1
|
Curran K, Kemper F, Hadley M. Time of day dependent changes in embryonic heart rate are detectable after maturation of rhythmic circadian gene expression in the eye, but before the heart in Xenopus laevis tadpoles cultured in LD. MICROPUBLICATION BIOLOGY 2024; 2024. [PMID: 39291148 PMCID: PMC11406323 DOI: 10.17912/micropub.biology.001277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 09/19/2024]
Abstract
We systematically characterized onset of expression of circadian genes in the embryonic eye and heart of Xenopus laevis tadpoles. We found that period1 (per1) and nr1d1 ( rev-erbα) were the first circadian genes to display significant 24-hour rhythms in the developing eye and heart in a 12-hour light-dark cycle (LD). Rhythmic expression of both oscillator and output genes were present in the eye by 2.75 days post fertilization (dpf), but not in 15 dpf hearts. Surprisingly, rhythmic oscillation of heart rate occurred after 3.2 dpf suggesting that heart rate may be controlled directly by light or indirectly by the pineal in LD.
Collapse
Affiliation(s)
- Kristen Curran
- Biology, University of Wisconsin-Whitewater, Whitewater, Wisconsin, United States
| | - Faith Kemper
- Biology, University of Wisconsin-Whitewater, Whitewater, Wisconsin, United States
- Wasseen, Inc. Milwaukee, Wisconsin, United States
| | - Morgan Hadley
- Biology, University of Wisconsin-Whitewater, Whitewater, Wisconsin, United States
- University of Kansas Medical Center, Kansas City, Kansas, United States
| |
Collapse
|
2
|
Yagita K. Emergence of the circadian clock oscillation during the developmental process in mammals. Curr Opin Genet Dev 2024; 84:102152. [PMID: 38266394 DOI: 10.1016/j.gde.2024.102152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/23/2023] [Accepted: 01/02/2024] [Indexed: 01/26/2024]
Abstract
The circadian clocks are cell-autonomous intrinsic oscillators existing throughout the body to coordinate intracellular and intercellular functions of each organ or tissue. The circadian clock oscillation gradually emerges during mid-to-late gestation in the mammalian developmental process. Recently, it has been revealed that the in vitro differentiation of mouse ES cells recapitulates the circadian clock development. Moreover, reprogramming of the cells results in the redisappearance of the clock, indicating that circadian clocks are tightly coupled with cellular differentiation. Interestingly, before the circadian clock develops, the embryo is governed under ultradian rhythms driven by the segmentation clock. This short review explores these observations, discussing the significance of the emergence of circadian clock oscillation during the mammalian developmental process.
Collapse
Affiliation(s)
- Kazuhiro Yagita
- Department of Physiology and Systems Bioscience, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan.
| |
Collapse
|
3
|
Comas M, De Pietri Tonelli D, Berdondini L, Astiz M. Ontogeny of the circadian system: a multiscale process throughout development. Trends Neurosci 2024; 47:36-46. [PMID: 38071123 DOI: 10.1016/j.tins.2023.11.004] [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: 07/23/2023] [Revised: 10/02/2023] [Accepted: 11/12/2023] [Indexed: 01/12/2024]
Abstract
The 24 h (circadian) timing system develops in mammals during the perinatal period. It carries out the essential task of anticipating daily recurring environmental changes to identify the best time of day for each molecular, cellular, and systemic process. Although significant knowledge has been acquired about the organization and function of the adult circadian system, relatively little is known about its ontogeny. During the perinatal period, the circadian system progressively gains functionality under the influence of the early environment. This review explores current evidence on the development of the circadian clock in mammals, highlighting the multilevel complexity of the process and the importance of gaining a better understanding of its underlying biology.
Collapse
Affiliation(s)
- Maria Comas
- Circadian Physiology of Neurons and Glia Laboratory, Achucarro Basque Center for Neuroscience, 48940 Leioa, Basque Country, Spain
| | | | - Luca Berdondini
- Microtechnology for Neuroelectronics, Fondazione Istituto Italiano di Tecnologia (IIT), 16163 Genova, Italy
| | - Mariana Astiz
- Circadian Physiology of Neurons and Glia Laboratory, Achucarro Basque Center for Neuroscience, 48940 Leioa, Basque Country, Spain; Ikerbasque - Basque Foundation for Science, Bilbao, Spain; Institute of Neurobiology, University of Lübeck, 23562 Lübeck, Germany.
| |
Collapse
|
4
|
Chen R, Routh BN, Gaudet AD, Fonken LK. Circadian Regulation of the Neuroimmune Environment Across the Lifespan: From Brain Development to Aging. J Biol Rhythms 2023; 38:419-446. [PMID: 37357738 PMCID: PMC10475217 DOI: 10.1177/07487304231178950] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023]
Abstract
Circadian clocks confer 24-h periodicity to biological systems, to ultimately maximize energy efficiency and promote survival in a world with regular environmental light cycles. In mammals, circadian rhythms regulate myriad physiological functions, including the immune, endocrine, and central nervous systems. Within the central nervous system, specialized glial cells such as astrocytes and microglia survey and maintain the neuroimmune environment. The contributions of these neuroimmune cells to both homeostatic and pathogenic demands vary greatly across the day. Moreover, the function of these cells changes across the lifespan. In this review, we discuss circadian regulation of the neuroimmune environment across the lifespan, with a focus on microglia and astrocytes. Circadian rhythms emerge in early life concurrent with neuroimmune sculpting of brain circuits and wane late in life alongside increasing immunosenescence and neurodegeneration. Importantly, circadian dysregulation can alter immune function, which may contribute to susceptibility to neurodevelopmental and neurodegenerative diseases. In this review, we highlight circadian neuroimmune interactions across the lifespan and share evidence that circadian dysregulation within the neuroimmune system may be a critical component in human neurodevelopmental and neurodegenerative diseases.
Collapse
Affiliation(s)
- Ruizhuo Chen
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
| | - Brandy N. Routh
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
- Institute for Neuroscience, The University of Texas at Austin, Austin, Texas
| | - Andrew D. Gaudet
- Institute for Neuroscience, The University of Texas at Austin, Austin, Texas
- Department of Psychology, The University of Texas at Austin, Austin, Texas
- Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, Texas
| | - Laura K. Fonken
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
- Institute for Neuroscience, The University of Texas at Austin, Austin, Texas
| |
Collapse
|
5
|
Van Gilst D, Puchkina AV, Roelants JA, Kervezee L, Dudink J, Reiss IKM, Van Der Horst GTJ, Vermeulen MJ, Chaves I. Effects of the neonatal intensive care environment on circadian health and development of preterm infants. Front Physiol 2023; 14:1243162. [PMID: 37719464 PMCID: PMC10500197 DOI: 10.3389/fphys.2023.1243162] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/18/2023] [Indexed: 09/19/2023] Open
Abstract
The circadian system in mammals ensures adaptation to the light-dark cycle on Earth and imposes 24-h rhythmicity on metabolic, physiological and behavioral processes. The central circadian pacemaker is located in the brain and is entrained by environmental signals called Zeitgebers. From here, neural, humoral and systemic signals drive rhythms in peripheral clocks in nearly every mammalian tissue. During pregnancy, disruption of the complex interplay between the mother's rhythmic signals and the fetal developing circadian system can lead to long-term health consequences in the offspring. When an infant is born very preterm, it loses the temporal signals received from the mother prematurely and becomes totally dependent on 24/7 care in the Neonatal Intensive Care Unit (NICU), where day/night rhythmicity is usually blurred. In this literature review, we provide an overview of the fetal and neonatal development of the circadian system, and short-term consequences of disruption of this process as occurs in the NICU environment. Moreover, we provide a theoretical and molecular framework of how this disruption could lead to later-life disease. Finally, we discuss studies that aim to improve health outcomes after preterm birth by studying the effects of enhancing rhythmicity in light and noise exposure.
Collapse
Affiliation(s)
- D. Van Gilst
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - A. V. Puchkina
- Department of Developmental Biology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - J. A. Roelants
- Department of Neonatal and Pediatric Intensive Care, Division of Neonatology, Erasmus University Medical Center Rotterdam-Sophia Children’s Hospital, Rotterdam, Netherlands
| | - L. Kervezee
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - J. Dudink
- Department of Neonatology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, Netherlands
| | - I. K. M. Reiss
- Department of Neonatal and Pediatric Intensive Care, Division of Neonatology, Erasmus University Medical Center Rotterdam-Sophia Children’s Hospital, Rotterdam, Netherlands
| | - G. T. J. Van Der Horst
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - M. J. Vermeulen
- Department of Neonatal and Pediatric Intensive Care, Division of Neonatology, Erasmus University Medical Center Rotterdam-Sophia Children’s Hospital, Rotterdam, Netherlands
| | - I. Chaves
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
6
|
Contreras-Correa ZE, Messman RD, Swanson RM, Lemley CO. Melatonin in Health and Disease: A Perspective for Livestock Production. Biomolecules 2023; 13:biom13030490. [PMID: 36979425 PMCID: PMC10046399 DOI: 10.3390/biom13030490] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/21/2023] [Accepted: 03/04/2023] [Indexed: 03/09/2023] Open
Abstract
Mounting evidence in the literature indicates an important role of endogenous and exogenous melatonin in driving physiological and molecular adaptations in livestock. Melatonin has been extensively studied in seasonally polyestrous animals whereby supplementation studies have been used to adjust circannual rhythms in herds of animals under abnormal photoperiodic conditions. Livestock undergo multiple metabolic and physiological adaptation processes throughout their production cycle which can result in decreased immune response leading to chronic illness, weight loss, or decreased production efficiency; however, melatonin’s antioxidant capacity and immunostimulatory properties could alleviate these effects. The cardiovascular system responds to melatonin and depending on receptor type and localization, melatonin can vasodilate or vasoconstrict several systemic arteries, thereby controlling whole animal nutrient partitioning via vascular resistance. Increased incidences of non-communicable diseases in populations exposed to circadian disruption have uncovered novel pathways of neurohormones, such as melatonin, influence health, and disease. Perturbations in immune function can negatively impact the growth and development of livestock which has been examined following melatonin supplementation. Specifically, melatonin can influence nutrient uptake, circulating nutrient profiles, and endocrine profiles controlling economically important livestock growth and development. This review focuses on the physiological, cellular, and molecular implications of melatonin on the health and disease of domesticated food animals.
Collapse
|
7
|
Wong SD, Wright KP, Spencer RL, Vetter C, Hicks LM, Jenni OG, LeBourgeois MK. Development of the circadian system in early life: maternal and environmental factors. J Physiol Anthropol 2022; 41:22. [PMID: 35578354 PMCID: PMC9109407 DOI: 10.1186/s40101-022-00294-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 05/01/2022] [Indexed: 12/25/2022] Open
Abstract
In humans, an adaptable internal biological system generates circadian rhythms that maintain synchronicity of behavior and physiology with the changing demands of the 24-h environment. Development of the circadian system begins in utero and continues throughout the first few years of life. Maturation of the clock can be measured through sleep/wake patterns and hormone secretion. Circadian rhythms, by definition, can persist in the absence of environmental input; however, their ability to adjust to external time cues is vital for adaptation and entrainment to the environment. The significance of these external factors that influence the emergence of a stable circadian clock in the first years of life remain poorly understood. Infants raised in our post-modern world face adverse external circadian signals, such as artificial light and mistimed hormonal cues via breast milk, which may increase interference with the physiological mechanisms that promote circadian synchronization. This review describes the very early developmental stages of the clock and common circadian misalignment scenarios that make the developing circadian system more susceptible to conflicting time cues and temporal disorder between the maternal, fetal, infant, and peripheral clocks.
Collapse
|
8
|
Costello HM, Johnston JG, Juffre A, Crislip GR, Gumz ML. Circadian clocks of the kidney: function, mechanism, and regulation. Physiol Rev 2022; 102:1669-1701. [PMID: 35575250 PMCID: PMC9273266 DOI: 10.1152/physrev.00045.2021] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 05/03/2022] [Accepted: 05/07/2022] [Indexed: 11/22/2022] Open
Abstract
An intrinsic cellular circadian clock is located in nearly every cell of the body. The peripheral circadian clocks within the cells of the kidney contribute to the regulation of a variety of renal processes. In this review, we summarize what is currently known regarding the function, mechanism, and regulation of kidney clocks. Additionally, the effect of extrarenal physiological processes, such as endocrine and neuronal signals, on kidney function is also reviewed. Circadian rhythms in renal function are an integral part of kidney physiology, underscoring the importance of considering time of day as a key biological variable. The field of circadian renal physiology is of tremendous relevance, but with limited physiological and mechanistic information on the kidney clocks this is an area in need of extensive investigation.
Collapse
Affiliation(s)
- Hannah M Costello
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
| | - Jermaine G Johnston
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
- North Florida/South Georgia Malcom Randall Department of Veterans Affairs Medical Center, Gainesville, Florida
| | - Alexandria Juffre
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida
| | - G Ryan Crislip
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
| | - Michelle L Gumz
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
- North Florida/South Georgia Malcom Randall Department of Veterans Affairs Medical Center, Gainesville, Florida
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, Florida
| |
Collapse
|
9
|
Malik A, Nalluri S, De A, Beligala D, Geusz ME. The Relevance of Circadian Clocks to Stem Cell Differentiation and Cancer Progression. NEUROSCI 2022; 3:146-165. [PMID: 39483369 PMCID: PMC11523739 DOI: 10.3390/neurosci3020012] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 03/22/2022] [Indexed: 11/03/2024] Open
Abstract
The molecular mechanism of circadian clocks depends on transcription-translation feedback loops (TTFLs) that have known effects on key cellular processes. However, the distinct role of circadian TTFLs in mammalian stem cells and other less differentiated cells remains poorly understood. Neural stem cells (NSCs) of the brain generate neurons and glia postnatally but also may become cancer stem cells (CSCs), particularly in astrocytomas. Evidence indicates clock TTFL impairment is needed for tumor growth and progression; although, this issue has been examined primarily in more differentiated cancer cells rather than CSCs. Similarly, few studies have examined circadian rhythms in NSCs. After decades of research, it is now well recognized that tumors consist of CSCs and a range of other cancer cells along with noncancerous stromal cells. The circadian properties of these many contributors to tumor properties and treatment outcome are being widely explored. New molecular tools and ones in development will likely enable greater discrimination of important circadian and non-circadian cells within malignancies at multiple stages of cancer progression and following therapy. Here, we focus on adult NSCs and glioma CSCs to address how cells at different stages of differentiation may harbor unique states of the molecular circadian clock influencing differentiation and cell fate.
Collapse
Affiliation(s)
- Astha Malik
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA;
| | - Shreya Nalluri
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA;
| | - Arpan De
- Department of Neurosurgery and Brain Tumor Center, Unit 1004, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA;
| | - Dilshan Beligala
- Department of Molecular Biology and Biotechnology, University of Peradeniya, Peradeniya 20400, Sri Lanka;
| | - Michael E Geusz
- Department of Biological Sciences, Bowling Green State University, Bowling Green, OH 43403, USA;
| |
Collapse
|
10
|
O G, Cascione S, Michielin F, Elvassore N. The emergence of the circadian clock network in hiPSC-derived hepatocytes on chip. Biochem Biophys Res Commun 2022; 601:109-115. [PMID: 35240497 DOI: 10.1016/j.bbrc.2022.02.046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 02/11/2022] [Indexed: 12/20/2022]
Abstract
The circadian clock has paramount implications in physiology and pathology. Although the circadian clock has been widely investigated in adults, up to now very little is known about how circadian rhythms emerge during embryonic development. Some studies about the ontology of the circadian system are focused on the development of the central pacemaker, whereas there is still no agreement about the development of the circadian clock in peripheral tissues. Our work represents the first attempt at investigating the onset of peripheral circadian clocks in the liver, which has a central role in controlling several aspects of human physiology. We profile the emergence of the circadian genes during the transition from the initial state of human pluripotency to the final state of hepatic maturation. We demonstrate that circadian rhythmicity is absent in human pluripotent stem cells, and it arises gradually during the process of hepatic commitment. The clock genes expression reaches a peak at the hepatic progenitor stage. At this point o hiPSC-derived f differentiation the gene oscillations start to be observed with a period of 13 h and approaches 24 h in a later stage when the clock primary feedback loop starts working properly. At the end of differentiation, circadian rhythmicity appears, with genes of primary and secondary feedback loops in antiphase (CLOCK, BMAL1 and REV-ERBα) a sign that the system becomes to be functional.
Collapse
Affiliation(s)
- Gagliano O
- Department of Industrail Engineering, University of Padova, Italy; Veneto Institute of Molecular Medicine, Padova, Italy
| | - S Cascione
- Department of Industrail Engineering, University of Padova, Italy; Veneto Institute of Molecular Medicine, Padova, Italy; San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Milan, Italy
| | - F Michielin
- Department of Industrail Engineering, University of Padova, Italy; Veneto Institute of Molecular Medicine, Padova, Italy; Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - N Elvassore
- Department of Industrail Engineering, University of Padova, Italy; Veneto Institute of Molecular Medicine, Padova, Italy; Great Ormond Street Institute of Child Health, University College London, London, United Kingdom.
| |
Collapse
|
11
|
Bery A, Bagchi U, Bergen AA, Felder-Schmittbuhl MP. Circadian clocks, retinogenesis and ocular health in vertebrates: new molecular insights. Dev Biol 2022; 484:40-56. [DOI: 10.1016/j.ydbio.2022.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/21/2022] [Accepted: 02/01/2022] [Indexed: 12/22/2022]
|
12
|
Dan H, Ruan T, Sampogna RV. Circadian Clock Regulation of Developmental Time in the Kidney. Cell Rep 2021; 31:107661. [PMID: 32433970 DOI: 10.1016/j.celrep.2020.107661] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 03/06/2020] [Accepted: 04/28/2020] [Indexed: 12/14/2022] Open
Abstract
We report the emergence of an endogenous circadian clock that regulates organogenesis in mouse fetal kidney. We detect circadian rhythms both in vivo with transcriptional profiling and ex vivo by bioluminescence. High-resolution structural analysis of embryonic explants reveals that global or local clock disruption results in defects that resemble human congenital abnormalities of the kidney. The onset of fetal rhythms strongly correlates with the timing of a distinct transition in branching and growth rates during a gestational window of high fetal growth demands. Defects in clock mutants typically have been attributed to accelerated aging; however, our study establishes a role for the fetal circadian clock as a developmental timer that regulates the pathways that control organogenesis, branching rate, and nephron number and thus plays a fundamental role in kidney development.
Collapse
Affiliation(s)
- Hanbin Dan
- Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Thomas Ruan
- Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Rosemary V Sampogna
- Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
13
|
Chirico N, Van Laake LW, Sluijter JPG, van Mil A, Dierickx P. Cardiac circadian rhythms in time and space: The future is in 4D. Curr Opin Pharmacol 2020; 57:49-59. [PMID: 33338891 DOI: 10.1016/j.coph.2020.11.006] [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: 09/10/2020] [Revised: 10/25/2020] [Accepted: 11/16/2020] [Indexed: 12/12/2022]
Abstract
The circadian clock synchronizes the body into 24-h cycles, thereby anticipating variations in tissue-specific diurnal tasks, such as response to increased cardiac metabolic demand during the active period of the day. As a result, blood pressure, heart rate, cardiac output, and occurrence of fatal cardiovascular events fluctuate in a diurnal manner. The heart contains different cell types that make up and reside in an environment of biochemical, mechanical, and topographical signaling. Cardiac architecture is essential for proper heart development as well as for maintenance of cell homeostasis and tissue repair. In this review, we describe the possibilities of studying circadian rhythmicity in the heart by using advanced in vitro systems that mimic the native cardiac 3D microenvironment which can be tuned in time and space. Harnessing the knowledge that originates from those in vitro models could significantly improve innovative cardiac modeling and regenerative strategies.
Collapse
Affiliation(s)
- Nino Chirico
- Regenerative Medicine Center, Circulatory Health Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands; Department of Cardiology and Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Linda W Van Laake
- Regenerative Medicine Center, Circulatory Health Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands; Department of Cardiology and Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Joost P G Sluijter
- Regenerative Medicine Center, Circulatory Health Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands; Department of Cardiology and Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Alain van Mil
- Regenerative Medicine Center, Circulatory Health Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands; Department of Cardiology and Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Pieterjan Dierickx
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104, USA; Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104, USA.
| |
Collapse
|
14
|
Bedont JL, Iascone DM, Sehgal A. The Lineage Before Time: Circadian and Nonclassical Clock Influences on Development. Annu Rev Cell Dev Biol 2020; 36:469-509. [PMID: 33021821 PMCID: PMC10826104 DOI: 10.1146/annurev-cellbio-100818-125454] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Diverse factors including metabolism, chromatin remodeling, and mitotic kinetics influence development at the cellular level. These factors are well known to interact with the circadian transcriptional-translational feedback loop (TTFL) after its emergence. What is only recently becoming clear, however, is how metabolism, mitosis, and epigenetics may become organized in a coordinated cyclical precursor signaling module in pluripotent cells prior to the onset of TTFL cycling. We propose that both the precursor module and the TTFL module constrain cellular identity when they are active during development, and that the emergence of these modules themselves is a key lineage marker. Here we review the component pathways underlying these ideas; how proliferation, specification, and differentiation decisions in both developmental and adult stem cell populations are or are not regulated by the classical TTFL; and emerging evidence that we propose implies a primordial clock that precedes the classical TTFL and influences early developmental decisions.
Collapse
Affiliation(s)
- Joseph Lewis Bedont
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
| | - Daniel Maxim Iascone
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
| | - Amita Sehgal
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
- The Howard Hughes Medical Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
15
|
Umemura Y, Yagita K. Development of the Circadian Core Machinery in Mammals. J Mol Biol 2020; 432:3611-3617. [DOI: 10.1016/j.jmb.2019.11.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/26/2019] [Accepted: 11/26/2019] [Indexed: 01/20/2023]
|
16
|
Contreras-Correa ZE, Lemire RL, Burnett DD, Lemley CO. Temporal transcript abundance of clock genes, angiogenic factors and nutrient sensing genes in bovine placental explants. Theriogenology 2020; 151:74-80. [PMID: 32311603 DOI: 10.1016/j.theriogenology.2020.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 03/11/2020] [Accepted: 04/04/2020] [Indexed: 10/24/2022]
Abstract
Recent research has shown expression of clock genes in peripheral tissue explants, targeting multiple pathways leading to the entrainment of circadian rhythms. Temporal variations are not solely regulated by photoperiod, but factors such as maternal feed availability can entrain fetal circadian clock. Currently, a paucity of information exists for clock gene expression and short-term temporal transcript abundance in the bovine placenta, which is essential for proper offspring development. Therefore, the objective of this study was to determine the effect of early to mid-gestational nutrient restriction on clock genes, angiogenic factors, and nutrient sensing genes mRNA transcript abundance in placental explants during a 24 h period. Placentomes from adequately fed and nutrient restricted heifers were collected via Cesarean section at day 180 of gestation; separated into caruncular and cotyledonary tissue and placed in culture media for a 24 h period. The mRNA transcript abundance of clock genes (ARNTL, CRY1, and PER2), angiogenic factors (HIF1A and VEGFA), and nutrient sensing genes (NAMPT and NR3C1) was determined every 4 h. Clock genes were expressed in caruncular and cotyledonary explant tissue. The caruncular explant transcript abundance of the clock genes was not influenced by time (P > 0.05); while ARNTL abundance decreased over time in the cotyledon explant (P < 0.05). A main effect of time was observed for HIF1A, VEGFA, and NR3C1 in the caruncular tissue (P < 0.05). Although, angiogenic factors and nutrient sensing genes in cotyledonary tissue displayed evident temporal variation in transcript abundance (P < 0.05). Nutrient restriction did not alter (P > 0.15) mRNA transcript abundance of clock genes, angiogenic factors, or nutrient sensing genes in either caruncular or cotyledonary tissue. Interestingly, these data may indicate limited transmission and synchronization of maternal and fetal temporal variations in transcript abundance. These findings demonstrate that multiple timepoint collections are needed in future studies due to the innate existence of temporal oscillations observed in the bovine placenta.
Collapse
Affiliation(s)
- Zully E Contreras-Correa
- Department of Animal and Dairy Sciences, Mississippi State University, Mississippi State, MS, United States.
| | - Racheal L Lemire
- Department of Animal and Dairy Sciences, Mississippi State University, Mississippi State, MS, United States.
| | - Derris D Burnett
- Department of Animal and Dairy Sciences, Mississippi State University, Mississippi State, MS, United States.
| | - Caleb O Lemley
- Department of Animal and Dairy Sciences, Mississippi State University, Mississippi State, MS, United States.
| |
Collapse
|
17
|
Tsuchiya Y, Umemura Y, Yagita K. Circadian clock and cancer: From a viewpoint of cellular differentiation. Int J Urol 2020; 27:518-524. [PMID: 32223039 DOI: 10.1111/iju.14231] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 02/27/2020] [Indexed: 12/18/2022]
Abstract
The circadian clock controls and adapts diverse physiological and behavioral processes according to Earth's 24-h cycle of environmental changes. The master pacemaker of the mammalian circadian clock resides in the hypothalamic suprachiasmatic nucleus, but almost all cells throughout the body show circadian oscillations in gene expression patterns and associated functions. Recent studies have shown that the circadian clock gradually develops during embryogenesis. Embryonic stem cells and induced pluripotent stem cells do not show circadian oscillations of gene expression, but gradually develop circadian clock oscillation during differentiation; thus, the developmental program of circadian clock emergence appears closely associated with cellular differentiation. Like embryonic stem cells, certain cancer cell types also lack the circadian clock. Given this similarity between embryonic stem cells and cancer cells, interest is growing in the contributions of circadian clock dysfunction to dedifferentiation and cancer development. In this review, we summarize recent advances in our understanding of circadian clock emergence during ontogenesis, and discuss possible associations with cellular differentiation and carcinogenesis. Considering the multiple physiological functions of circadian rhythms, circadian abnormalities might contribute to a host of diseases, including cancer. Insights on circadian function could lead to the identification of biomarkers for cancer diagnosis and prognosis, as well as novel targets for treatment.
Collapse
Affiliation(s)
- Yoshiki Tsuchiya
- Department of Physiology and Systems Bioscience, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yasuhiro Umemura
- Department of Physiology and Systems Bioscience, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuhiro Yagita
- Department of Physiology and Systems Bioscience, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
18
|
Arata Y, Takagi H. Quantitative Studies for Cell-Division Cycle Control. Front Physiol 2019; 10:1022. [PMID: 31496950 PMCID: PMC6713215 DOI: 10.3389/fphys.2019.01022] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 07/24/2019] [Indexed: 11/13/2022] Open
Abstract
The cell-division cycle (CDC) is driven by cyclin-dependent kinases (CDKs). Mathematical models based on molecular networks, as revealed by molecular and genetic studies, have reproduced the oscillatory behavior of CDK activity. Thus, one basic system for representing the CDC is a biochemical oscillator (CDK oscillator). However, genetically clonal cells divide with marked variability in their total duration of a single CDC round, exhibiting non-Gaussian statistical distributions. Therefore, the CDK oscillator model does not account for the statistical nature of cell-cycle control. Herein, we review quantitative studies of the statistical properties of the CDC. Over the past 70 years, studies have shown that the CDC is driven by a cluster of molecular oscillators. The CDK oscillator is coupled to transcriptional and mitochondrial metabolic oscillators, which cause deterministic chaotic dynamics for the CDC. Recent studies in animal embryos have raised the possibility that the dynamics of molecular oscillators underlying CDC control are affected by allometric volume scaling among the cellular compartments. Considering these studies, we discuss the idea that a cluster of molecular oscillators embedded in different cellular compartments coordinates cellular physiology and geometry for successful cell divisions.
Collapse
Affiliation(s)
| | - Hiroaki Takagi
- Department of Physics, School of Medicine, Nara Medical University, Nara, Japan
| |
Collapse
|
19
|
Mendez N, Torres-Farfan C, Salazar E, Bascur P, Bastidas C, Vergara K, Spichiger C, Halabi D, Vio CP, Richter HG. Fetal Programming of Renal Dysfunction and High Blood Pressure by Chronodisruption. Front Endocrinol (Lausanne) 2019; 10:362. [PMID: 31244775 PMCID: PMC6563621 DOI: 10.3389/fendo.2019.00362] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 05/22/2019] [Indexed: 12/28/2022] Open
Abstract
Adverse prenatal conditions are known to impose significant trade-offs impinging on health and disease balance during adult life. Among several deleterious factors associated with complicated pregnancy, alteration of the gestational photoperiod remains largely unknown. Previously, we reported that prenatal manipulation of the photoperiod has adverse effects on the mother, fetus, and adult offspring; including cardiac hypertrophy. Here, we investigated whether chronic photoperiod shifting (CPS) during gestation may program adult renal function and blood pressure regulation. To this end, pregnant rats were subjected to CPS throughout pregnancy to evaluate the renal effects on the fetus and adult offspring. In the kidney at 18 days of gestation, both clock and clock-controlled gene expression did not display a daily pattern, although there were recurrent weaves of transcriptional activity along the 24 h in the control group. Using DNA microarray, significant differential expression was found for 1,703 transcripts in CPS relative to control fetal kidney (835 up-regulated and 868 down-regulated). Functional genomics assessment revealed alteration of diverse gene networks in the CPS fetal kidney, including regulation of transcription, aldosterone-regulated Na+ reabsorption and connective tissue differentiation. In adult offspring at 90 days of age, circulating proinflammatory cytokines IL-1β and IL-6 were increased under CPS conditions. In these individuals, CPS did not modify kidney clock gene expression but had effects on different genes with specific functions in the nephron. Next, we evaluated several renal markers and the response of blood pressure to 4%NaCl in the diet for 4 weeks (i.e., at 150 days of age). CPS animals displayed elevated systolic blood pressure in basal conditions that remained elevated in response to 4%NaCl, relative to control conditions. At this age, CPS modified the expression of Nhe3, Ncc, Atp1a1, Nr3c1 (glucocorticoid receptor), and Nr3c2 (mineralocorticoid receptor); while Nkcc, Col3A1, and Opn were modified in the CPS 4%+NaCl group. Furthermore, CPS decreased protein expression of Kallikrein and COX-2, both involved in sodium handling. In conclusion, gestational chronodisruption programs kidney dysfunction at different levels, conceivably underlying the prehypertensive phenotype observed in the adult CPS offspring.
Collapse
Affiliation(s)
- Natalia Mendez
- Laboratory of Developmental Chronobiology, Institute of Anatomy, Histology, and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Claudia Torres-Farfan
- Laboratory of Developmental Chronobiology, Institute of Anatomy, Histology, and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Centro Interdisciplinario de Estudios del Sistema Nervioso, Universidad Austral de Chile, Valdivia, Chile
| | - Esteban Salazar
- Laboratory of Developmental Chronobiology, Institute of Anatomy, Histology, and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Pía Bascur
- Laboratory of Developmental Chronobiology, Institute of Anatomy, Histology, and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Carla Bastidas
- Laboratory of Developmental Chronobiology, Institute of Anatomy, Histology, and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Karina Vergara
- Laboratory of Developmental Chronobiology, Institute of Anatomy, Histology, and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Carlos Spichiger
- Faculty of Sciences, Institute of Biochemistry and Microbiology, Universidad Austral de Chile, Valdivia, Chile
| | - Diego Halabi
- Faculty of Medicine, School of Dentistry, Universidad Austral de Chile, Valdivia, Chile
| | - Carlos P. Vio
- Center of Aging and Regeneration CARE, Department of Physiology, Pontificia Universidad Católica de Chile, Valdivia, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastian, Santiago, Chile
| | - Hans G. Richter
- Laboratory of Developmental Chronobiology, Institute of Anatomy, Histology, and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
- *Correspondence: Hans G. Richter
| |
Collapse
|
20
|
Canaple L, Gréchez-Cassiau A, Delaunay F, Dkhissi-Benyahya O, Samarut J. Maternal eating behavior is a major synchronizer of fetal and postnatal peripheral clocks in mice. Cell Mol Life Sci 2018; 75:3991-4005. [PMID: 29804258 PMCID: PMC11105238 DOI: 10.1007/s00018-018-2845-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 05/14/2018] [Accepted: 05/22/2018] [Indexed: 12/12/2022]
Abstract
Most living organisms show circadian rhythms in physiology and behavior. These oscillations are generated by endogenous circadian clocks, present in virtually all cells where they control key biological processes. To study peripheral clocks in vivo, we developed an original model, the Rev-Luc mouse to follow noninvasively and longitudinally Rev-Luc oscillations in peripheral clocks using in vivo bioluminescence imaging. We found in vitro and in vivo a robust diurnal rhythm of Rev-Luc, mainly in liver, intestine, kidney and adipose tissues. We further confirmed in vivo that Rev-Luc peripheral tissues are food-entrainable oscillators, not affected by age or sex. These data strongly support the relevance of the Rev-Luc model for circadian studies, especially to investigate in vivo the establishment and the entrainment of the rhythm throughout ontogenesis. We then showed that Rev-Luc expression develops dynamically and gradually, both in amplitude and in phase, during fetal and postnatal development. We also demonstrate for the first time that the immature peripheral circadian system of offspring in utero is mainly entrained by maternal cues from feeding regimen. The prenatal entrainment will also differentially determine the Rev-Luc expression in pups before weaning underlining the importance of the maternal chrononutrition on the circadian system entrainment of the offspring.
Collapse
Affiliation(s)
- Laurence Canaple
- Université de Lyon, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon; CNRS UMR 5242, Institut de Génomique Fonctionnelle de Lyon, 46 allée d'Italie, 69364, Lyon, France.
| | - Aline Gréchez-Cassiau
- Université Côte d'Azur, CNRS UMR7277, INSERM U1091, Institut de Biologie Valrose, Bâtiment de Sciences Naturelles, 28 Avenue Valrose, 06108, Nice Cedex 2, France
| | - Franck Delaunay
- Université Côte d'Azur, CNRS UMR7277, INSERM U1091, Institut de Biologie Valrose, Bâtiment de Sciences Naturelles, 28 Avenue Valrose, 06108, Nice Cedex 2, France
| | - Ouria Dkhissi-Benyahya
- Université de Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500, Bron, France
| | - Jacques Samarut
- Université de Lyon, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon; CNRS UMR 5242, Institut de Génomique Fonctionnelle de Lyon, 46 allée d'Italie, 69364, Lyon, France.
| |
Collapse
|
21
|
Sumová A, Čečmanová V. Mystery of rhythmic signal emergence within the suprachiasmatic nuclei. Eur J Neurosci 2018; 51:300-309. [PMID: 30188597 DOI: 10.1111/ejn.14141] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 08/07/2018] [Accepted: 08/28/2018] [Indexed: 12/21/2022]
Abstract
The circadian system provides organisms with a temporal organization that optimizes their adaptation to environmental fluctuations on a 24-hr basis. In mammals, the circadian clock in the suprachiasmatic nuclei (SCN) develops during the perinatal period. The rhythmicity first appears at the level of individual SCN neurons during the fetal stage, and this step is often misinterpreted as the time of complete SCN clock development. However, the process is only finalized when the SCN begin to play a role of the central clock in the body, that is, when they are able to generate robust rhythmicity at the cell population level, entrain the rhythmic signal with external light-dark cycles and convey this signal to the rest of the body. The development is gradual and correlates with morphological maturation of the SCN structural complexity, which is based on intercellular network formation. The aim of this review is to summarize events related to the first emergence of circadian oscillations in the fetal SCN clock. Although a large amount of data on ontogenesis of the circadian system have been accumulated, how exactly the immature SCN converts into a functional central clock has still remained rather elusive. In this review, the hypothesis of how the SCN attains its rhythmicity at the tissue level is discussed in context with the recent advances in the field. For an extensive summary of the complete ontogenetic development of the circadian system, the readers are referred to other previously published reviews.
Collapse
Affiliation(s)
- Alena Sumová
- Department of Neurohumoral Regulations, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Vendula Čečmanová
- Department of Neurohumoral Regulations, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
22
|
Varcoe TJ, Gatford KL, Kennaway DJ. Maternal circadian rhythms and the programming of adult health and disease. Am J Physiol Regul Integr Comp Physiol 2017; 314:R231-R241. [PMID: 29141950 DOI: 10.1152/ajpregu.00248.2017] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The in utero environment is inherently rhythmic, with the fetus subjected to circadian changes in temperature, substrates, and various maternal hormones. Meanwhile, the fetus is developing an endogenous circadian timing system, preparing for life in an external environment where light, food availability, and other environmental factors change predictably and repeatedly every 24 h. In humans, there are many situations that can disrupt circadian rhythms, including shift work, international travel, insomnias, and circadian rhythm disorders (e.g., advanced/delayed sleep phase disorder), with a growing consensus that this chronodisruption can have deleterious consequences for an individual's health and well-being. However, the impact of chronodisruption during pregnancy on the health of both the mother and fetus is not well understood. In this review, we outline circadian timing system ontogeny in mammals and examine emerging research from animal models demonstrating long-term negative implications for progeny health following maternal chronodisruption during pregnancy.
Collapse
Affiliation(s)
- Tamara J Varcoe
- Robinson Research Institute, Adelaide Medical School, University of Adelaide , Adelaide, South Australia , Australia
| | - Kathryn L Gatford
- Robinson Research Institute, Adelaide Medical School, University of Adelaide , Adelaide, South Australia , Australia
| | - David J Kennaway
- Robinson Research Institute, Adelaide Medical School, University of Adelaide , Adelaide, South Australia , Australia
| |
Collapse
|
23
|
Xu J, Wang Y, Xu Y, Zhou C. Clock gene Bmal1 in mice embryo is dispensable for early embryo development but critical for live birth. BIOL RHYTHM RES 2017. [DOI: 10.1080/09291016.2017.1307975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Jian Xu
- Reproductive Medicine Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Reproductive Medicine Center, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yizi Wang
- Reproductive Medicine Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yanwen Xu
- Reproductive Medicine Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Canquan Zhou
- Reproductive Medicine Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
24
|
Mark PJ, Crew RC, Wharfe MD, Waddell BJ. Rhythmic Three-Part Harmony: The Complex Interaction of Maternal, Placental and Fetal Circadian Systems. J Biol Rhythms 2017; 32:534-549. [PMID: 28920512 DOI: 10.1177/0748730417728671] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
From the perspective of circadian biology, mammalian pregnancy presents an unusual biological scenario in which an entire circadian system (i.e., that of the fetus) is embodied within another (i.e., that of the mother). Moreover, both systems are likely to be influenced at their interface by a third player, the placenta. Successful pregnancy requires major adaptations in maternal physiology, many of which involve circadian changes that support the high metabolic demands of the growing fetus. A functional role for maternal circadian adaptations is implied by the effects of circadian disruption, which result in pregnancy complications including higher risks for miscarriage, preterm labor, and low birth weight. Various aspects of fetal physiology lead to circadian variation, at least in late gestation, but it remains unclear what drives this rhythmicity. It likely involves contributions from the maternal environment and possibly from the placenta and the developing intrinsic molecular clocks within fetal tissues. The role of the placenta is of particular significance because it serves not only to relay signals about the external environment (via the mother) but may also exhibit its own circadian rhythmicity. This review considers how the fetus may be influenced by dynamic circadian signals from the mother and the placenta during gestation, and how, in the face of these changing influences, a new fetal circadian system emerges. Particular emphasis is placed on the role of endocrine signals, most notably melatonin and glucocorticoids, as mediators of maternal-fetal circadian interactions, and on the expression of the clock gene in the 3 compartments. Further study is required to understand how the mother, placenta, and fetus interact across pregnancy to optimize circadian adaptations that support adequate growth and development of the fetus and its transition to postnatal life in a circadian environment.
Collapse
Affiliation(s)
- Peter J Mark
- School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Rachael C Crew
- School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Michaela D Wharfe
- School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Brendan J Waddell
- School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
25
|
Involvement of posttranscriptional regulation of Clock in the emergence of circadian clock oscillation during mouse development. Proc Natl Acad Sci U S A 2017; 114:E7479-E7488. [PMID: 28827343 DOI: 10.1073/pnas.1703170114] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Circadian clock oscillation emerges in mouse embryo in the later developmental stages. Although circadian clock development is closely correlated with cellular differentiation, the mechanisms of its emergence during mammalian development are not well understood. Here, we demonstrate an essential role of the posttranscriptional regulation of Clock subsequent to the cellular differentiation for the emergence of circadian clock oscillation in mouse fetal hearts and mouse embryonic stem cells (ESCs). In mouse fetal hearts, no apparent oscillation of cell-autonomous molecular clock was detectable around E10, whereas oscillation was clearly visible in E18 hearts. Temporal RNA-sequencing analysis using mouse fetal hearts reveals many fewer rhythmic genes in E10-12 hearts (63, no core circadian genes) than in E17-19 hearts (483 genes), suggesting the lack of functional circadian transcriptional/translational feedback loops (TTFLs) of core circadian genes in E10 mouse fetal hearts. In both ESCs and E10 embryos, CLOCK protein was absent despite the expression of Clock mRNA, which we showed was due to Dicer/Dgcr8-dependent translational suppression of CLOCK. The CLOCK protein is required for the discernible molecular oscillation in differentiated cells, and the posttranscriptional regulation of Clock plays a role in setting the timing for the emergence of the circadian clock oscillation during mammalian development.
Collapse
|
26
|
Chaix A, Zarrinpar A, Panda S. The circadian coordination of cell biology. J Cell Biol 2017; 215:15-25. [PMID: 27738003 PMCID: PMC5057284 DOI: 10.1083/jcb.201603076] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 09/21/2016] [Indexed: 02/07/2023] Open
Abstract
Chaix et al. review how cells generate circadian oscillations and how circadian clocks control cell biology. Circadian clocks are cell-autonomous timing mechanisms that organize cell functions in a 24-h periodicity. In mammals, the main circadian oscillator consists of transcription–translation feedback loops composed of transcriptional regulators, enzymes, and scaffolds that generate and sustain daily oscillations of their own transcript and protein levels. The clock components and their targets impart rhythmic functions to many gene products through transcriptional, posttranscriptional, translational, and posttranslational mechanisms. This, in turn, temporally coordinates many signaling pathways, metabolic activity, organelles’ structure and functions, as well as the cell cycle and the tissue-specific functions of differentiated cells. When the functions of these circadian oscillators are disrupted by age, environment, or genetic mutation, the temporal coordination of cellular functions is lost, reducing organismal health and fitness.
Collapse
Affiliation(s)
- Amandine Chaix
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Amir Zarrinpar
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037 Division of Gastroenterology, University of California, San Diego, La Jolla, CA 92093
| | - Satchidananda Panda
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037
| |
Collapse
|
27
|
Aoyama S, Shibata S. The Role of Circadian Rhythms in Muscular and Osseous Physiology and Their Regulation by Nutrition and Exercise. Front Neurosci 2017; 11:63. [PMID: 28261043 PMCID: PMC5306200 DOI: 10.3389/fnins.2017.00063] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 01/27/2017] [Indexed: 01/13/2023] Open
Abstract
The mammalian circadian clock regulates the day and night cycles of various physiological functions. The circadian clock system consists of a central clock in the suprachiasmatic nucleus (SCN) of the hypothalamus and peripheral clocks in peripheral tissues. According to the results of circadian transcriptomic studies in several tissues, the majority of rhythmic genes are expressed in a tissue-specific manner and are influenced by tissue-specific circadian rhythms. Here we review the diurnal variations of musculoskeletal functions and discuss the impact of the circadian clock on homeostasis in skeletal muscle and bone. Peripheral clocks are controlled by not only photic stimulation from the central clock in the SCN but also by external cues, such as feeding and exercise. In this review, we discuss the effects of feeding and exercise on the circadian clock and diurnal variation of musculoskeletal functions. We also discuss the therapeutic potential of chrono-nutrition and chrono-exercise on circadian disturbances and the failure of homeostasis in skeletal muscle and bone.
Collapse
Affiliation(s)
- Shinya Aoyama
- Organization for University Research Initiatives, Waseda UniversityTokyo, Japan; Laboratory of Physiology and Pharmacology, School of Advanced Science and Engineering, Waseda UniversityTokyo, Japan
| | - Shigenobu Shibata
- Laboratory of Physiology and Pharmacology, School of Advanced Science and Engineering, Waseda University Tokyo, Japan
| |
Collapse
|
28
|
Schiaffino S, Blaauw B, Dyar KA. The functional significance of the skeletal muscle clock: lessons from Bmal1 knockout models. Skelet Muscle 2016; 6:33. [PMID: 27752300 PMCID: PMC5062818 DOI: 10.1186/s13395-016-0107-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 09/28/2016] [Indexed: 01/11/2023] Open
Abstract
The circadian oscillations of muscle genes are controlled either directly by the intrinsic muscle clock or by extrinsic factors, such as feeding, hormonal signals, or neural influences, which are in turn regulated by the central pacemaker, the suprachiasmatic nucleus of the hypothalamus. A unique feature of circadian rhythms in skeletal muscle is motor neuron-dependent contractile activity, which can affect the oscillation of a number of muscle genes independently of the muscle clock. The role of the intrinsic muscle clock has been investigated using different Bmal1 knockout (KO) models. A comparative analysis of these models reveals that the dramatic muscle wasting and premature aging caused by global conventional KO are not present in muscle-specific Bmal1 KO or in global Bmal1 KO induced in the adult, therefore must reflect the loss of Bmal1 function during development in non-muscle tissues. On the other hand, muscle-specific Bmal1 knockout causes impaired muscle glucose uptake and metabolism, supporting a major role of the muscle clock in anticipating the sleep-to-wake transition, when glucose becomes the predominant fuel for the skeletal muscle.
Collapse
Affiliation(s)
- Stefano Schiaffino
- Venetian Institute of Molecular Medicine (VIMM), Via Orus 2, 35129 Padova, Italy
| | - Bert Blaauw
- Venetian Institute of Molecular Medicine (VIMM), Via Orus 2, 35129 Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Kenneth A. Dyar
- Molecular Endocrinology, Institute for Diabetes and Obesity, Helmholtz Zentrum München, Munich, Germany
| |
Collapse
|
29
|
Bruce KD, Szczepankiewicz D, Sihota KK, Ravindraanandan M, Thomas H, Lillycrop KA, Burdge GC, Hanson MA, Byrne CD, Cagampang FR. Altered cellular redox status, sirtuin abundance and clock gene expression in a mouse model of developmentally primed NASH. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:584-93. [PMID: 27040510 PMCID: PMC4874946 DOI: 10.1016/j.bbalip.2016.03.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 03/07/2016] [Accepted: 03/25/2016] [Indexed: 02/06/2023]
Abstract
Background We have previously shown that high fat (HF) feeding during pregnancy primes the development of non-alcoholic steatohepatits (NASH) in the adult offspring. However, the underlying mechanisms are unclear. Aims Since the endogenous molecular clock can regulate hepatic lipid metabolism, we investigated whether exposure to a HF diet during development could alter hepatic clock gene expression and contribute to NASH onset in later life. Methods Female mice were fed either a control (C, 7% kcal fat) or HF (45% kcal fat) diet. Offspring were fed either a C or HF diet resulting in four offspring groups: C/C, C/HF, HF/C and HF/HF. NAFLD progression, cellular redox status, sirtuin expression (Sirt1, Sirt3), and the expression of core clock genes (Clock, Bmal1, Per2, Cry2) and clock-controlled genes involved in lipid metabolism (Rev-Erbα, Rev-Erbβ, RORα, and Srebp1c) were measured in offspring livers. Results Offspring fed a HF diet developed NAFLD. However HF fed offspring of mothers fed a HF diet developed NASH, coupled with significantly reduced NAD+/NADH (p < 0.05, HF/HF vs C/C), Sirt1 (p < 0.001, HF/HF vs C/C), Sirt3 (p < 0.01, HF/HF vs C/C), perturbed clock gene expression, and elevated expression of genes involved lipid metabolism, such as Srebp1c (p < 0.05, C/HF and HF/HF vs C/C). Conclusion Our results suggest that exposure to excess dietary fat during early and post-natal life increases the susceptibility to develop NASH in adulthood, involving altered cellular redox status, reduced sirtuin abundance, and desynchronized clock gene expression. Offspring of mothers fed a high fat diet show severe fatty liver in later life. HF feeding is associated with altered cellular redox status and reduced sirtuin gene expression. HF feeding desynchronises the expression of core clock genes and lipogenic transcription factors. Exposure to a HF diet during development causes changes in liver metabolism that precede severe fatty liver disease.
Collapse
Affiliation(s)
- Kimberley D Bruce
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK; University of Colorado Anschutz Medical Campus, Endocrinology, Metabolism and Diabetes, Aurora, USA.
| | - Dawid Szczepankiewicz
- Poznań University of Life Sciences, Department of Animal Physiology and Biochemistry, Poznań, Poland
| | - Kiran K Sihota
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Manoj Ravindraanandan
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Hugh Thomas
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Karen A Lillycrop
- Centre for Biological Sciences, Institute of Developmental Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, Southampton, UK
| | - Graham C Burdge
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Mark A Hanson
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Christopher D Byrne
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Felino R Cagampang
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| |
Collapse
|
30
|
Yang G, Chen L, Grant GR, Paschos G, Song WL, Musiek ES, Lee V, McLoughlin SC, Grosser T, Cotsarelis G, FitzGerald GA. Timing of expression of the core clock gene Bmal1 influences its effects on aging and survival. Sci Transl Med 2016; 8:324ra16. [PMID: 26843191 PMCID: PMC4870001 DOI: 10.1126/scitranslmed.aad3305] [Citation(s) in RCA: 230] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 12/29/2015] [Indexed: 12/11/2022]
Abstract
The absence of Bmal1, a core clock gene, results in a loss of circadian rhythms, an acceleration of aging, and a shortened life span in mice. To address the importance of circadian rhythms in the aging process, we generated conditional Bmal1 knockout mice that lacked the BMAL1 protein during adult life and found that wild-type circadian variations in wheel-running activity, heart rate, and blood pressure were abolished. Ocular abnormalities and brain astrogliosis were conserved irrespective of the timing of Bmal1 deletion. However, life span, fertility, body weight, blood glucose levels, and age-dependent arthropathy, which are altered in standard Bmal1 knockout mice, remained unaltered, whereas atherosclerosis and hair growth improved, in the conditional adult-life Bmal1 knockout mice, despite abolition of clock function. Hepatic RNA-Seq revealed that expression of oscillatory genes was dampened in the adult-life Bmal1 knockout mice, whereas overall gene expression was largely unchanged. Thus, many phenotypes in conventional Bmal1 knockout mice, hitherto attributed to disruption of circadian rhythms, reflect the loss of properties of BMAL1 that are independent of its role in the clock. These findings prompt reevaluation of the systemic consequences of disruption of the molecular clock.
Collapse
Affiliation(s)
- Guangrui Yang
- The Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lihong Chen
- The Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gregory R Grant
- The Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA. Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Georgios Paschos
- The Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wen-Liang Song
- The Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Erik S Musiek
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Vivian Lee
- Department of Ophthalmology, University of Pennsylvania Scheie Eye Institute, Philadelphia, PA 19104, USA
| | - Sarah C McLoughlin
- The Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tilo Grosser
- The Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - George Cotsarelis
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Garret A FitzGerald
- The Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
31
|
|
32
|
Abstract
Since the kidney is integral to maintenance of fluid and ion homeostasis, and therefore blood pressure regulation, its proper function is paramount. Circadian fluctuations in blood pressure, renal blood flow, glomerular filtration rate, and sodium and water excretion have been documented for decades, if not longer. Recent studies on the role of circadian clock proteins in the regulation of a variety of renal transport genes suggest that the molecular clock in the kidney controls circadian fluctuations in renal function. The circadian clock appears to be a critical regulator of renal function with important implications for the treatment of renal pathologies, which include chronic kidney disease and hypertension. The development, regulation, and mechanism of the kidney clock are reviewed here.
Collapse
Affiliation(s)
- Kristen Solocinski
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, FloridaDepartment of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida
| | - Michelle L Gumz
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, FloridaDepartment of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida
| |
Collapse
|
33
|
Mouralidarane A, Soeda J, Sugden D, Bocianowska A, Carter R, Ray S, Saraswati R, Cordero P, Novelli M, Fusai G, Vinciguerra M, Poston L, Taylor PD, Oben JA. Maternal obesity programs offspring non-alcoholic fatty liver disease through disruption of 24-h rhythms in mice. Int J Obes (Lond) 2015; 39:1339-48. [PMID: 25971926 DOI: 10.1038/ijo.2015.85] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Revised: 03/25/2015] [Accepted: 05/06/2015] [Indexed: 02/08/2023]
Abstract
BACKGROUND Maternal obesity increases offspring propensity to metabolic dysfunctions and to non-alcoholic fatty liver disease (NAFLD), which may lead to cirrhosis or liver cancer. The circadian clock is a transcriptional/epigenetic molecular machinery synchronising physiological processes to coordinate energy utilisation within a 24-h light/dark period. Alterations in rhythmicity have profound effects on metabolic pathways, which we sought to investigate in offspring with programmed NAFLD. METHODS Mice were fed a standard or an obesogenic diet (OD), before and throughout pregnancy, and during lactation. Offspring were weaned onto standard or an OD at 3 weeks postpartum and housed in 12:12 light/dark conditions. Biochemical and histological indicators of NAFLD and fibrosis, analysis of canonical clock genes with methylation status and locomotor activity were investigated at 6 months. RESULTS We show that maternal obesity interacts with an obesogenic post-weaning diet to promote the development of NAFLD with disruption of canonical metabolic rhythmicity gene expression in the liver. We demonstrate hypermethylation of BMAL-1 (brain and muscle Arnt like-1) and Per2 promoter regions and altered 24-h rhythmicity of hepatic pro-inflammatory and fibrogenic mediators. CONCLUSIONS These data implicate disordered circadian rhythms in NAFLD and suggest that disruption of this system during critical developmental periods may be responsible for the onset of chronic liver disease in adulthood.
Collapse
Affiliation(s)
- A Mouralidarane
- Institute for Liver and Digestive Health, University College London, Royal Free Hospital, London, UK
- Women's Health Academic Centre, King's College London, St Thomas' Hospital, London, UK
| | - J Soeda
- Institute for Liver and Digestive Health, University College London, Royal Free Hospital, London, UK
| | - D Sugden
- Women's Health Academic Centre, King's College London, St Thomas' Hospital, London, UK
| | - A Bocianowska
- Women's Health Academic Centre, King's College London, St Thomas' Hospital, London, UK
| | - R Carter
- Institute for Liver and Digestive Health, University College London, Royal Free Hospital, London, UK
| | - S Ray
- Institute for Liver and Digestive Health, University College London, Royal Free Hospital, London, UK
| | - R Saraswati
- Histopathology Department, University College Hospital, University College London, London, UK
| | - P Cordero
- Institute for Liver and Digestive Health, University College London, Royal Free Hospital, London, UK
| | - M Novelli
- Histopathology Department, University College Hospital, University College London, London, UK
| | - G Fusai
- Department of Liver Medicine and Transplant, Sheila Sherlock Liver Centre, University College London, Royal Free Hospital, London, UK
| | - M Vinciguerra
- Institute for Liver and Digestive Health, University College London, Royal Free Hospital, London, UK
- Gastroenterology Unit, Casa Sollievo della Sofferenza Hospital, San Giovanni Rotondo, Italy
- School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - L Poston
- Women's Health Academic Centre, King's College London, St Thomas' Hospital, London, UK
| | - P D Taylor
- Women's Health Academic Centre, King's College London, St Thomas' Hospital, London, UK
| | - J A Oben
- Institute for Liver and Digestive Health, University College London, Royal Free Hospital, London, UK
| |
Collapse
|
34
|
Wreschnig D, Dolatshad H, Davis FC. Embryonic development of circadian oscillations in the mouse hypothalamus. J Biol Rhythms 2015; 29:299-310. [PMID: 25238858 DOI: 10.1177/0748730414545086] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Circadian rhythms in mammals are regulated by the hypothalamic suprachiasmatic nucleus (SCN). The generation of circadian oscillations is a cell-autonomous property, and coupling among cells is essential for the SCN to function as a pacemaker. The development of SCN anatomy and cytology has been extensively studied, but the point in development when the SCN first has the capacity to generate circadian oscillations has not been established. We therefore examined the development of circadian oscillations using per2::luc mice in which bioluminescence tracks the expression of the circadian clock protein, PER2. In vitro, hypothalamic explants first expressed consistent oscillations when isolated between 15 and 16 days postfertilization (e15). Oscillations were more robust at later ages. Explants from other brain areas did not express oscillations, indicating that the development of oscillations is not a general property of embryonic tissue. SCN explants obtained on e14 did not initially express oscillations but developed them in vitro over 4 to 6 d. Although coupling among cells is required for the long-term expression of tissue-level oscillations, explants from mice lacking the coupling peptide vasoactive intestinal peptide still developed oscillations. In the mouse, the capacity to generate molecular oscillations on e15 coincides with the completion of neurogenesis and SCN-specific transcription factor expression. Thus, within a day of its genesis at an age approximately equivalent to the end of the first trimester in humans, the SCN develops the capacity to express circadian oscillations and autonomously develops mechanisms sufficient to couple and synchronize its cells.
Collapse
Affiliation(s)
- Daniel Wreschnig
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | - Hamid Dolatshad
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | - Fred C Davis
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
35
|
Olejníková L, Polidarová L, Paušlyová L, Sládek M, Sumová A. Diverse development and higher sensitivity of the circadian clocks to changes in maternal-feeding regime in a rat model of cardio-metabolic disease. Chronobiol Int 2015; 32:531-47. [DOI: 10.3109/07420528.2015.1014095] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
36
|
Landgraf D, Achten C, Dallmann F, Oster H. Embryonic development and maternal regulation of murine circadian clock function. Chronobiol Int 2014; 32:416-27. [DOI: 10.3109/07420528.2014.986576] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
37
|
Mészáros K, Pruess L, Szabó AJ, Gondan M, Ritz E, Schaefer F. Development of the circadian clockwork in the kidney. Kidney Int 2014; 86:915-22. [DOI: 10.1038/ki.2014.199] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 04/03/2014] [Accepted: 04/10/2014] [Indexed: 11/09/2022]
|
38
|
Du Pré BC, Van Veen TAB, Young ME, Vos MA, Doevendans PA, Van Laake LW. Circadian rhythms in cell maturation. Physiology (Bethesda) 2014; 29:72-83. [PMID: 24382873 DOI: 10.1152/physiol.00036.2013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Circadian rhythms are of major importance in mammalian physiology and disease. In this review, we give an overview of the present knowledge on origination of circadian rhythms. We discuss the development of both master and peripheral clocks and compare the origination of circadian rhythms in utero and in vitro.
Collapse
Affiliation(s)
- Bastiaan C Du Pré
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | |
Collapse
|
39
|
Curran KL, Allen L, Porter BB, Dodge J, Lope C, Willadsen G, Fisher R, Johnson N, Campbell E, VonBergen B, Winfrey D, Hadley M, Kerndt T. Circadian genes, xBmal1 and xNocturnin, modulate the timing and differentiation of somites in Xenopus laevis. PLoS One 2014; 9:e108266. [PMID: 25238599 PMCID: PMC4169625 DOI: 10.1371/journal.pone.0108266] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 08/20/2014] [Indexed: 02/06/2023] Open
Abstract
We have been investigating whether xBmal1 and xNocturnin play a role in somitogenesis, a cyclic developmental process with an ultradian period. Previous work from our lab shows that circadian genes (xPeriod1, xPeriod2, xBmal1, and xNocturnin) are expressed in developing somites. Somites eventually form the vertebrae, muscles of the back, and dermis. In Xenopus, a pair of somites is formed about every 50 minutes from anterior to posterior. We were intrigued by the co-localization of circadian genes in an embryonic tissue known to be regulated by an ultradian clock. Cyclic expression of genes involved in Notch signaling has been implicated in the somite clock. Disruption of Notch signaling in humans has been linked to skeletal defects in the vertebral column. We found that both depletion (morpholino) and overexpression (mRNA) of xBMAL1 protein (bHLH transcription factor) or xNOCTURNIN protein (deadenylase) on one side of the developing embryo led to a significant decrease in somite number with respect to the untreated side (p<0.001). These manipulations also significantly affect expression of a somite clock component (xESR9; p<0.05). We observed opposing effects on somite size. Depletion of xBMAL1 or xNOCTURNIN caused a statistically significant decrease in somite area (quantified using NIH ImageJ; p<0.002), while overexpression of these proteins caused a significant dose dependent increase in somite area (p<0.02; p<0.001, respectively). We speculate that circadian genes may play two separate roles during somitogenesis. Depletion and overexpression of xBMAL1 and NOCTURNIN both decrease somite number and influence expression of a somite clock component, suggesting that these proteins may modulate the timing of the somite clock in the undifferentiated presomitic mesoderm. The dosage dependent effects on somite area suggest that xBMAL1 and xNOCTURNIN may also act during somite differentiation to promote myogenesis.
Collapse
Affiliation(s)
- Kristen L. Curran
- University of Wisconsin-Whitewater, Department of Biological Sciences, Whitewater, Wisconsin, United States of America
| | - Latoya Allen
- University of Wisconsin-Whitewater, Department of Biological Sciences, Whitewater, Wisconsin, United States of America
| | - Brittany Bronson Porter
- University of Wisconsin-Whitewater, Department of Biological Sciences, Whitewater, Wisconsin, United States of America
| | - Joseph Dodge
- University of Wisconsin-Whitewater, Department of Biological Sciences, Whitewater, Wisconsin, United States of America
| | - Chelsea Lope
- University of Wisconsin-Whitewater, Department of Biological Sciences, Whitewater, Wisconsin, United States of America
| | - Gail Willadsen
- University of Wisconsin-Whitewater, Department of Biological Sciences, Whitewater, Wisconsin, United States of America
| | - Rachel Fisher
- University of Wisconsin-Whitewater, Department of Biological Sciences, Whitewater, Wisconsin, United States of America
| | - Nicole Johnson
- University of Wisconsin-Whitewater, Department of Biological Sciences, Whitewater, Wisconsin, United States of America
| | - Elizabeth Campbell
- University of Wisconsin-Whitewater, Department of Biological Sciences, Whitewater, Wisconsin, United States of America
| | - Brett VonBergen
- University of Wisconsin-Whitewater, Department of Biological Sciences, Whitewater, Wisconsin, United States of America
| | - Devon Winfrey
- University of Wisconsin-Whitewater, Department of Biological Sciences, Whitewater, Wisconsin, United States of America
| | - Morgan Hadley
- University of Wisconsin-Whitewater, Department of Biological Sciences, Whitewater, Wisconsin, United States of America
| | - Thomas Kerndt
- University of Wisconsin-Whitewater, Department of Biological Sciences, Whitewater, Wisconsin, United States of America
| |
Collapse
|
40
|
Gonsalvez DG, Li-Yuen-Fong M, Cane KN, Stamp LA, Young HM, Anderson CR. Different neural crest populations exhibit diverse proliferative behaviors. Dev Neurobiol 2014; 75:287-301. [DOI: 10.1002/dneu.22229] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 08/16/2014] [Accepted: 09/02/2014] [Indexed: 01/02/2023]
Affiliation(s)
- David G. Gonsalvez
- Department of Anatomy and Neuroscience; University of Melbourne; Victoria 3010 Australia
| | - Mathew Li-Yuen-Fong
- Department of Anatomy and Neuroscience; University of Melbourne; Victoria 3010 Australia
| | - Kylie N. Cane
- Department of Anatomy and Neuroscience; University of Melbourne; Victoria 3010 Australia
| | - Lincon A. Stamp
- Department of Anatomy and Neuroscience; University of Melbourne; Victoria 3010 Australia
| | - Heather M. Young
- Department of Anatomy and Neuroscience; University of Melbourne; Victoria 3010 Australia
| | - Colin R. Anderson
- Department of Anatomy and Neuroscience; University of Melbourne; Victoria 3010 Australia
| |
Collapse
|
41
|
Brown SA. Circadian clock-mediated control of stem cell division and differentiation: beyond night and day. Development 2014; 141:3105-11. [DOI: 10.1242/dev.104851] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
A biological ‘circadian’ clock conveys diurnal regulation upon nearly all aspects of behavior and physiology to optimize them within the framework of the solar day. From digestion to cardiac function and sleep, both cellular and systemic processes show circadian variations that coincide with diurnal need. However, recent research has shown that this same timekeeping mechanism might have been co-opted to optimize other aspects of development and physiology that have no obvious link to the 24 h day. For example, clocks have been suggested to underlie heterogeneity in stem cell populations, to optimize cycles of cell division during wound healing, and to alter immune progenitor differentiation and migration. Here, I review these circadian mechanisms and propose that they could serve as metronomes for a surprising variety of physiologically and medically important functions that far exceed the daily timekeeping roles for which they probably evolved.
Collapse
Affiliation(s)
- Steven A. Brown
- Institute of Pharmacology and Toxicology, University of Zürich, 190 Winterthurerstrasse, Zürich 8057, Switzerland
| |
Collapse
|
42
|
Polidarová L, Olejníková L, Paušlyová L, Sládek M, Soták M, Pácha J, Sumová A. Development and entrainment of the colonic circadian clock during ontogenesis. Am J Physiol Gastrointest Liver Physiol 2014; 306:G346-56. [PMID: 24337008 DOI: 10.1152/ajpgi.00340.2013] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Colonic morphology and function change significantly during ontogenesis. In mammals, many colonic physiological functions are temporally controlled by the circadian clock in the colon, which is entrained by the central circadian clock in the suprachiasmatic nuclei (SCN). The aim of this present study was to ascertain when and how the circadian clock in the colon develops during the perinatal period and whether maternal cues and/or the developing pup SCN may influence the ontogenesis of the colonic clock. Daily profiles of clock genes Per1, Per2, Cry1, Cry2, Rev-erbα, Bmal1, and Clock expression in the colon underwent significant modifications since embryonic day 20 (E20) through postnatal days (P) 2, 10, 20, and 30 via changes in the mutual phasing among the individual clock gene expression rhythms, their relative phasing to the light-dark regime, and their amplitudes. An adult-like state was achieved around P20. The foster study revealed that during the prenatal period, the maternal circadian phase may partially modulate development of the colonic clock. Postnatally, the absence and/or presence of rhythmic maternal care affected the phasing of the clock gene expression profiles in pups at P10 and P20. A reversal in the colonic clock phase between P10 and P20 occurred in the absence of rhythmic signals from the pup SCN. The data demonstrate ontogenetic maturation of the colonic clock and stress the importance of prenatal and postnatal maternal rhythmic signals for its development. These data may contribute to the understanding of colonic function-related diseases in newborn children.
Collapse
Affiliation(s)
- Lenka Polidarová
- Department of Neurohumoral Regulations, Institute of Physiology, Academy of Science of the Czech Republic, Prague, Czech Republic; and
| | | | | | | | | | | | | |
Collapse
|
43
|
Cell and tissue-autonomous development of the circadian clock in mouse embryos. FEBS Lett 2013; 588:459-65. [DOI: 10.1016/j.febslet.2013.12.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Revised: 12/10/2013] [Accepted: 12/10/2013] [Indexed: 11/23/2022]
|
44
|
O’Neil D, Mendez-Figueroa H, Mistretta TA, Su C, Lane RH, Aagaard KM. Dysregulation of Npas2 leads to altered metabolic pathways in a murine knockout model. Mol Genet Metab 2013; 110:378-87. [PMID: 24067359 PMCID: PMC3874417 DOI: 10.1016/j.ymgme.2013.08.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 08/27/2013] [Indexed: 12/15/2022]
Abstract
In our primate model of maternal high fat diet exposure, we have described that fetal epigenomic modifications to the peripheral circadian Npas2 are associated with persistent alterations in fetal hepatic metabolism and non-alcoholic fatty liver. As the interaction of circadian response with metabolism is not well understood, we employed a murine knockout model to characterize the molecular mechanisms with which Npas2 reprograms the fetal hepatic metabolic response. cDNA was generated from Npas2-/- and +/+ (wild type) livers at day 2 (newborn) and at 25 weeks (adult) of life. Newborn samples were analyzed by exon array (n = 3/cohort). Independent pathway analysis software determined that the primary dysregulated pathway(s) in the Npas2-/- animals uniformly converged on lipid metabolism. Of particular interest, Ppargc1a, which integrates circadian and metabolism pathways, was significantly (p < .01) over expressed in newborn (1.7 fold) and adult (1.8 fold) Npas2-/- animals. These findings are consistent with an essential role for Npas2 in programming the peripheral circadian response and hepatic metabolism, which has not been previously described.
Collapse
Affiliation(s)
- Derek O’Neil
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine; Baylor College of Medicine; Houston, TX, 77030; USA
- Translational Biology and Molecular Medicine Program; Baylor College of Medicine; Houston, TX, 77030; USA
| | - Hector Mendez-Figueroa
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine; Baylor College of Medicine; Houston, TX, 77030; USA
| | - Toni-Ann Mistretta
- Department of Pathology; Texas Children’s Hospital, Baylor College of Medicine; Houston, TX, 77030; USA
| | - Chunliu Su
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine; Baylor College of Medicine; Houston, TX, 77030; USA
| | - Robert H. Lane
- Department of Pediatrics; University of Utah; Salt Lake City, UT, 84112; USA
| | - Kjersti M. Aagaard
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine; Baylor College of Medicine; Houston, TX, 77030; USA
- Translational Biology and Molecular Medicine Program; Baylor College of Medicine; Houston, TX, 77030; USA
| |
Collapse
|
45
|
Brooks E, Canal MM. Development of circadian rhythms: role of postnatal light environment. Neurosci Biobehav Rev 2013; 37:551-60. [PMID: 23454636 DOI: 10.1016/j.neubiorev.2013.02.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 01/31/2013] [Accepted: 02/12/2013] [Indexed: 10/27/2022]
Abstract
Mammals are born with an immature circadian system, which completes its development postnatally. Evidence suggests that the environment experienced by a newborn will impact and shape its development, which will have future consequences at the levels of circadian system function, circadian behaviour and physiology, and potentially, the animal's long-term health and welfare. Here we review the various stages in postnatal development of the circadian system, and discuss the data available on the long-term effects of early environment, in particular light environment, on the animal's brain, physiology and behaviour.
Collapse
Affiliation(s)
- Elisabeth Brooks
- University of Manchester, Faculty of Life Sciences, AV Hill Building, Oxford Road, Manchester M13 9PT, UK
| | | |
Collapse
|
46
|
Boden MJ, Varcoe TJ, Kennaway DJ. Circadian regulation of reproduction: from gamete to offspring. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2013; 113:387-97. [PMID: 23380455 DOI: 10.1016/j.pbiomolbio.2013.01.003] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 12/10/2012] [Accepted: 01/22/2013] [Indexed: 01/19/2023]
Abstract
Few challenges are more critical to the survival of a species than reproduction. To ensure reproductive success, myriad aspects of physiology and behaviour need to be tightly orchestrated within the animal, as well as timed appropriately with the external environment. This is accomplished through an endogenous circadian timing system generated at the cellular level through a series of interlocked transcription/translation feedback loops, leading to the overt expression of circadian rhythms. These expression patterns are found throughout the body, and are intimately interwoven with both the timing and function of the reproductive process. In this review we highlight the many aspects of reproductive physiology in which circadian rhythms are known to play a role, including regulation of the estrus cycle, the LH surge and ovulation, the production and maturation of sperm and the timing of insemination and fertilisation. We will also describe roles for circadian rhythms in support of the preimplantation embryo in the oviduct, implantation/placentation, as well as the control of parturition and early postnatal life. There are several key differences in physiology between humans and the model systems used for the study of circadian disruption, and these challenges to interpretation will be discussed as part of this review.
Collapse
Affiliation(s)
- M J Boden
- Robinson Institute, Research Centre for Reproductive Health, Department of Obstetrics and Gynaecology, University of Adelaide, Medical School, Adelaide, SA 5005, Australia.
| | | | | |
Collapse
|
47
|
Guilding C, Scott F, Bechtold DA, Brown TM, Wegner S, Piggins HD. Suppressed cellular oscillations in after-hours mutant mice are associated with enhanced circadian phase-resetting. J Physiol 2012. [PMID: 23207594 PMCID: PMC3591715 DOI: 10.1113/jphysiol.2012.242198] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Within the core molecular clock, protein phosphorylation and degradation play a vital role in determining circadian period. The 'after-hours' (Afh) mutation in mouse slows the degradation of the core clock protein Cryptochrome, lengthening the period of the molecular clock in the suprachiasmatic nuclei (SCN) and behavioural wheel-running rhythms. However, we do not yet know how the Afh mutation affects other aspects of physiology or the activity of circadian oscillators in other brain regions. Here we report that daily rhythms of metabolism and ingestive behaviours are altered in these animals, as are PERIOD2::LUCIFERASE (PER2::LUC) rhythms in mediobasal hypothalamic nuclei, which influence these behaviours. Overall there is a trend towards period lengthening and a decrease in amplitude of PER2::LUC rhythms throughout the brain. Imaging of single cells from the arcuate and dorsomedial hypothalamic nuclei revealed this reduction in tissue oscillator amplitude to be due to a decrease in the amplitude, rather than a desynchrony, of single cells. Consistent with existing models of oscillator function, this cellular phenotype was associated with a greater susceptibility to phase-shifting stimuli in vivo and in vitro, with light evoking high-amplitude Type 0 resetting in Afh mutant mice. Together, these findings reveal unexpected consequences of the Afh mutation on the amplitude and synchrony of individual cellular oscillators in the SCN.
Collapse
Affiliation(s)
- Clare Guilding
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | | | | | | | | | | |
Collapse
|
48
|
Wu G, Zhu J, He F, Wang W, Hu S, Yu J. Gene and genome parameters of mammalian liver circadian genes (LCGs). PLoS One 2012; 7:e46961. [PMID: 23071677 PMCID: PMC3468600 DOI: 10.1371/journal.pone.0046961] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 09/07/2012] [Indexed: 11/19/2022] Open
Abstract
The mammalian circadian system controls various physiology processes and behavior responses by regulating thousands of circadian genes with rhythmic expressions. In this study, we redefined circadian-regulated genes based on published results in the mouse liver and compared them with other gene groups defined relative to circadian regulations, especially the non-circadian-regulated genes expressed in liver at multiple molecular levels from gene position to protein expression based on integrative analyses of different datasets from the literature. Based on the intra-tissue analysis, the liver circadian genes or LCGs show unique features when compared to other gene groups. First, LCGs in general have less neighboring genes and larger in both genomic and 3'-UTR lengths but shorter in CDS (coding sequence) lengths. Second, LCGs have higher mRNA and protein abundance, higher temporal expression variations, and shorter mRNA half-life. Third, more than 60% of LCGs form major co-expression clusters centered in four temporal windows: dawn, day, dusk, and night. In addition, larger and smaller LCGs are found mainly expressed in the day and night temporal windows, respectively, and we believe that LCGs are well-partitioned into the gene expression regulatory network that takes advantage of gene size, expression constraint, and chromosomal architecture. Based on inter-tissue analysis, more than half of LCGs are ubiquitously expressed in multiple tissues but only show rhythmical expression in one or limited number of tissues. LCGs show at least three-fold lower expression variations across the temporal windows than those among different tissues, and this observation suggests that temporal expression variations regulated by the circadian system is relatively subtle as compared with the tissue expression variations formed during development. Taken together, we suggest that the circadian system selects gene parameters in a cost effective way to improve tissue-specific functions by adapting temporal variations from the environment over evolutionary time scales.
Collapse
Affiliation(s)
- Gang Wu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- Graduate University of Chinese Academy of Sciences, Beijing, China
| | - Jiang Zhu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Fuhong He
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- Laboratory of Disease Genomics and Individualized Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Weiwei Wang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Songnian Hu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Jun Yu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- * E-mail:
| |
Collapse
|
49
|
Waddell B, Wharfe M, Crew R, Mark P. A rhythmic placenta? Circadian variation, clock genes and placental function. Placenta 2012; 33:533-9. [DOI: 10.1016/j.placenta.2012.03.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 03/22/2012] [Indexed: 12/19/2022]
|
50
|
Epigenetic Control of Circadian Clock Operation during Development. GENETICS RESEARCH INTERNATIONAL 2012; 2012:845429. [PMID: 22567402 PMCID: PMC3335631 DOI: 10.1155/2012/845429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Revised: 12/22/2011] [Accepted: 01/13/2012] [Indexed: 11/17/2022]
Abstract
The molecular players of circadian clock oscillation have been identified and extensively characterized. The epigenetic mechanisms behind the circadian gene expression control has also been recently studied, although there are still details to be illucidated. In this review, we briefly summarize the current understanding of the mammalian clock. We also provide evidence for the lack of circadian oscillation in particular cell types. As the circadian clock has intimate interaction with the various cellular functions in different type of cells, it must have plasticity and specicity in its operation within different epigenetic environments. The lack of circadian oscillation in certain cells provide an unique opportunity to study the required epigenetic environment in the cell that permit circadian oscillation and to idenfify key influencing factors for proper clock function. How epigenetic mechansims, including DNA methylaiton and chromatin modifications, participate in control of clock oscillation still awaits future studies at the genomic scale.
Collapse
|