, Irs1, Irs2, Pi3kca, and Adipor2, compared to the flat and standard profile. Reduced intracellular IRS1 Tyr608, AKT Ser473, AMPK Thr172, and ACC Ser79 phosphorylations were also observed. CONCLUSIONS The current study demonstrated that late-in-the-day cortisol exposure modulates insulin sensitivity-related gene expression and intracellular insulin signaling in skeletal muscle cells.
Collapse
Affiliation(s)
- Mariarosaria Negri
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy
| | - Claudia Pivonello
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy
| | - Chiara Simeoli
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy
| | - Gilda Di Gennaro
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy
| | - Mary Anna Venneri
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Francesca Sciarra
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Rosario Ferrigno
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy
| | - Cristina de Angelis
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy
| | - Emilia Sbardella
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Maria Cristina De Martino
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy
| | - Annamaria Colao
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy
- UNESCO Chair for Health Education and Sustainable Development, Federico II University, Naples, Italy
| | - Andrea M Isidori
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Rosario Pivonello
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy
- UNESCO Chair for Health Education and Sustainable Development, Federico II University, Naples, Italy
| |
Collapse
|
36
|
Cox KH, Takahashi JS. Introduction to the Clock System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1344:3-20. [PMID: 34773223 DOI: 10.1007/978-3-030-81147-1_1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Circadian (24-h) rhythms dictate almost everything we do, setting our clocks for specific times of sleeping and eating, as well as optimal times for many other basic functions. The physiological systems that coordinate circadian rhythms are intricate, but at their core, they all can be distilled down to cell-autonomous rhythms that are then synchronized within and among tissues. At first glance, these cell-autonomous rhythms may seem rather straight-forward, but years of research in the field has shown that they are strikingly complex, responding to many different external signals, often with remarkable tissue-specificity. To understand the cellular clock system, it is important to be familiar with the major players, which consist of pairs of proteins in a triad of transcriptional/translational feedback loops. In this chapter, we will go through each of the core protein pairs one-by-one, summarizing the literature as to their regulation and their broader impacts on circadian gene expression. We will conclude by briefly examining the human genetics literature, as well as providing perspectives on the future of the study of the molecular clock.
Collapse
Affiliation(s)
- Kimberly H Cox
- Department of Neuroscience and Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joseph S Takahashi
- Department of Neuroscience and Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA. .,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
37
|
Wang F, Xie N, Wu Y, Zhang Q, Zhu Y, Dai M, Zhou J, Pan J, Tang M, Cheng Q, Shi B, Guo Q, Li X, Xie L, Wang B, Yang D, Weng Q, Guo L, Ye J, Pan M, Zhang S, Zhou H, Zhen C, Liu P, Ning K, Brackenridge L, Hardiman PJ, Qu F. Association between circadian rhythm disruption and polycystic ovary syndrome. Fertil Steril 2020; 115:771-781. [PMID: 33358334 DOI: 10.1016/j.fertnstert.2020.08.1425] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 08/27/2020] [Accepted: 08/28/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To explore the association of circadian rhythm disruption with polycystic ovary syndrome (PCOS) and the potential underlying mechanism in ovarian granulosa cells (GCs). DESIGN Multicenter questionnaire-based survey, in vivo and ex vivo studies. SETTING Twelve hospitals in China, animal research center, and research laboratory of a women's hospital. PATIENTS/ANIMALS A total of 436 PCOS case subjects and 715 control subjects were recruited for the survey. In vivo and ex vivo studies were conducted in PCOS-model rats and on ovarian GCs collected from women with PCOS and control subjects. INTERVENTION(S) The PCOS rat model was established with the use of testosterone propionate. MAIN OUTCOME MEASURE(S) Assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq), RNA sequencing, rhythmicity analysis, functional enrichment analysis. RESULT(S) There was a significant correlation between night shift work and PCOS. PCOS-model rats presented distinct differences in the circadian variation of corticotropin-releasing hormone, adrenocorticotropic hormone, prolactin, and a 4-h phase delay in thyrotropic hormone levels. The motif enrichment analysis of ATAC-seq revealed the absence of clock-related transcription factors in specific peaks of PCOS group, and RNA sequencing ex vivo at various time points over 24 hours demonstrated the differential rhythmic expression patterns of women with PCOS. Kyoto Encyclopedia of Genes and Genomes analysis further highlighted metabolic dysfunction, including both carbohydrate and amino acid metabolism and the tricarboxylic acid cycle. CONCLUSION(S) There is a significant association of night shift work with PCOS, and genome-wide chronodisruption exists in ovarian GCs.
Collapse
Affiliation(s)
- Fangfang Wang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Institute for Women's Health, University College London, London, United Kingdom
| | - Ningning Xie
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Yan Wu
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Qing Zhang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Yuhang Zhu
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Minchen Dai
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Jue Zhou
- College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China; Institute for Women's Health, University College London, London, United Kingdom
| | - Jiexue Pan
- First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Mengling Tang
- School of Public Health, Zhejiang University, Hangzhou, China
| | - Qi Cheng
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Biwei Shi
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Qinyuan Guo
- Maternal and Child Healthcare Hospital of Liuzhou, Liuzhou, China
| | - Xinling Li
- Maternal and Child Healthcare Hospital of Liuzhou, Liuzhou, China
| | - Lifeng Xie
- Maternal and Child Healthcare Hospital of Liuzhou, Liuzhou, China
| | - Bing Wang
- Second Hospital of Jiaxing, Jiaxing, China
| | - Dongxia Yang
- Second Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qing Weng
- First People's Hospital of Yuhang District of Hangzhou, Hangzhou, China
| | - Lanzhong Guo
- Dongyang Women's and Children's Hospital, Dongyang, China
| | - Jisheng Ye
- Dongyang Women's and Children's Hospital, Dongyang, China
| | - Mingwo Pan
- Guangdong Women and Children Hospital, Guangzhou, China
| | - Shuyi Zhang
- Baiyin City Maternity and Childcare Hospital, Baiyin, China
| | - Hua Zhou
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Cailan Zhen
- People's Hospital of Lucheng, Lucheng, China
| | - Ping Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second Hospital, Chengdu, People's Republic of China
| | - Ke Ning
- Department of Social Science, Institute of Education, University College London, London, United Kingdom
| | - Lisa Brackenridge
- Institute for Women's Health, University College London, London, United Kingdom
| | - Paul J Hardiman
- Institute for Women's Health, University College London, London, United Kingdom
| | - Fan Qu
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Institute for Women's Health, University College London, London, United Kingdom.
| |
Collapse
|
38
|
Tripathi LP, Itoh MN, Takeda Y, Tsujino K, Kondo Y, Kumanogoh A, Mizuguchi K. Integrative Analysis Reveals Common and Unique Roles of Tetraspanins in Fibrosis and Emphysema. Front Genet 2020; 11:585998. [PMID: 33424923 PMCID: PMC7793877 DOI: 10.3389/fgene.2020.585998] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 10/28/2020] [Indexed: 11/17/2022] Open
Abstract
While both chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF) are multifactorial disorders characterized by distinct clinical and pathological features, their commonalities and differences have not been fully elucidated. We sought to investigate the preventive roles of tetraspanins Cd151 and Cd9 -that are involved in diverse cellular processes in lung pathophysiology- in pulmonary fibrosis and emphysema, respectively, and to obtain a deeper understanding of their underlying molecular mechanisms toward facilitating improved therapeutic outcomes. Using an integrative approach, we examined the transcriptomic changes in the lungs of Cd151- and Cd9-deficient mice using functional-enrichment-analysis, pathway-perturbation-analysis and protein-protein-interaction (PPI) network analysis. Circadian-rhythm, extracellular-matrix (ECM), cell-adhesion and inflammatory responses and associated factors were prominently influenced by Cd151-deletion. Conversely, cellular-junctions, focal-adhesion, vascular-remodeling, and TNF-signaling were deeply impacted by Cd9-deletion. We also highlighted a “common core” of factors and signaling cascades that underlie the functions of both Cd151 and Cd9 in lung pathology. Circadian dysregulation following Cd151-deletion seemingly facilitated progressive fibrotic lung phenotype. Conversely, TGF-β signaling attenuation and TNF-signaling activation emerged as potentially novel functionaries of Cd9-deletion-induced emphysema. Our findings offer promising avenues for developing novel therapeutic treatments for pulmonary fibrosis and emphysema.
Collapse
Affiliation(s)
- Lokesh P Tripathi
- National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Japan.,Artificial Intelligence Center for Health and Biomedical Research (ArCHER), National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Japan
| | - Mari N Itoh
- National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Japan.,Artificial Intelligence Center for Health and Biomedical Research (ArCHER), National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Japan
| | - Yoshito Takeda
- Department of Respiratory Medicine, Allergy and Rheumatic Diseases, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kazuyuki Tsujino
- Department of Respiratory Medicine, Allergy and Rheumatic Diseases, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yasushi Kondo
- Research Division, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine, Allergy and Rheumatic Diseases, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kenji Mizuguchi
- National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Japan.,Artificial Intelligence Center for Health and Biomedical Research (ArCHER), National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Japan.,Institute for Protein Research, Osaka University, Suita, Japan
| |
Collapse
|
39
|
The circadian machinery links metabolic disorders and depression: A review of pathways, proteins and potential pharmacological interventions. Life Sci 2020; 265:118809. [PMID: 33249097 DOI: 10.1016/j.lfs.2020.118809] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/11/2020] [Accepted: 11/18/2020] [Indexed: 12/16/2022]
Abstract
Circadian rhythms are responsible for regulating a number of physiological processes. The central oscillator is located within the suprachiasmatic nucleus (SCN) of the hypothalamus and the SCN synchronises the circadian clocks that are found in our peripheral organs through neural and humoral signalling. At the molecular level, biological clocks consist of transcription-translation feedback loops (TTFLs) and these pathways are influenced by transcription factors, post-translational modifications, signalling pathways and epigenetic modifiers. When disruptions occur in the circadian machinery, the activities of the proteins implicated in this network and the expression of core clock or clock-controlled genes (CCGs) can be altered. Circadian misalignment can also arise when there is desychronisation between our internal clocks and environmental stimuli. There is evidence in the literature demonstrating that disturbances in the circadian rhythm contribute to the pathophysiology of several diseases and disorders. This includes the metabolic syndrome and recently, it has been suggested that the 'circadian syndrome' may be a more appropriate term to use to not only describe the cardio-metabolic risk factors but also the associated comorbidities. Here we overview the molecular architecture of circadian clocks in mammals and provide insight into the effects of shift work, exposure to artificial light, food intake and stress on the circadian rhythm. The relationship between circadian rhythms, metabolic disorders and depression is reviewed and this is a topic that requires further investigation. We also describe how particular proteins involved in the TTFLs can be potentially modulated by small molecules, including pharmacological interventions and dietary compounds.
Collapse
|
40
|
Does a high-fat diet affect the circadian clock, or is it the other way around? A systematic review. Nutr Res 2020; 84:1-13. [PMID: 33213889 DOI: 10.1016/j.nutres.2020.10.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 09/25/2020] [Accepted: 10/08/2020] [Indexed: 10/23/2022]
Abstract
This paper reviews studies that addressed the influence of diet on circadian rhythmicity in mice and, in turn, circadian clock chronodisruption and its role in the development of metabolic disorders. Studies from the past 14 years were selected via a systematic search conducted using the PubMed electronic database. After applying the inclusion and exclusion criteria, 291 studies were selected, of which 13 were chosen using the following inclusion criteria: use of a high-fat diet for mice, evaluation of clock gene expression, and the association between chronodisruption and lipid metabolism disorders. These studies reported changes in animals' biological clock when they developed metabolic disorders by consuming a high-fat diet. It was also evident that some clock gene mutations or deletions triggered metabolic changes. Disturbances of clock gene machinery may play important roles in lipid metabolism and the development of atherosclerotic processes. However, many metabolic processes also affect the function of clock genes and circadian systems. In summary, this review's results may provide new insights into the reciprocal regulation of energy homeostasis and the biological clock.
Collapse
|
41
|
Kim YH, Lazar MA. Transcriptional Control of Circadian Rhythms and Metabolism: A Matter of Time and Space. Endocr Rev 2020; 41:5835826. [PMID: 32392281 PMCID: PMC7334005 DOI: 10.1210/endrev/bnaa014] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/04/2020] [Indexed: 02/07/2023]
Abstract
All biological processes, living organisms, and ecosystems have evolved with the Sun that confers a 24-hour periodicity to life on Earth. Circadian rhythms arose from evolutionary needs to maximize daily organismal fitness by enabling organisms to mount anticipatory and adaptive responses to recurrent light-dark cycles and associated environmental changes. The clock is a conserved feature in nearly all forms of life, ranging from prokaryotes to virtually every cell of multicellular eukaryotes. The mammalian clock comprises transcription factors interlocked in negative feedback loops, which generate circadian expression of genes that coordinate rhythmic physiology. In this review, we highlight previous and recent studies that have advanced our understanding of the transcriptional architecture of the mammalian clock, with a specific focus on epigenetic mechanisms, transcriptomics, and 3-dimensional chromatin architecture. In addition, we discuss reciprocal ways in which the clock and metabolism regulate each other to generate metabolic rhythms. We also highlight implications of circadian biology in human health, ranging from genetic and environment disruptions of the clock to novel therapeutic opportunities for circadian medicine. Finally, we explore remaining fundamental questions and future challenges to advancing the field forward.
Collapse
Affiliation(s)
- Yong Hoon Kim
- Institute for Diabetes, Obesity, and Metabolism, and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Mitchell A Lazar
- Institute for Diabetes, Obesity, and Metabolism, and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
42
|
Kinoshita C, Okamoto Y, Aoyama K, Nakaki T. MicroRNA: A Key Player for the Interplay of Circadian Rhythm Abnormalities, Sleep Disorders and Neurodegenerative Diseases. Clocks Sleep 2020; 2:282-307. [PMID: 33089205 PMCID: PMC7573810 DOI: 10.3390/clockssleep2030022] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 07/21/2020] [Indexed: 02/06/2023] Open
Abstract
Circadian rhythms are endogenous 24-h oscillators that regulate the sleep/wake cycles and the timing of biological systems to optimize physiology and behavior for the environmental day/night cycles. The systems are basically generated by transcription-translation feedback loops combined with post-transcriptional and post-translational modification. Recently, evidence is emerging that additional non-coding RNA-based mechanisms are also required to maintain proper clock function. MicroRNA is an especially important factor that plays critical roles in regulating circadian rhythm as well as many other physiological functions. Circadian misalignment not only disturbs the sleep/wake cycle and rhythmic physiological activity but also contributes to the development of various diseases, such as sleep disorders and neurodegenerative diseases. The patient with neurodegenerative diseases often experiences profound disruptions in their circadian rhythms and/or sleep/wake cycles. In addition, a growing body of recent evidence implicates sleep disorders as an early symptom of neurodegenerative diseases, and also suggests that abnormalities in the circadian system lead to the onset and expression of neurodegenerative diseases. The genetic mutations which cause the pathogenesis of familial neurodegenerative diseases have been well studied; however, with the exception of Huntington's disease, the majority of neurodegenerative diseases are sporadic. Interestingly, the dysfunction of microRNA is increasingly recognized as a cause of sporadic neurodegenerative diseases through the deregulated genes related to the pathogenesis of neurodegenerative disease, some of which are the causative genes of familial neurodegenerative diseases. Here we review the interplay of circadian rhythm disruption, sleep disorders and neurodegenerative disease, and its relation to microRNA, a key regulator of cellular processes.
Collapse
Affiliation(s)
- Chisato Kinoshita
- Department of Pharmacology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan; (C.K.); (Y.O.); (K.A.)
| | - Yayoi Okamoto
- Department of Pharmacology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan; (C.K.); (Y.O.); (K.A.)
- Teikyo University Support Center for Women Physicians and Researchers, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
| | - Koji Aoyama
- Department of Pharmacology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan; (C.K.); (Y.O.); (K.A.)
| | - Toshio Nakaki
- Department of Pharmacology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan; (C.K.); (Y.O.); (K.A.)
- Faculty of Pharma-Science, Teikyo University, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
| |
Collapse
|
43
|
O'Connell EJ, Martinez CA, Liang YG, Cistulli PA, Cook KM. Out of breath, out of time: interactions between HIF and circadian rhythms. Am J Physiol Cell Physiol 2020; 319:C533-C540. [PMID: 32726159 DOI: 10.1152/ajpcell.00305.2020] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Humans have internal circadian clocks that ensure that important physiological functions occur at specific times of the day. These molecular clocks are regulated at the genomic level and exist in most cells of the body. Multiple circadian resetting cues have been identified, including light, temperature, and food. Recently, oxygen has been identified as a resetting cue, and emerging science indicates that this occurs through interactions at the cellular level between the circadian transcription-translation feedback loop and the hypoxia-inducible pathway (hypoxia-inducible factor; subject of the 2019 Nobel Prize in Physiology or Medicine). This review will cover recently identified relationships between HIF and proteins of the circadian clock. Interactions between the circadian clock and hypoxia could have wide-reaching implications for human diseases, and understanding the molecular mechanisms regulating these overlapping pathways may open up new strategies for drug discovery.
Collapse
Affiliation(s)
- Emma J O'Connell
- University of Sydney, Faculty of Medicine and Health and Charles Perkins Centre, Camperdown, New South Wales, Australia
| | - Chloe-Anne Martinez
- University of Sydney, Faculty of Medicine and Health and Charles Perkins Centre, Camperdown, New South Wales, Australia
| | - Yichuan G Liang
- University of Sydney, Faculty of Medicine and Health and Charles Perkins Centre, Camperdown, New South Wales, Australia
| | - Peter A Cistulli
- University of Sydney, Faculty of Medicine and Health and Charles Perkins Centre, Camperdown, New South Wales, Australia
| | - Kristina M Cook
- University of Sydney, Faculty of Medicine and Health and Charles Perkins Centre, Camperdown, New South Wales, Australia
| |
Collapse
|
44
|
Egstrand S, Nordholm A, Morevati M, Mace ML, Hassan A, Naveh-Many T, Rukov JL, Gravesen E, Olgaard K, Lewin E. A molecular circadian clock operates in the parathyroid gland and is disturbed in chronic kidney disease associated bone and mineral disorder. Kidney Int 2020; 98:1461-1475. [PMID: 32721445 DOI: 10.1016/j.kint.2020.06.034] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 01/16/2023]
Abstract
Circadian rhythms in metabolism, hormone secretion, cell cycle and locomotor activity are regulated by a molecular circadian clock with the master clock in the suprachiasmatic nucleus of the central nervous system. However, an internal clock is also expressed in several peripheral tissues. Although about 10% of all genes are regulated by clock machinery an internal molecular circadian clock in the parathyroid glands has not previously been investigated. Parathyroid hormone secretion exhibits a diurnal variation and parathyroid hormone gene promoter contains an E-box like element, a known target of circadian clock proteins. Therefore, we examined whether an internal molecular circadian clock is operating in parathyroid glands, whether it is entrained by feeding and how it responds to chronic kidney disease. As uremia is associated with extreme parathyroid growth and since disturbed circadian rhythm is related to abnormal growth, we examined the expression of parathyroid clock and clock-regulated cell cycle genes in parathyroid glands of normal and uremic rats. Circadian clock genes were found to be rhythmically expressed in normal parathyroid glands and this clock was minimally entrained by feeding. Diurnal regulation of parathyroid glands was next examined. Significant rhythmicity of fibroblast-growth-factor-receptor-1, MafB and Gata3 was found. In uremic rats, deregulation of circadian clock genes and the cell cycle regulators, Cyclin D1, c-Myc, Wee1 and p27, which are influenced by the circadian clock, was found in parathyroid glands as well as the aorta. Thus, a circadian clock operates in parathyroid glands and this clock and downstream cell cycle regulators are disturbed in uremia and may contribute to dysregulated parathyroid proliferation in secondary hyperparathyroidism.
Collapse
Affiliation(s)
- Søren Egstrand
- Nephrological Department, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark; Nephrological Department, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Anders Nordholm
- Nephrological Department, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark; Nephrological Department, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Marya Morevati
- Nephrological Department, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Maria L Mace
- Nephrological Department, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Alia Hassan
- Minerva Center for Calcium and Bone Metabolism, Nephrology Services, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Tally Naveh-Many
- Minerva Center for Calcium and Bone Metabolism, Nephrology Services, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Jakob L Rukov
- Nephrological Department, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Eva Gravesen
- Nephrological Department, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Klaus Olgaard
- Nephrological Department, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ewa Lewin
- Nephrological Department, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark; Nephrological Department, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW Blood pressure (BP) exhibits strong diurnal variations that have been shown to be important for normal physiology and health. In this review, we highlight recent advances in both basic and clinic research on how the circadian clock affects these BP rhythms. RECENT FINDINGS Tissue-specific and inducible knockout rodent models have provided novel ways to dissect how circadian clocks regulate BP rhythms and demonstrated that loss of these rhythms is associated with the development of disease. The use of circadian-specific research protocols has translated findings from rodent models to humans, providing insight into circadian control of BP, as well as how sleep, activity, and other factors influence diurnal BP rhythms. Circadian mechanisms play an important role in the regulation of diurnal BP rhythms. Future research needs to extend these findings to clinical populations and determine the extent to which circadian factors may play a role in the development of novel treatment approaches to the management of hypertension.
Collapse
Affiliation(s)
- Megan K Rhoads
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Vikhram Balagee
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - S Justin Thomas
- Department of Psychiatry, University of Alabama at Birmingham, SC1010, 1720 2nd Avenue South, Birmingham, AL, 35294-0017, USA.
| |
Collapse
|
46
|
Abstract
Background: Sleep disturbances are a common symptom of major depressive disorder (MDD). Sleep is highly regulated by circadian rhythms, controlled by circadian genes, that act through a series of feedback loops to regulate the sleep-wake cycle.Objectives: To the best of our knowledge, a systematic review regarding the core circadian genes and their role in MDD has not been published recently. Also, a review of these genes and their role in sleep disturbances in depressed individuals appears to have never been done. We decided to integrate both concepts into one comprehensive review.Method: The review was done using the appropriate search terms in the following search engines: OVID Medline, Embase, PsycINFO and Pubmed.Results: Based on the data reviewed, none of the circadian genes appear to be associated with MDD, but some are more promising than others. These genes are: CRY1, CRY2, PER2 and NPAS2. When investigating the role of circadian genes in sleep disturbances among individuals with MDD, the most promising candidate gene is TIMELESS. Although the results in this area are limited.Conclusion: Given the promising leads from this review, future studies should investigate circadian genes in sleep disturbances among the depressed population.
Collapse
Affiliation(s)
- Lindsay Melhuish Beaupre
- Institute of Medical Sciences, University of Toronto, Toronto, Canada.,Centre for Addiction & Mental Health, Neurogenetics Section, Toronto, Canada
| | - Gregory M Brown
- Centre for Addiction & Mental Health, Neurogenetics Section, Toronto, Canada.,Department of Psychiatry, University of Toronto, Toronto, Canada
| | - James L Kennedy
- Institute of Medical Sciences, University of Toronto, Toronto, Canada.,Centre for Addiction & Mental Health, Neurogenetics Section, Toronto, Canada.,Department of Psychiatry, University of Toronto, Toronto, Canada
| |
Collapse
|
47
|
De Nobrega AK, Luz KV, Lyons LC. Resetting the Aging Clock: Implications for Managing Age-Related Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1260:193-265. [PMID: 32304036 DOI: 10.1007/978-3-030-42667-5_9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Worldwide, individuals are living longer due to medical and scientific advances, increased availability of medical care and changes in public health policies. Consequently, increasing attention has been focused on managing chronic conditions and age-related diseases to ensure healthy aging. The endogenous circadian system regulates molecular, physiological and behavioral rhythms orchestrating functional coordination and processes across tissues and organs. Circadian disruption or desynchronization of circadian oscillators increases disease risk and appears to accelerate aging. Reciprocally, aging weakens circadian function aggravating age-related diseases and pathologies. In this review, we summarize the molecular composition and structural organization of the circadian system in mammals and humans, and evaluate the technological and societal factors contributing to the increasing incidence of circadian disorders. Furthermore, we discuss the adverse effects of circadian dysfunction on aging and longevity and the bidirectional interactions through which aging affects circadian function using examples from mammalian research models and humans. Additionally, we review promising methods for managing healthy aging through behavioral and pharmacological reinforcement of the circadian system. Understanding age-related changes in the circadian clock and minimizing circadian dysfunction may be crucial components to promote healthy aging.
Collapse
Affiliation(s)
- Aliza K De Nobrega
- Department of Biological Science, Program in Neuroscience, Florida State University, Tallahassee, FL, USA
| | - Kristine V Luz
- Department of Biological Science, Program in Neuroscience, Florida State University, Tallahassee, FL, USA
| | - Lisa C Lyons
- Department of Biological Science, Program in Neuroscience, Florida State University, Tallahassee, FL, USA.
| |
Collapse
|
48
|
Molecular mechanisms and physiological importance of circadian rhythms. Nat Rev Mol Cell Biol 2019; 21:67-84. [PMID: 31768006 DOI: 10.1038/s41580-019-0179-2] [Citation(s) in RCA: 675] [Impact Index Per Article: 112.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2019] [Indexed: 12/12/2022]
Abstract
To accommodate daily recurring environmental changes, animals show cyclic variations in behaviour and physiology, which include prominent behavioural states such as sleep-wake cycles but also a host of less conspicuous oscillations in neurological, metabolic, endocrine, cardiovascular and immune functions. Circadian rhythmicity is created endogenously by genetically encoded molecular clocks, whose components cooperate to generate cyclic changes in their own abundance and activity, with a periodicity of about a day. Throughout the body, such molecular clocks convey temporal control to the function of organs and tissues by regulating pertinent downstream programmes. Synchrony between the different circadian oscillators and resonance with the solar day is largely enabled by a neural pacemaker, which is directly responsive to certain environmental cues and able to transmit internal time-of-day representations to the entire body. In this Review, we discuss aspects of the circadian clock in Drosophila melanogaster and mammals, including the components of these molecular oscillators, the function and mechanisms of action of central and peripheral clocks, their synchronization and their relevance to human health.
Collapse
|
49
|
Diurnal oscillations of endogenous H 2O 2 sustained by p66 Shc regulate circadian clocks. Nat Cell Biol 2019; 21:1553-1564. [PMID: 31768048 DOI: 10.1038/s41556-019-0420-4] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 10/01/2019] [Indexed: 02/04/2023]
Abstract
Redox balance, an essential feature of healthy physiological steady states, is regulated by circadian clocks, but whether or how endogenous redox signalling conversely regulates clockworks in mammals remains unknown. Here, we report circadian rhythms in the levels of endogenous H2O2 in mammalian cells and mouse livers. Using an unbiased method to screen for H2O2-sensitive transcription factors, we discovered that rhythmic redox control of CLOCK directly by endogenous H2O2 oscillations is required for proper intracellular clock function. Importantly, perturbations in the rhythm of H2O2 levels induced by the loss of p66Shc, which oscillates rhythmically in the liver and suprachiasmatic nucleus (SCN) of mice, disturb the rhythmic redox control of CLOCK function, reprogram hepatic transcriptome oscillations, lengthen the circadian period in mice and modulate light-induced clock resetting. Our findings suggest that redox signalling rhythms are intrinsically coupled to the circadian system through reversible oxidative modification of CLOCK and constitute essential mechanistic timekeeping components in mammals.
Collapse
|
50
|
Dong Z, Zhang G, Qu M, Gimple RC, Wu Q, Qiu Z, Prager BC, Wang X, Kim LJY, Morton AR, Dixit D, Zhou W, Huang H, Li B, Zhu Z, Bao S, Mack SC, Chavez L, Kay SA, Rich JN. Targeting Glioblastoma Stem Cells through Disruption of the Circadian Clock. Cancer Discov 2019; 9:1556-1573. [PMID: 31455674 PMCID: PMC6983300 DOI: 10.1158/2159-8290.cd-19-0215] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 05/29/2019] [Accepted: 08/01/2019] [Indexed: 12/13/2022]
Abstract
Glioblastomas are highly lethal cancers, containing self-renewing glioblastoma stem cells (GSC). Here, we show that GSCs, differentiated glioblastoma cells (DGC), and nonmalignant brain cultures all displayed robust circadian rhythms, yet GSCs alone displayed exquisite dependence on core clock transcription factors, BMAL1 and CLOCK, for optimal cell growth. Downregulation of BMAL1 or CLOCK in GSCs induced cell-cycle arrest and apoptosis. Chromatin immunoprecipitation revealed that BMAL1 preferentially bound metabolic genes and was associated with active chromatin regions in GSCs compared with neural stem cells. Targeting BMAL1 or CLOCK attenuated mitochondrial metabolic function and reduced expression of tricarboxylic acid cycle enzymes. Small-molecule agonists of two independent BMAL1-CLOCK negative regulators, the cryptochromes and REV-ERBs, downregulated stem cell factors and reduced GSC growth. Combination of cryptochrome and REV-ERB agonists induced synergistic antitumor efficacy. Collectively, these findings show that GSCs co-opt circadian regulators beyond canonical circadian circuitry to promote stemness maintenance and metabolism, offering novel therapeutic paradigms. SIGNIFICANCE: Cancer stem cells are highly malignant tumor-cell populations. We demonstrate that GSCs selectively depend on circadian regulators, with increased binding of the regulators in active chromatin regions promoting tumor metabolism. Supporting clinical relevance, pharmacologic targeting of circadian networks specifically disrupted cancer stem cell growth and self-renewal.This article is highlighted in the In This Issue feature, p. 1469.
Collapse
Affiliation(s)
- Zhen Dong
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center and Sanford Consortium for Regenerative Medicine, University of California, San Diego, California
| | - Guoxin Zhang
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center and Sanford Consortium for Regenerative Medicine, University of California, San Diego, California
| | - Meng Qu
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Ryan C Gimple
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center and Sanford Consortium for Regenerative Medicine, University of California, San Diego, California
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Qiulian Wu
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center and Sanford Consortium for Regenerative Medicine, University of California, San Diego, California
| | - Zhixin Qiu
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center and Sanford Consortium for Regenerative Medicine, University of California, San Diego, California
| | - Briana C Prager
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center and Sanford Consortium for Regenerative Medicine, University of California, San Diego, California
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Xiuxing Wang
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center and Sanford Consortium for Regenerative Medicine, University of California, San Diego, California
| | - Leo J Y Kim
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center and Sanford Consortium for Regenerative Medicine, University of California, San Diego, California
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Andrew R Morton
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Deobrat Dixit
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center and Sanford Consortium for Regenerative Medicine, University of California, San Diego, California
| | - Wenchao Zhou
- Department of Cancer Biology, Center for Cancer Stem Cell Research, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Haidong Huang
- Department of Cancer Biology, Center for Cancer Stem Cell Research, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Bin Li
- Ludwig Institute for Cancer Research, La Jolla, California
| | - Zhe Zhu
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center and Sanford Consortium for Regenerative Medicine, University of California, San Diego, California
| | - Shideng Bao
- Department of Cancer Biology, Center for Cancer Stem Cell Research, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Stephen C Mack
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Lukas Chavez
- Department of Medicine, University of California, San Diego, California
| | - Steve A Kay
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California.
| | - Jeremy N Rich
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center and Sanford Consortium for Regenerative Medicine, University of California, San Diego, California.
| |
Collapse
|