1
|
Sharma SA, Oladejo SO, Kuang Z. Chemical interplay between gut microbiota and epigenetics: Implications in circadian biology. Cell Chem Biol 2025; 32:61-82. [PMID: 38776923 PMCID: PMC11569273 DOI: 10.1016/j.chembiol.2024.04.016] [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: 12/15/2023] [Revised: 03/22/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024]
Abstract
Circadian rhythms are intrinsic molecular mechanisms that synchronize biological functions with the day/night cycle. The mammalian gut is colonized by a myriad of microbes, collectively named the gut microbiota. The microbiota impacts host physiology via metabolites and structural components. A key mechanism is the modulation of host epigenetic pathways, especially histone modifications. An increasing number of studies indicate the role of the microbiota in regulating host circadian rhythms. However, the mechanisms remain largely unknown. Here, we summarize studies on microbial regulation of host circadian rhythms and epigenetic pathways, highlight recent findings on how the microbiota employs host epigenetic machinery to regulate circadian rhythms, and discuss its impacts on host physiology, particularly immune and metabolic functions. We further describe current challenges and resources that could facilitate research on microbiota-epigenetic-circadian rhythm interactions to advance our knowledge of circadian disorders and possible therapeutic avenues.
Collapse
Affiliation(s)
- Samskrathi Aravinda Sharma
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15213, USA
| | - Sarah Olanrewaju Oladejo
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15213, USA
| | - Zheng Kuang
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15213, USA.
| |
Collapse
|
2
|
Ragozzino FJ, Karatsoreos IN, Peters JH. Principles of synaptic encoding of brainstem circadian rhythms. Exp Physiol 2024; 109:2006-2010. [PMID: 38308846 PMCID: PMC11607608 DOI: 10.1113/ep090867] [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: 10/15/2023] [Accepted: 01/16/2024] [Indexed: 02/05/2024]
Abstract
Circadian regulation of autonomic tone and reflex pathways pairs physiological processes with the daily light cycle. However, the underlying mechanisms mediating these changes on autonomic neurocircuitry are only beginning to be understood. The brainstem nucleus of the solitary tract (NTS) and adjacent nuclei, including the area postrema and dorsal motor nucleus of the vagus, are key candidates for rhythmic control of some aspects of the autonomic nervous system. Recent findings have contributed to a working model of circadian regulation in the brainstem which manifests from the transcriptional, to synaptic, to circuit levels of organization. Vagal afferent neurons and the NTS possess rhythmic clock gene expression, rhythmic action potential firing, and our recent findings demonstrate rhythmic spontaneous glutamate release. In addition, postsynaptic conductances also vary across the day producing subtle changes in membrane depolarization which govern synaptic efficacy. Together these coordinated pre- and postsynaptic changes provide nuanced control of synaptic transmission across the day to tune the sensitivity of primary afferent input and likely govern reflex output. Further, given the important role for the brainstem in integrating cues such as feeding, cardiovascular function and temperature, it may also be an underappreciated locus in mediating the effects of such non-photic entraining cues. This short review focuses on the neurophysiological principles that govern NTS synaptic transmission and how circadian rhythms impacted them across the day.
Collapse
Affiliation(s)
- Forrest J. Ragozzino
- Department of Integrative Physiology and Neuroscience, College of Veterinary MedicineWashington State UniversityPullmanWashingtonUSA
| | - Ilia N. Karatsoreos
- Department of Psychological and Brain SciencesUniversity of Massachusetts AmherstAmherstMassachusettsUSA
| | - James H. Peters
- Department of Integrative Physiology and Neuroscience, College of Veterinary MedicineWashington State UniversityPullmanWashingtonUSA
| |
Collapse
|
3
|
Rao F, Xue T. Circadian-independent light regulation of mammalian metabolism. Nat Metab 2024; 6:1000-1007. [PMID: 38831000 DOI: 10.1038/s42255-024-01051-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 04/16/2024] [Indexed: 06/05/2024]
Abstract
The daily light-dark cycle is a key zeitgeber (time cue) for entraining an organism's biological clock, whereby light sensing by retinal photoreceptors, particularly intrinsically photosensitive retinal ganglion cells, stimulates the suprachiasmatic nucleus of the hypothalamus, a central pacemaker that in turn orchestrates the rhythm of peripheral metabolic activities. Non-rhythmic effects of light on metabolism have also been long known, and their transduction mechanisms are only beginning to unfold. Here, we summarize emerging evidence that, in mammals, light exposure or deprivation profoundly affects glucose homeostasis, thermogenesis and other metabolic activities in a clock-independent manner. Such light regulation could involve melanopsin-based, intrinsically photosensitive retinal ganglion cell-initiated brain circuits via the suprachiasmatic nucleus of the hypothalamus and other nuclei, or direct stimulation of opsins expressed in the hypothalamus, adipose tissue, blood vessels and skin to regulate sympathetic tone, lipolysis, glucose uptake, mitochondrial activation, thermogenesis, food intake, blood pressure and melanogenesis. These photic signalling events may coordinate with circadian-based mechanisms to maintain metabolic homeostasis, with dysregulation of this system underlying metabolic diseases caused by aberrant light exposure, such as environmental night light and shift work.
Collapse
Affiliation(s)
- Feng Rao
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
| | - Tian Xue
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
4
|
Han C, Lim JY, Koike N, Kim SY, Ono K, Tran CK, Mangutov E, Kim E, Zhang Y, Li L, Pradhan AA, Yagita K, Chen Z, Yoo SH, Burish MJ. Regulation of headache response and transcriptomic network by the trigeminal ganglion clock. Headache 2024; 64:195-210. [PMID: 38288634 PMCID: PMC10961824 DOI: 10.1111/head.14670] [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: 07/02/2023] [Revised: 12/06/2023] [Accepted: 12/20/2023] [Indexed: 02/17/2024]
Abstract
OBJECTIVE To characterize the circadian features of the trigeminal ganglion in a mouse model of headache. BACKGROUND Several headache disorders, such as migraine and cluster headache, are known to exhibit distinct circadian rhythms of attacks. The circadian basis for these rhythmic pain responses, however, remains poorly understood. METHODS We examined trigeminal ganglion ex vivo and single-cell cultures from Per2::LucSV reporter mice and performed immunohistochemistry. Circadian behavior and transcriptomics were investigated using a novel combination of trigeminovascular and circadian models: a nitroglycerin mouse headache model with mechanical thresholds measured every 6 h, and trigeminal ganglion RNA sequencing measured every 4 h for 24 h. Finally, we performed pharmacogenomic analysis of gene targets for migraine, cluster headache, and trigeminal neuralgia treatments as well as trigeminal ganglion neuropeptides; this information was cross-referenced with our cycling genes from RNA sequencing data to identify potential targets for chronotherapy. RESULTS The trigeminal ganglion demonstrates strong circadian rhythms in both ex vivo and single-cell cultures, with core circadian proteins found in both neuronal and non-neuronal cells. Using our novel behavioral model, we showed that nitroglycerin-treated mice display circadian rhythms of pain sensitivity which were abolished in arrhythmic Per1/2 double knockout mice. Furthermore, RNA-sequencing analysis of the trigeminal ganglion revealed 466 genes that displayed circadian oscillations in the control group, including core clock genes and clock-regulated pain neurotransmitters. In the nitroglycerin group, we observed a profound circadian reprogramming of gene expression, as 331 of circadian genes in the control group lost rhythm and another 584 genes gained rhythm. Finally, pharmacogenetics analysis identified 10 genes in our trigeminal ganglion circadian transcriptome that encode target proteins of current medications used to treat migraine, cluster headache, or trigeminal neuralgia. CONCLUSION Our study unveiled robust circadian rhythms in the trigeminal ganglion at the behavioral, transcriptomic, and pharmacogenetic levels. These results support a fundamental role of the clock in pain pathophysiology. PLAIN LANGUAGE SUMMARY Several headache diseases, such as migraine and cluster headache, have headaches that occur at the same time each day. We learned that the trigeminal ganglion, an important pain structure in several headache diseases, has a 24-hour cycle that might be related to this daily cycle of headaches. Our genetic analysis suggests that some medications may be more effective in treating migraine and cluster headache when taken at specific times of the day.
Collapse
Affiliation(s)
- Chorong Han
- Department of Biochemistry and Molecular Biology, UTHealth Houston, Houston, Texas, USA
| | - Ji Ye Lim
- Department of Biochemistry and Molecular Biology, UTHealth Houston, Houston, Texas, USA
| | - Nobuya Koike
- Department of Physiology and Systems Bioscience, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Sun Young Kim
- Department of Biochemistry and Molecular Biology, UTHealth Houston, Houston, Texas, USA
| | - Kaori Ono
- Department of Biochemistry and Molecular Biology, UTHealth Houston, Houston, Texas, USA
| | - Celia K. Tran
- Department of Biochemistry and Molecular Biology, UTHealth Houston, Houston, Texas, USA
| | - Elizaveta Mangutov
- Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Eunju Kim
- Department of Biochemistry and Molecular Biology, UTHealth Houston, Houston, Texas, USA
| | - Yanping Zhang
- Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Lingyong Li
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Amynah A. Pradhan
- Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Kazuhiro Yagita
- Department of Physiology and Systems Bioscience, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Zheng Chen
- Department of Biochemistry and Molecular Biology, UTHealth Houston, Houston, Texas, USA
| | - Seung-Hee Yoo
- Department of Biochemistry and Molecular Biology, UTHealth Houston, Houston, Texas, USA
| | - Mark J. Burish
- Department of Neurosurgery, UTHealth Houston, Houston, Texas, USA
| |
Collapse
|
5
|
Engin A. Misalignment of Circadian Rhythms in Diet-Induced Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:27-71. [PMID: 39287848 DOI: 10.1007/978-3-031-63657-8_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The biological clocks of the circadian timing system coordinate cellular and physiological processes and synchronize them with daily cycles. While the central clock in the suprachiasmatic nucleus (SCN) is mainly synchronized by the light/dark cycles, the peripheral clocks react to other stimuli, including the feeding/fasting state, nutrients, sleep-wake cycles, and physical activity. During the disruption of circadian rhythms due to genetic mutations or social and occupational obligations, incorrect arrangement between the internal clock system and environmental rhythms leads to the development of obesity. Desynchronization between the central and peripheral clocks by altered timing of food intake and diet composition leads to uncoupling of the peripheral clocks from the central pacemaker and to the development of metabolic disorders. The strong coupling of the SCN to the light-dark cycle creates a situation of misalignment when food is ingested during the "wrong" time of day. Food-anticipatory activity is mediated by a self-sustained circadian timing, and its principal component is a food-entrainable oscillator. Modifying the time of feeding alone greatly affects body weight, whereas ketogenic diet (KD) influences circadian biology, through the modulation of clock gene expression. Night-eating behavior is one of the causes of circadian disruption, and night eaters have compulsive and uncontrolled eating with severe obesity. By contrast, time-restricted eating (TRE) restores circadian rhythms through maintaining an appropriate daily rhythm of the eating-fasting cycle. The hypothalamus has a crucial role in the regulation of energy balance rather than food intake. While circadian locomotor output cycles kaput (CLOCK) expression levels increase with high-fat diet-induced obesity, peroxisome proliferator-activated receptor-alpha (PPARα) increases the transcriptional level of brain and muscle aryl hydrocarbon receptor nuclear translocator (ARNT)-like 1 (BMAL1) in obese subjects. In this context, effective timing of chronotherapies aiming to correct SCN-driven rhythms depends on an accurate assessment of the SCN phase. In fact, in a multi-oscillator system, local rhythmicity and its disruption reflects the disruption of either local clocks or central clocks, thus imposing rhythmicity on those local tissues, whereas misalignment of peripheral oscillators is due to exosome-based intercellular communication.Consequently, disruption of clock genes results in dyslipidemia, insulin resistance, and obesity, while light exposure during the daytime, food intake during the daytime, and sleeping during the biological night promote circadian alignment between the central and peripheral clocks. Thus, shift work is associated with an increased risk of obesity, diabetes, and cardiovascular diseases because of unusual eating times as well as unusual light exposure and disruption of the circadian rhythm.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
6
|
Cincotta AH. Brain Dopamine-Clock Interactions Regulate Cardiometabolic Physiology: Mechanisms of the Observed Cardioprotective Effects of Circadian-Timed Bromocriptine-QR Therapy in Type 2 Diabetes Subjects. Int J Mol Sci 2023; 24:13255. [PMID: 37686060 PMCID: PMC10487918 DOI: 10.3390/ijms241713255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/19/2023] [Accepted: 07/27/2023] [Indexed: 09/10/2023] Open
Abstract
Despite enormous global efforts within clinical research and medical practice to reduce cardiovascular disease(s) (CVD), it still remains the leading cause of death worldwide. While genetic factors clearly contribute to CVD etiology, the preponderance of epidemiological data indicate that a major common denominator among diverse ethnic populations from around the world contributing to CVD is the composite of Western lifestyle cofactors, particularly Western diets (high saturated fat/simple sugar [particularly high fructose and sucrose and to a lesser extent glucose] diets), psychosocial stress, depression, and altered sleep/wake architecture. Such Western lifestyle cofactors are potent drivers for the increased risk of metabolic syndrome and its attendant downstream CVD. The central nervous system (CNS) evolved to respond to and anticipate changes in the external (and internal) environment to adapt survival mechanisms to perceived stresses (challenges to normal biological function), including the aforementioned Western lifestyle cofactors. Within the CNS of vertebrates in the wild, the biological clock circuitry surveils the environment and has evolved mechanisms for the induction of the obese, insulin-resistant state as a survival mechanism against an anticipated ensuing season of low/no food availability. The peripheral tissues utilize fat as an energy source under muscle insulin resistance, while increased hepatic insulin resistance more readily supplies glucose to the brain. This neural clock function also orchestrates the reversal of the obese, insulin-resistant condition when the low food availability season ends. The circadian neural network that produces these seasonal shifts in metabolism is also responsive to Western lifestyle stressors that drive the CNS clock into survival mode. A major component of this natural or Western lifestyle stressor-induced CNS clock neurophysiological shift potentiating the obese, insulin-resistant state is a diminution of the circadian peak of dopaminergic input activity to the pacemaker clock center, suprachiasmatic nucleus. Pharmacologically preventing this loss of circadian peak dopaminergic activity both prevents and reverses existing metabolic syndrome in a wide variety of animal models of the disorder, including high fat-fed animals. Clinically, across a variety of different study designs, circadian-timed bromocriptine-QR (quick release) (a unique formulation of micronized bromocriptine-a dopamine D2 receptor agonist) therapy of type 2 diabetes subjects improved hyperglycemia, hyperlipidemia, hypertension, immune sterile inflammation, and/or adverse cardiovascular event rate. The present review details the seminal circadian science investigations delineating important roles for CNS circadian peak dopaminergic activity in the regulation of peripheral fuel metabolism and cardiovascular biology and also summarizes the clinical study findings of bromocriptine-QR therapy on cardiometabolic outcomes in type 2 diabetes subjects.
Collapse
|
7
|
Yazdani V, Zeraattalab-Motlagh S, Jayedi A, Majdi M, Mirrafiei A, Martami F, Djafarian K, Shab-Bidar S. The association between self-reported nocturnal sleep duration, irregularity in daily energy intake and diet quality in a sample of Iranian adults. Public Health Nutr 2023; 26:1609-1616. [PMID: 37039130 PMCID: PMC10410366 DOI: 10.1017/s1368980023000502] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/02/2023] [Accepted: 01/18/2023] [Indexed: 04/12/2023]
Abstract
OBJECTIVE Evidence on the relationship between sleep duration and irregularity in daily energy intake with diet quality in Iranian adults is scarce. We aimed to evaluate the association of sleep duration with diet quality and irregularity in daily energy intake. DESIGN This is a cross-sectional study. SETTING The study was performed in healthcare centres in Tehran. PARTICIPANTS 739 adults aged 20-59 years were recruited. Dietary intake was assessed by a FFQ and three 24-h dietary recalls. Diet quality was assessed using the Healthy Eating Index-2015 (HEI-2015). An irregularity score of daily energy intake was calculated based on the deviation from the 3-d mean energy intake. Sleep duration was estimated using self-reported nocturnal sleep duration by each person. RESULTS The mean age of the study participants was 44·4 ± 10·7 years; 70 % were women. The mean nocturnal sleep duration, HEI score and irregularity score were 6·7 ± 1·22 h/d, 52·5 ± 8·55 and 22·9 + 19, respectively. After adjusting for potential confounders, sleep duration was not associated with adherence to HEI-2015 (OR: 1·16; 95 % CI 0·77, 1·74). Longer sleep duration was marginally associated with a lower odd of irregularity in daily energy intake. However, after adjustment for various confounders, this association was not significant (OR: 0·82; 95 % CI 0·50, 1·33; Ptrend = 0·45). No significant interaction was observed between sleep duration and irregularity in daily energy intake in relation to adherence to HEI-2015 (Pinteraction = 0·48). CONCLUSIONS We found that sleep duration was not associated with adherence to HEI-2015 and irregularity in daily energy intake. Further prospective studies are warranted.
Collapse
Affiliation(s)
- Vida Yazdani
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran1416753955, Islamic Republic of Iran
| | - Sheida Zeraattalab-Motlagh
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran1416753955, Islamic Republic of Iran
| | - Ahmad Jayedi
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran1416753955, Islamic Republic of Iran
| | - Maryam Majdi
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran1416753955, Islamic Republic of Iran
| | - Amin Mirrafiei
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran1416753955, Islamic Republic of Iran
| | - Fahimeh Martami
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran1416753955, Islamic Republic of Iran
| | - Kurosh Djafarian
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Islamic Republic of Iran
| | - Sakineh Shab-Bidar
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran1416753955, Islamic Republic of Iran
| |
Collapse
|
8
|
Ragozzino FJ, Peterson B, Karatsoreos IN, Peters JH. Circadian regulation of glutamate release pathways shapes synaptic throughput in the brainstem nucleus of the solitary tract (NTS). J Physiol 2023; 601:1881-1896. [PMID: 36975145 PMCID: PMC10192157 DOI: 10.1113/jp284370] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/06/2023] [Indexed: 03/29/2023] Open
Abstract
Circadian regulation of autonomic reflex pathways pairs physiological function with the daily light cycle. The brainstem nucleus of the solitary tract (NTS) is a key candidate for rhythmic control of the autonomic nervous system. Here we investigated circadian regulation of NTS neurotransmission and synaptic throughput using patch-clamp electrophysiology in brainstem slices from mice. We found that spontaneous quantal glutamate release onto NTS neurons showed strong circadian rhythmicity, with the highest rate of release during the light phase and the lowest in the dark, that were sufficient to drive day/night differences in constitutive postsynaptic action potential firing. In contrast, afferent evoked action potential throughput was enhanced during the dark and diminished in the light. Afferent-driven synchronous release pathways showed a similar decrease in release probability that did not explain the enhanced synaptic throughput during the night. However, analysis of postsynaptic membrane properties revealed diurnal changes in conductance, which, when coupled with the circadian changes in glutamate release pathways, tuned synaptic throughput between the light and dark phases. These coordinated pre-/postsynaptic changes encode nuanced control over synaptic performance and pair NTS action potential firing and vagal throughput with time of day. KEY POINTS: Vagal afferent neurons relay information from peripheral organs to the brainstem nucleus of the solitary tract (NTS) to initiate autonomic reflex pathways as well as providing important controls of food intake, digestive function and energy balance. Vagally mediated reflexes and behaviours are under strong circadian regulation. Diurnal fluctuations in presynaptic vesicle release pathways and postsynaptic membrane conductances provide nuanced control over NTS action potential firing and vagal synaptic throughput. Coordinated pre-/postsynaptic changes represent a fundamental mechanism mediating daily changes in vagal afferent signalling and autonomic function.
Collapse
Affiliation(s)
- Forrest J. Ragozzino
- Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - BreeAnne Peterson
- Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Ilia N. Karatsoreos
- Department of Psychological and Brain Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - James H. Peters
- Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| |
Collapse
|
9
|
Ye BJ. Association between Shift Work and Metabolic Syndrome: A 4-Year Retrospective Cohort Study. Healthcare (Basel) 2023; 11:healthcare11060802. [PMID: 36981459 PMCID: PMC10048347 DOI: 10.3390/healthcare11060802] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/11/2023] Open
Abstract
(1) Background: Previous studies on the association between shift work and metabolic syndrome have had inconsistent results. This may be due to the cross-sectional study design and non-objective data used in those studies. Hence, this study aimed to identify risk factors for Metabolic syndrome using objective information provided by the relevant companies and longitudinal data provided in health examinations. (2) Methods: In total, 1211 male workers of three manufacturing companies, including shift workers, were surveyed annually for 4 years. Data on age, smoking, drinking, physical activity, length of shift work, type of shift, past history, waist circumference, blood pressure, blood sugar, triglyceride, and high-density cholesterol (HDL) were collected and analyzed using generalized estimating equations (GEE) to identify the risk factors for Metabolic syndrome. (3) Results: In the multivariate analysis of Metabolic syndrome risk factors, age (OR = 1.078, 95% CI: 1.045–1.112), current smoking (OR = 1.428, 95% CI: 1.815–5.325), and BMI (OR = 1.498, 95% CI: 1.338–1.676) were statistically significant for day workers (n= 510). Additionally, for shift workers (N = 701), age (OR = 1.064, 95% CI: 1.008–1.174), current smoking (OR = 2.092, 95% CI: 1.854–8.439), BMI (OR = 1.471, 95% CI: 1.253–1.727) and length of shift work (OR = 1.115, 95% CI: 1.010-1.320) were statistically significant. Shift work was associated with a higher risk of Metabolic syndrome (OR = 1.093, 95% CI: 1.137–2.233) compared to day workers. For shift workers, shift work for more than 20 years was associated with Metabolic syndrome (OR = 2.080, 95% CI: 1.911–9.103), but the dose–response relationship was not statistically significant. (4) Conclusions: This study revealed that age, current smoking, BMI, and shift work are potential risk factors for Metabolic syndrome. In particular, the length of shift work (>20 years) is a potential risk factor for Metabolic syndrome in shift workers. To prevent metabolic syndrome in shift workers, health managers need to actively accommodate shift workers (especially those who have worked for more than 20 years), current smokers, and obese people. A long-term cohort study based on objective data is needed to identify the chronic health impact and the risk factors of shift work.
Collapse
Affiliation(s)
- Byeong-Jin Ye
- Department of Occupational and Environmental Medicine & Institute of Environmental and Occupational Medicine, Busan Paik Hospital, Inje University, Busan 47392, Republic of Korea
| |
Collapse
|
10
|
Tiwari A, Rathor P, Trivedi PK, Ch R. Multi-Omics Reveal Interplay between Circadian Dysfunction and Type2 Diabetes. BIOLOGY 2023; 12:301. [PMID: 36829576 PMCID: PMC9953493 DOI: 10.3390/biology12020301] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023]
Abstract
Type 2 diabetes is one of the leading threats to human health in the 21st century. It is a metabolic disorder characterized by a dysregulated glucose metabolism resulting from impaired insulin secretion or insulin resistance. More recently, accumulated epidemiological and animal model studies have confirmed that circadian dysfunction caused by shift work, late meal timing, and sleep loss leads to type 2 diabetes. Circadian rhythms, 24-h endogenous biological oscillations, are a fundamental feature of nearly all organisms and control many physiological and cellular functions. In mammals, light synchronizes brain clocks and feeding is a main stimulus that synchronizes the peripheral clocks in metabolic tissues, such as liver, pancreas, muscles, and adipose tissues. Circadian arrhythmia causes the loss of synchrony of the clocks of these metabolic tissues and leads to an impaired pancreas β-cell metabolism coupled with altered insulin secretion. In addition to these, gut microbes and circadian rhythms are intertwined via metabolic regulation. Omics approaches play a significant role in unraveling how a disrupted circadian metabolism causes type 2 diabetes. In the present review, we emphasize the discoveries of several genes, proteins, and metabolites that contribute to the emergence of type 2 diabetes mellitus (T2D). The implications of these discoveries for comprehending the circadian clock network in T2D may lead to new therapeutic solutions.
Collapse
Affiliation(s)
- Ashutosh Tiwari
- Metabolomics Lab, CSIR-Central Institute of Medicinal & Aromatic Plants (CIMAP), Lucknow 226015, India
| | - Priya Rathor
- Metabolomics Lab, CSIR-Central Institute of Medicinal & Aromatic Plants (CIMAP), Lucknow 226015, India
| | - Prabodh Kumar Trivedi
- Department of Biotechnology, CSIR-Central Institute of Medicinal & Aromatic Plants (CIMAP), Lucknow 226015, India
- Academy of Council of Scientific and Industrial Research (ACSIR), Gaziabad 201002, India
| | - Ratnasekhar Ch
- Metabolomics Lab, CSIR-Central Institute of Medicinal & Aromatic Plants (CIMAP), Lucknow 226015, India
- Academy of Council of Scientific and Industrial Research (ACSIR), Gaziabad 201002, India
- School of Biological Sciences, Queen’s University Belfast, Belfast BT9 5DL, UK
| |
Collapse
|
11
|
Teixeira GP, Guimarães KC, Soares AGNS, Marqueze EC, Moreno CRC, Mota MC, Crispim CA. Role of chronotype in dietary intake, meal timing, and obesity: a systematic review. Nutr Rev 2022; 81:75-90. [PMID: 35771674 DOI: 10.1093/nutrit/nuac044] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
CONTEXT Recent studies show that dietary habits and obesity seem to be influenced by chronotype, which reflects an individual's preference for the timing of sleeping, eating, and activity in a 24-hour period. OBJECTIVE This review aimed to analyze the association of chronotype with dietary habits, namely energy and macronutrient intakes, meal timing, and eating patterns, as well as with obesity. DATA SOURCES PubMed/MEDLINE, LILACS, and Google Scholar databases were searched between 2004 and 2020. Study selection was performed by 2 authors independently; disagreements on eligibility of articles were resolved by a third author. After assessment of 12 060 abstracts, 43 studies (21 articles on obesity; 13 on food consumption, meal timing, and eating patterns; and 9 that addressed both obesity and dietary behavior) were included. DATA EXTRACTION A standard form was used to extract study design, country, number of participants, method of chronotype determination, and main findings. DATA ANALYSIS Approximately 95% of included studies showed an association between eveningness and at least 1 unhealthy eating habit. Morningness was associated with regular consumption of fresh and minimally processed foods. In addition, about 47% of studies showed a higher association between late types and obesity. CONCLUSION Late types are more likely to present unhealthy eating habits, such as eating late at night, skipping breakfast often, and eating processed/ultraprocessed foods, while early types are more likely to have healthy and protective habits, such as eating early and eating predominantly fresh/minimally processed foods. Intermediate types tend to have a pattern of health and eating more similar to early types than to late types. Late types are also more likely to present higher weight and body mass index than early or intermediate types. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42021256078.
Collapse
Affiliation(s)
- Gabriela P Teixeira
- are with the Graduate Program in Health Sciences, Faculty of Medicine, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Kisian C Guimarães
- are with the Graduate Program in Health Sciences, Faculty of Medicine, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Ana Gabriela N S Soares
- are with the Nutrition Course, Faculty of Medicine, Federal University of Uberlândia, Minas Gerais Uberlândia, Brazil
| | - Elaine C Marqueze
- are with the School of Public Health, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Cláudia R C Moreno
- are with the School of Public Health, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Maria C Mota
- are with the Graduate Program in Health Sciences, Faculty of Medicine, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Cibele A Crispim
- are with the Graduate Program in Health Sciences, Faculty of Medicine, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil.,are with the Nutrition Course, Faculty of Medicine, Federal University of Uberlândia, Minas Gerais Uberlândia, Brazil
| |
Collapse
|
12
|
Herrera Moro Chao D, Kirchner MK, Pham C, Foppen E, Denis RGP, Castel J, Morel C, Montalban E, Hassouna R, Bui LC, Renault J, Mouffle C, García-Cáceres C, Tschöp MH, Li D, Martin C, Stern JE, Luquet SH. Hypothalamic astrocytes control systemic glucose metabolism and energy balance. Cell Metab 2022; 34:1532-1547.e6. [PMID: 36198294 PMCID: PMC9615252 DOI: 10.1016/j.cmet.2022.09.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 06/23/2022] [Accepted: 09/02/2022] [Indexed: 01/29/2023]
Abstract
The hypothalamus is key in the control of energy balance. However, strategies targeting hypothalamic neurons have failed to provide viable options to treat most metabolic diseases. Conversely, the role of astrocytes in systemic metabolic control has remained largely unexplored. Here, we show that obesity promotes anatomically restricted remodeling of hypothalamic astrocyte activity. In the paraventricular nucleus (PVN) of the hypothalamus, chemogenetic manipulation of astrocytes results in bidirectional control of neighboring neuron activity, autonomic outflow, glucose metabolism, and energy balance. This process recruits a mechanism involving the astrocytic control of ambient glutamate levels, which becomes defective in obesity. Positive or negative chemogenetic manipulation of PVN astrocyte Ca2+ signals, respectively, worsens or improves metabolic status of diet-induced obese mice. Collectively, these findings highlight a yet unappreciated role for astrocytes in the direct control of systemic metabolism and suggest potential targets for anti-obesity strategy.
Collapse
Affiliation(s)
| | - Matthew K Kirchner
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA; Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA 30302, USA
| | - Cuong Pham
- Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR8246, INSERM U1130, Sorbonne Universite, Paris 75005, France
| | - Ewout Foppen
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Raphael G P Denis
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France; Institut Cochin, Université Paris Cité, INSERM U1016, CNRS UMR 8104, 75014 Paris, France
| | - Julien Castel
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Chloe Morel
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Enrica Montalban
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Rim Hassouna
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Linh-Chi Bui
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Justine Renault
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Christine Mouffle
- Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR8246, INSERM U1130, Sorbonne Universite, Paris 75005, France
| | - Cristina García-Cáceres
- Helmholtz Diabetes Center (HDC) & German Center for Diabetes Research (DZD), Helmholtz Zentrum München, Neuherberg, 85764, Germany; Division of Metabolic Diseases, Technische Universität München, Munich, 80333, Germany; Medizinische Klinik and Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Matthias H Tschöp
- Helmholtz Diabetes Center (HDC) & German Center for Diabetes Research (DZD), Helmholtz Zentrum München, Neuherberg, 85764, Germany; Division of Metabolic Diseases, Technische Universität München, Munich, 80333, Germany
| | - Dongdong Li
- Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR8246, INSERM U1130, Sorbonne Universite, Paris 75005, France
| | - Claire Martin
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Javier E Stern
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA; Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA 30302, USA
| | - Serge H Luquet
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France.
| |
Collapse
|
13
|
Vaca-Dempere M, Kumar A, Sica V, Muñoz-Cánoves P. Running skeletal muscle clocks on time- the determining factors. Exp Cell Res 2022; 413:112989. [PMID: 35081395 DOI: 10.1016/j.yexcr.2021.112989] [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: 03/29/2021] [Revised: 12/08/2021] [Accepted: 12/19/2021] [Indexed: 11/23/2022]
Abstract
Circadian rhythms generate 24 h-long oscillations, which are key regulators of many aspects of behavior and physiology. Recent circadian transcriptome studies have discovered rhythmicity at the transcriptional level of hundreds of skeletal muscle genes, with roles in skeletal muscle growth, maintenance, and metabolic functions. These rhythms allow this tissue to perform molecular functions at the appropriate time of the day in order to anticipate environmental changes. However, while the last decade of research has characterized several aspects of the skeletal muscle molecular clock, many still are unexplored, including its functions, regulatory mechanisms, and interactions with other tissues. The central clock is believed to be located in the suprachiasmatic nucleus (SCN) of the brain hypothalamus, providing entrainment to peripheral organs through humoral and neuronal signals. However, these mechanisms of action are still unknown. Conversely, muscle tissue can be entrained through extrinsic, SCN-independent factors, such as feeding and physical activity. In this review, we provide an overview of the recent research about the extrinsic and intrinsic factors required for skeletal muscle clock regulation. Furthermore, we discuss the need for future studies to elucidate the mechanisms behind this regulation, which will in turn help dissect the role of circadian disruption at the onset of aging and diseases.
Collapse
Affiliation(s)
- Mireia Vaca-Dempere
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), 08003, Barcelona, Spain
| | - Arun Kumar
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), 08003, Barcelona, Spain
| | - Valentina Sica
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), 08003, Barcelona, Spain
| | - Pura Muñoz-Cánoves
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), 08003, Barcelona, Spain; ICREA, 08010, Barcelona, Spain; Spanish National Center on Cardiovascular Research (CNIC), 28029, Madrid, Spain.
| |
Collapse
|
14
|
Malik S, Stokes Iii J, Manne U, Singh R, Mishra MK. Understanding the significance of biological clock and its impact on cancer incidence. Cancer Lett 2022; 527:80-94. [PMID: 34906624 PMCID: PMC8816870 DOI: 10.1016/j.canlet.2021.12.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 12/12/2022]
Abstract
The circadian clock is an essential timekeeper that controls, for humans, the daily rhythm of biochemical, physiological, and behavioral functions. Irregular performance or disruption in circadian rhythms results in various diseases, including cancer. As a factor in cancer development, perturbations in circadian rhythms can affect circadian homeostasis in energy balance, lead to alterations in the cell cycle, and cause dysregulation of chromatin remodeling. However, knowledge gaps remain in our understanding of the relationship between the circadian clock and cancer. Therefore, a mechanistic understanding by which circadian disruption enhances cancer risk is needed. This review article outlines the importance of the circadian clock in tumorigenesis and summarizes underlying mechanisms in the clock and its carcinogenic mechanisms, highlighting advances in chronotherapy for cancer treatment.
Collapse
Affiliation(s)
- Shalie Malik
- Cancer Biology Research and Training, Department of Biological Sciences, Alabama State University, Montgomery, AL, USA; Department of Zoology and Dr. Giri Lal Gupta Institute of Public Health and Public Affairs, University of Lucknow, Lucknow, UP, India
| | - James Stokes Iii
- Department of Biological and Environmental Sciences, Auburn University, Montgomery, AL, USA
| | - Upender Manne
- Departments of Pathology, Surgery and Epidemiology, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rajesh Singh
- Department of Microbiology, Biochemistry, and Immunology, Cancer Health Equity Institute, Morehouse School of Medicine, Atlanta, GA, USA
| | - Manoj K Mishra
- Cancer Biology Research and Training, Department of Biological Sciences, Alabama State University, Montgomery, AL, USA.
| |
Collapse
|
15
|
Melatonin ameliorates cardiac remodelling in fructose-induced metabolic syndrome rat model by using genes encoding cardiac potassium ion channels. Mol Biol Rep 2021; 48:5811-5819. [PMID: 34347240 DOI: 10.1007/s11033-021-06526-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 06/27/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Metabolic syndrome comprises a group of disorders, including cardiac abnormalities. Ventricular arrhythmias observed in metabolic syndrome are due to the impaired ventricular repolarization. This study aims to determine the effects of melatonin on cardiac ventricular repolarization in metabolic syndrome rat model. METHODS AND RESULTS Sprague-Dawley rats were divided into control (n = 8), melatonin (n = 8), metabolic syndrome (n = 8) and metabolic syndrome + melatonin (n = 8) groups. Fructose (200 g/lt/day) was added into the drinking water during 8 weeks of rats to induce metabolic syndrome model. In the last two weeks, melatonin (20 mg/kg/day) was administered via oral gavage. Blood pressure measurements and ECG recordings were taken at three different times. Blood and left ventricular tissue samples were harvested and the KCNQ1,3 and KCNH2 gene expressions were analysed by qRT-PCR method. We observed insulin resistance, hyperglycemia, dyslipidemia and higher systolic blood pressure in metabolic syndrome group (p < 0.01, for all). Prolonged QT interval was observed in metabolic syndrome group (p < 0.001). The expression levels of the KCNQ genes encoding the Kv7 channel was significantly reduced, however KCNH2 gene which encodes Kv11.1 channel was increased in metabolic syndrome group compared to control group (p < 0.05, p < 0.001, respectively). Melatonin significantly normalised the prolongation on QT interval in metabolic syndrome group (p < 0.001) and the expressions of the KCNQ (p < 0.002) and KCNH2 genes (p = 0.003). CONCLUSIONS The present study revealed that melatonin had ameliorative effects on ventricular repolarization by improving the prolonged QT duration in rats with metabolic syndrome and this effect was generated by the KCNQ and KCNH2 gene families.
Collapse
|
16
|
Kurhaluk N, Tkachenko H, Lukash O. Photoperiod-induced alterations in biomarkers of oxidative stress and biochemical pathways in rats of different ages: Focus on individual physiological reactivity. Chronobiol Int 2021; 38:1673-1691. [PMID: 34121553 DOI: 10.1080/07420528.2021.1939364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Effects of photoperiodicity caused by both the age and individual physiological reactivity estimated by resistance to hypobaric hypoxia on the levels of lipid peroxidation, protein oxidation (aldehydic and ketonic derivatives), total antioxidant capacity, activities of antioxidant enzymes (superoxide dismutase, catalase, glutathione peroxidase, and glutathione reductase), and biochemical parameters of aerobic and anaerobic pathways in hepatic tissue depending on the blood melatonin level were studied. The study was carried out on 96 6- and 21-month-old male rats divided into hypoxia resistance groups (LR, low resistance, HR, high resistance). The analyses were conducted at four photoperiods: winter (January), spring (March), summer (July), and autumn (October). Our results indicate a significant effect of melatonin, i.e. over 80%, revealed by the complete statistical model of the studied biomarkers of oxidative stress and oxygen-dependent parameters of metabolism. The effects of melatonin vary with age and between photoperiods, which in turn was determined by individual physiological reactivity. In terms of the photoperiods, the melatonin content in the group of the adult animals with low resistance to hypoxia decreased from winter to summer. In a group of old animals in comparison with adults, the melatonin content in all the studied photoperiods was much lower as well, regardless of their hypoxia resistance. In the group of old animals with low resistance to hypoxia, the melatonin content decreased throughout the photoperiods as follows: winter, autumn, summer, and spring. As can be concluded, spring is a critical period for old animals, particularly those with low hypoxia resistance. The important role of melatonin in these processes was also confirmed by our correlation analysis between oxidative stress biomarkers, energy-related metabolites, and antioxidant enzymes in the hepatic tissue of rats of different ages, with different resistance to hypoxia, and in different photoperiods. The melatonin concentration in the blood of highly resistant rats was higher than in those with low resistance to hypoxia. Melatonin determines the individual constitutional level of resistance to hypoxia and is responsible for individual enzymatic antioxidative responses, depending on the four photoperiods. Our studies have shown that melatonin levels are related to the redox characteristics of antioxidant defenses against lipid peroxidation and oxidative modification of proteins in old rats with low resistance to hypoxia, compared to a group of highly resistant adults. Finally, the melatonin-related mechanisms of antioxidative protection depend on metabolic processes in hepatic tissue and exhibit photoperiodical variability in adult and old rats.
Collapse
Affiliation(s)
- Natalia Kurhaluk
- Department of Zoology and Animal Physiology, Institute of Biology and Earth Sciences, Pomeranian University in Słupsk, Słupsk, Poland
| | - Halyna Tkachenko
- Department of Zoology and Animal Physiology, Institute of Biology and Earth Sciences, Pomeranian University in Słupsk, Słupsk, Poland
| | - Oleksandr Lukash
- Department of Ecology and Nature Protection, T.G. Shevchenko National University "Chernihiv Collegium", Chernihiv, Ukraine
| |
Collapse
|
17
|
Abstract
OBJECTIVES Core clock genes regulate tissue-specific transcriptome oscillations that synchronize physiologic processes throughout the body, held in phase by the central circadian rhythm. The central circadian rhythm rapidly dampens with onset of critical illness, but the effect of critical illness on gene expression oscillations is unknown. The objective of this study was to characterize the rhythmicity and phase coherence of core clock genes and the broader transcriptome after onset of critical illness. DESIGN Cross-sectional study. SETTING ICUs and hospital clinical research unit. PATIENTS Critically ill patients within the first day of presenting from the community and healthy volunteers. INTERVENTIONS Usual care (critically ill patients) and modified constant routine (healthy volunteers). MEASUREMENTS AND MAIN RESULTS We studied 15 critically ill patients, including 10 with sepsis and five with intracerebral hemorrhage, and 11 healthy controls. The central circadian rhythm and rest-activity rhythms were profiled by continuous wrist actigraphy, and serum melatonin sampled every 2 hours along with whole blood for RNA isolation over 24 hours. The gene expression transcriptome was obtained by RNA sequencing. Core clock genes were analyzed for rhythmicity by cosinor fit. Significant circadian rhythmicity was identified in five of six core clock genes in healthy controls, but none in critically ill patients. TimeSignature, a validated algorithm based on 41 genes, was applied to assess overall transcriptome phase coherence. Median absolute error of TimeSignature was higher in individual critically ill patients than healthy patients (4.90 vs 1.48 hr) and was correlated with encephalopathy severity by Glasgow Coma Scale in critically ill patients (rho, -0.54; p = 0.036). CONCLUSIONS Gene expression rhythms rapidly become abnormal during critical illness. The association between disrupted transcriptome rhythms and encephalopathy suggests a path for future work to elucidate the underlying pathophysiology.
Collapse
|
18
|
Arellanes-Licea EDC, Pérez-Mendoza M, Carmona-Castro A, Díaz-Muñoz M, Miranda-Anaya M. Obese Neotomodon alstoni mice exhibit sexual dimorphism in the daily profile of circulating melatonin and clock proteins PER1 and BMAL1 in the hypothalamus and peripheral oscillators. Chronobiol Int 2021; 38:584-597. [PMID: 33393371 DOI: 10.1080/07420528.2020.1860999] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 12/02/2020] [Accepted: 12/03/2020] [Indexed: 12/13/2022]
Abstract
Obesity is a global health threat and a risk factor for several metabolic conditions. Though circadian dysfunction has been considered among the multiple causes of obesity, little work has been done to explore the relationship between obesity, circadian dysfunction, and sexual dimorphism. The Neotomodon alstoni mouse is a suitable model for such research. This study employed N. alstoni mice in a chronobiological analysis to determine whether there is circadian desynchronization of relative PER1 and BMAL1 protein levels in the hypothalamus, liver, visceral white adipose tissue, kidney, and heart. It also compared differences between sexes and lean and obese N. alstoni adult mice, by recording behavior and daily circulating serum melatonin as markers of circadian output. We found that obese mice display reduced locomotor activity. Additionally, Cosinor analyses of the relative expression of PER1 and BMAL1 show differences between lean and obese mice in a sex-linked manner. The PER1 24 h rhythm was absent in all tissues of obese males and significant in the tissues of obese females. The BMAL1 24 h rhythm also was significant in most of the tissues tested in lean males, whereas it was significant and shifted the acrophase (peak time of rhythm) in most of the tissues in obese females. Both lean male and female mice showed a rhythmic 24 h pattern of circulating serum melatonin. This daily profile was not only absent in obese mice of both sexes but showed sexual dimorphism. Obese male mice showed lower circulating levels of melatonin compared to lean male mice, but they were higher in obese females compared to lean females. Our results suggest that obesity in N. alstoni is associated with an internal circadian desynchronization in a sex-dependent manner. Overall, this study reinforces the need for further research on the neuroendocrinology of obesity and circadian rhythms using this biological model.
Collapse
Affiliation(s)
- Elvira Del Carmen Arellanes-Licea
- Unidad Multidisciplinaria de Docencia e Investigación, Facultad de Ciencias, Universidad Nacional Autónoma de México, Querétaro, México
| | - Moisés Pérez-Mendoza
- Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Querétaro, México
| | - Agustín Carmona-Castro
- Departamento de Biología Celular, Facultad de Ciencias, Ciudad Universitaria, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Mauricio Díaz-Muñoz
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| | - Manuel Miranda-Anaya
- Unidad Multidisciplinaria de Docencia e Investigación, Facultad de Ciencias, Universidad Nacional Autónoma de México, Querétaro, México
| |
Collapse
|
19
|
Circadian rhythms: influence on physiology, pharmacology, and therapeutic interventions. J Pharmacokinet Pharmacodyn 2021; 48:321-338. [PMID: 33797011 PMCID: PMC8015932 DOI: 10.1007/s10928-021-09751-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 03/19/2021] [Indexed: 12/20/2022]
Abstract
Circadian rhythms are ubiquitous phenomena that recur daily in a self-sustaining, entrainable, and oscillatory manner, and orchestrate a wide range of molecular, physiological, and behavioral processes. Circadian clocks are comprised of a hierarchical network of central and peripheral clocks that generate, sustain, and synchronize the circadian rhythms. The functioning of the peripheral clock is regulated by signals from autonomic innervation (from the central clock), endocrine networks, feeding, and other external cues. The critical role played by circadian rhythms in maintaining both systemic and tissue-level homeostasis is well established, and disruption of the rhythm has direct consequence for human health, disorders, and diseases. Circadian oscillations in both pharmacokinetics and pharmacodynamic processes are known to affect efficacy and toxicity of several therapeutic agents. A variety of modeling approaches ranging from empirical to more complex systems modeling approaches have been applied to characterize circadian biology and its influence on drug actions, optimize time of dosing, and identify opportunities for pharmacological modulation of the clock mechanisms and their downstream effects. In this review, we summarize current understanding of circadian rhythms and its influence on physiology, pharmacology, and therapeutic interventions, and discuss the role of chronopharmacometrics in gaining new insights into circadian rhythms and its applications in chronopharmacology.
Collapse
|
20
|
Abstract
The circadian clock controls several aspects of mammalian physiology and orchestrates the daily oscillations of biological processes and behavior. Our circadian rhythms are driven by an endogenous central clock in the brain that synchronizes with clocks in peripheral tissues, thereby regulating our immune system and the severity of infections. These rhythms affect the pharmacokinetics and efficacy of therapeutic agents and vaccines. The core circadian regulatory circuits and clock-regulated host pathways provide fertile ground to identify novel antiviral therapies. An increased understanding of the role circadian systems play in regulating virus infection and the host response to the virus will inform our clinical management of these diseases. This review provides an overview of the experimental and clinical evidence reporting on the interplay between the circadian clock and viral infections, highlighting the importance of virus-clock research.
Collapse
Affiliation(s)
- Helene Borrmann
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | | | - Xiaodong Zhuang
- Xiaodong Zhuang, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7FZ, UK; e-mail:
| |
Collapse
|
21
|
Romo-Nava F, Buijs RM, McElroy SL. The use of melatonin to mitigate the adverse metabolic side effects of antipsychotics. HANDBOOK OF CLINICAL NEUROLOGY 2021; 179:371-382. [PMID: 34225976 DOI: 10.1016/b978-0-12-819975-6.00024-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Antipsychotic drugs are efficacious first-line treatments for many individuals diagnosed with a psychiatric illness. However, their adverse metabolic side-effect profile, which resembles the metabolic syndrome, represents a significant clinical problem that increases morbidity and limits treatment adherence. Moreover, the mechanisms involved in antipsychotic-induced adverse metabolic effects (AMEs) are unknown and mitigating strategies and interventions are limited. However, recent clinical trials show that nightly administration of exogenous melatonin may mitigate or even prevent antipsychotic-induced AMEs. This clinical evidence in combination with recent preclinical data implicate the circadian system in antipsychotic-induced AMEs and their mitigation. In this chapter, we provide an overview on the circadian system and its involvement in antipsychotic-induced AMEs, as well as the potential beneficial effect of nightly melatonin administration to mitigate them.
Collapse
Affiliation(s)
- Francisco Romo-Nava
- Lindner Center of HOPE Research Institute, Lindner Center of HOPE, Mason, OH, United States; Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH, United States.
| | - Ruud M Buijs
- Hypothalamic Integration Mechanisms Laboratory, Department of Cellular Biology and Physiology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - Susan L McElroy
- Lindner Center of HOPE Research Institute, Lindner Center of HOPE, Mason, OH, United States; Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
22
|
Gopalakrishnan S, Kannan NN. Only time will tell: the interplay between circadian clock and metabolism. Chronobiol Int 2020; 38:149-167. [PMID: 33345624 DOI: 10.1080/07420528.2020.1842436] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In most organisms ranging from cyanobacteria to humans, the endogenous timekeeping system temporally coordinates the behavioral, physiological, and metabolic processes with a periodicity close to 24 h. The timing of these daily rhythms is orchestrated by the synchronized oscillations of both the central pacemaker in the brain and the peripheral clocks located across multiple organs and tissues. A growing body of evidence suggests that the central circadian clock and peripheral clocks residing in the metabolically active tissues are incredibly well coordinated to confer coherent metabolic homeostasis. The interplay between nutrient metabolism and circadian rhythms can occur at various levels supported by the molecular clock network, multiple systemic mechanisms, and the neuroendocrine signaling pathways. While studies suggest the reciprocal regulation between circadian clock and metabolism, it is important to understand the precise mechanisms and the underlying pathways involved in the cross-talk among circadian oscillators and diverse metabolic networks. In addition to the internal synchronization of the metabolic rhythms, feeding time is considered as a potential external synchronization cue that fine tunes the timing of the circadian rhythms in metabolic peripheral clocks. A deeper understanding of how the timing of food intake and the diet composition drive the tissue-specific metabolic rhythms across the body is concomitantly important to develop novel therapeutic strategies for the metabolic disorders arising from circadian misalignment. This review summarizes the recent advancements in the circadian clock regulation of nutrient metabolism and discusses the current understanding of the metabolic feedback signals that link energy metabolism with the circadian clock.
Collapse
Affiliation(s)
- Swetha Gopalakrishnan
- Chronobiology Laboratory, School of Biology, Indian Institute of Science Education and Research (IISER) , Thiruvananthapuram, India
| | - Nisha N Kannan
- Chronobiology Laboratory, School of Biology, Indian Institute of Science Education and Research (IISER) , Thiruvananthapuram, India
| |
Collapse
|
23
|
The mRNA-Binding Protein RBM3 Regulates Activity Patterns and Local Synaptic Translation in Cultured Hippocampal Neurons. J Neurosci 2020; 41:1157-1173. [PMID: 33310754 PMCID: PMC7888222 DOI: 10.1523/jneurosci.0921-20.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 10/14/2020] [Accepted: 11/12/2020] [Indexed: 12/18/2022] Open
Abstract
The activity and the metabolism of the brain change rhythmically during the day/night cycle. Such rhythmicity is also observed in cultured neurons from the suprachiasmatic nucleus, which is a critical center in rhythm maintenance. However, this issue has not been extensively studied in cultures from areas less involved in timekeeping, as the hippocampus. Using neurons cultured from the hippocampi of newborn rats (both male and female), we observed significant time-dependent changes in global activity, in synaptic vesicle dynamics, in synapse size, and in synaptic mRNA amounts. A transcriptome analysis of the neurons, performed at different times over 24 h, revealed significant changes only for RNA-binding motif 3 (Rbm3). RBM3 amounts changed, especially in synapses. RBM3 knockdown altered synaptic vesicle dynamics and changed the neuronal activity patterns. This procedure also altered local translation in synapses, albeit it left the global cellular translation unaffected. We conclude that hippocampal cultured neurons can exhibit strong changes in their activity levels over 24 h, in an RBM3-dependent fashion. SIGNIFICANCE STATEMENT This work is important in several ways. First, the discovery of relatively regular activity patterns in hippocampal cultures implies that future studies using this common model will need to take the time parameter into account, to avoid misinterpretation. Second, our work links these changes in activity strongly to RBM3, in a fashion that is independent of the canonical clock mechanisms, which is a very surprising observation. Third, we describe here probably the first molecule (RBM3) whose manipulation affects translation specifically in synapses, and not at the whole-cell level. This is a key finding for the rapidly growing field of local synaptic translation.
Collapse
|
24
|
Otamas A, Grant PJ, Ajjan RA. Diabetes and atherothrombosis: The circadian rhythm and role of melatonin in vascular protection. Diab Vasc Dis Res 2020; 17:1479164120920582. [PMID: 32506946 PMCID: PMC7607413 DOI: 10.1177/1479164120920582] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Obesity-related euglycaemic insulin resistance clusters with cardiometabolic risk factors, contributing to the development of both type 2 diabetes and cardiovascular disease. An increased thrombotic tendency in diabetes stems from platelet hyperactivity, enhanced activity of prothrombotic coagulation factors and impaired fibrinolysis. Furthermore, a low-grade inflammatory response and increased oxidative stress accelerate the atherosclerotic process and, together with an enhanced thrombotic environment, result in premature and more severe cardiovascular disease. The disruption of circadian cycles in man secondary to chronic obesity and loss of circadian cues is implicated in the increased risk of developing diabetes and cardiovascular disease. Levels of melatonin, the endogenous synchronizer of circadian rhythm, are reduced in individuals with vascular disease and those with deranged glucose metabolism. The anti-inflammatory, antihypertensive, antioxidative and antithrombotic activities of melatonin make it a potential therapeutic agent to reduce the risk of vascular occlusive disease in diabetes. The mechanisms behind melatonin-associated reduction in procoagulant response are not fully known. Current evidence suggests that melatonin inhibits platelet aggregation and might affect the coagulation cascade, altering fibrin clot structure and/or resistance to fibrinolysis. Large-scale clinical trials are warranted to investigate the effects of modulating the circadian clock on insulin resistance, glycaemia and cardiovascular outcome.
Collapse
Affiliation(s)
- Anastasia Otamas
- The LIGHT Laboratories, Leeds Institute of Cardiovascular and Metabolic Medicine and Leeds Teaching Hospitals Trust, University of Leeds, Leeds, UK
| | - Peter J Grant
- The LIGHT Laboratories, Leeds Institute of Cardiovascular and Metabolic Medicine and Leeds Teaching Hospitals Trust, University of Leeds, Leeds, UK
| | - Ramzi A Ajjan
- The LIGHT Laboratories, Leeds Institute of Cardiovascular and Metabolic Medicine and Leeds Teaching Hospitals Trust, University of Leeds, Leeds, UK
| |
Collapse
|
25
|
Pellegrini M, Cioffi I, Evangelista A, Ponzo V, Goitre I, Ciccone G, Ghigo E, Bo S. Effects of time-restricted feeding on body weight and metabolism. A systematic review and meta-analysis. Rev Endocr Metab Disord 2020; 21:17-33. [PMID: 31808043 DOI: 10.1007/s11154-019-09524-w] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Restriction in meal timing has emerged as a promising dietary approach for the management of obesity and dysmetabolic diseases. The present systematic review and meta-analysis summarized the most recent evidence on the effect of time-restricted feeding (TRF) on weight-loss and cardiometabolic variables in comparison with unrestricted-time regimens. Studies involving TRF regimen were systematically searched up to January 2019. Effect size was expressed as weighted mean difference (WMD) and 95% confidence intervals (CI). A total of 11 studies, 5 randomized controlled trials and 6 observational, were included. All selected studies had a control group without time restriction; hours of fasting ranged from 12-h until 20-h and study duration from 4 to 8-weeks. Most studies involved the Ramadan fasting. TRF determined a greater weight-loss than control regimens (11 studies, n = 485 subjects) (WMD: -1.07 kg, 95%CI: -1.74 to -0.40; p = 0.002; I2 = 56.2%), unrelated to study design. The subgroup analysis showed an inverse association between TRF and fat free mass in observational studies (WMD: -1.33 kg, 95%CI: -2.55 to -0.11; p = 0.03; I2 = 0%). An overall significant reduction in fasting glucose concentrations was observed with TRF regimens (7 studies, n = 363 subjects) (WMD: -1.71 mg/dL, 95%CI: -3.20 to -0.21; p = 0.03; I2 = 0%), above all in trials (WMD:-2.45 mg/dL, 95%CI: -4.72 to -0.17; p = 0.03; I2 = 0%). No between-group differences in the other variables were found. TRF regimens achieved a superior effect in promoting weight-loss and reducing fasting glucose compared to approaches with unrestricted time in meal consumption. However, long-term and well-designed trials are needed to draw definitive conclusions.
Collapse
Affiliation(s)
- Marianna Pellegrini
- Department of Medical Sciences, University of Turin, c.so AM Dogliotti 14, 10126, Turin, Italy
| | - Iolanda Cioffi
- Clinical Medicine and Surgery, Federico II University Hospital, Naples, Italy
| | - Andrea Evangelista
- Unit of Clinical Epidemiology, CPO, "Città della Salute e della Scienza" Hospital of Turin, Turin, Italy
| | - Valentina Ponzo
- Department of Medical Sciences, University of Turin, c.so AM Dogliotti 14, 10126, Turin, Italy
| | - Ilaria Goitre
- Department of Medical Sciences, University of Turin, c.so AM Dogliotti 14, 10126, Turin, Italy
| | - Giovannino Ciccone
- Unit of Clinical Epidemiology, CPO, "Città della Salute e della Scienza" Hospital of Turin, Turin, Italy
| | - Ezio Ghigo
- Department of Medical Sciences, University of Turin, c.so AM Dogliotti 14, 10126, Turin, Italy
| | - Simona Bo
- Department of Medical Sciences, University of Turin, c.so AM Dogliotti 14, 10126, Turin, Italy.
| |
Collapse
|
26
|
Goyal M, Goel A, Singh R, Chowdhury N, Verma N, Tiwari S, Deepak KK. Circadian rhythm of airways caliber and its autonomic modulation. Chronobiol Int 2020; 37:845-855. [PMID: 32077322 DOI: 10.1080/07420528.2020.1731525] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The autonomic nervous system (ANS) is one of the effector pathways for circadian variation of many physiological parameters. Autonomic tone and airways caliber have been reported to exhibit circadian variation in separate studies. A simultaneous investigation of heart rate variability (HRV) and airway caliber might ascertain how airway caliber is modulated by autonomic tone. This study was planned to identify the variations in airway caliber and autonomic function tone during a 24-hour span. A total of 56 healthy male subjects with almost similar daily routines were studied. Time domain, frequency domain and nonlinear analysis of R-R interval from 5 min electrocardiogram (ECG) was done seven times during the daytime wake span at 3-hour intervals starting at 05:00 h in the morning until 23:00 h in the night. Simultaneously peak expiratory flow rate (PEFR) was determined using a mini Wright's peak flow meter. Rhythmometric analysis was done for PEFR and HRV parameters. Significant circadian variation in low frequency (LF) and high frequency (HF) variance was identified in this group of healthy subjects. The circadian rhythm of LF variance was characterized by a gradual increase and corresponding reciprocal change in HF variance from morning until night. The LF/HF ratio and SD2/SD1 ratio reflecting sympatho-vagal balance showed low to high values from morning to evening. The acrophase of the PEFR temporal pattern is similar to that of LF power and almost opposite in phase to that of HF power. PEFR is positively correlated with LF power. The circadian rhythm of airway caliber co-varies with cardiac autonomic tone. It appears that the temporal pattern of cardiac autonomic tone precedes in time that of airways caliber, thereby suggesting the latter operates under the modulatory effect of the 24-hour pattern in sympatho-vagal balance.
Collapse
Affiliation(s)
- Manish Goyal
- Department of Physiology, All India Institute of Medical Sciences , Bhubaneswar, India
| | - Arun Goel
- Department of Physiology, All India Institute of Medical Sciences , Rishikesh, India
| | - Ruchi Singh
- Department of Physiology, All India Institute of Medical Sciences , Bhopal, India
| | - Nilotpal Chowdhury
- Department of Physiology, All India Institute of Medical Sciences , Rishikesh, India
| | - Narsingh Verma
- Department of Physiology, King George's Medical University , Lucknow, India
| | - Sunita Tiwari
- Department of Physiology, King George's Medical University , Lucknow, India
| | - Kishore Kumar Deepak
- Department of Physiology, All India Institute of Medical Sciences , Delhi, India
| |
Collapse
|
27
|
Pilorz V, Astiz M, Heinen KO, Rawashdeh O, Oster H. The Concept of Coupling in the Mammalian Circadian Clock Network. J Mol Biol 2020; 432:3618-3638. [PMID: 31926953 DOI: 10.1016/j.jmb.2019.12.037] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 12/22/2019] [Accepted: 12/23/2019] [Indexed: 12/13/2022]
Abstract
The circadian clock network regulates daily rhythms in mammalian physiology and behavior to optimally adapt the organism to the 24-h day/night cycle. A central pacemaker, the hypothalamic suprachiasmatic nucleus (SCN), coordinates subordinate cellular oscillators in the brain, as well as in peripheral organs to align with each other and external time. Stability and coordination of this vast network of cellular oscillators is achieved through different levels of coupling. Although coupling at the molecular level and across the SCN is well established and believed to define its function as pacemaker structure, the notion of coupling in other tissues and across the whole system is less well understood. In this review, we describe the different levels of coupling in the mammalian circadian clock system - from molecules to the whole organism. We highlight recent advances in gaining knowledge of the complex organization and function of circadian network regulation and its significance for the generation of stable but plastic intrinsic 24-h rhythms.
Collapse
Affiliation(s)
- Violetta Pilorz
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behavior and Metabolism, Marie-Curie-Strasse, 23562, Luebeck, Germany
| | - Mariana Astiz
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behavior and Metabolism, Marie-Curie-Strasse, 23562, Luebeck, Germany
| | - Keno Ole Heinen
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behavior and Metabolism, Marie-Curie-Strasse, 23562, Luebeck, Germany
| | - Oliver Rawashdeh
- The University of Queensland, School of Biomedical Sciences, Faculty of Medicine, St Lucia Qld, 4071, Australia
| | - Henrik Oster
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behavior and Metabolism, Marie-Curie-Strasse, 23562, Luebeck, Germany.
| |
Collapse
|
28
|
Szechtman H, Harvey BH, Woody EZ, Hoffman KL. The Psychopharmacology of Obsessive-Compulsive Disorder: A Preclinical Roadmap. Pharmacol Rev 2020; 72:80-151. [PMID: 31826934 DOI: 10.1124/pr.119.017772] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
This review evaluates current knowledge about obsessive-compulsive disorder (OCD), with the goal of providing a roadmap for future directions in research on the psychopharmacology of the disorder. It first addresses issues in the description and diagnosis of OCD, including the structure, measurement, and appropriate description of the disorder and issues of differential diagnosis. Current pharmacotherapies for OCD are then reviewed, including monotherapy with serotonin reuptake inhibitors and augmentation with antipsychotic medication and with psychologic treatment. Neuromodulatory therapies for OCD are also described, including psychosurgery, deep brain stimulation, and noninvasive brain stimulation. Psychotherapies for OCD are then reviewed, focusing on behavior therapy, including exposure and response prevention and cognitive therapy, and the efficacy of these interventions is discussed, touching on issues such as the timing of sessions, the adjunctive role of pharmacotherapy, and the underlying mechanisms. Next, current research on the neurobiology of OCD is examined, including work probing the role of various neurotransmitters and other endogenous processes and etiology as clues to the neurobiological fault that may underlie OCD. A new perspective on preclinical research is advanced, using the Research Domain Criteria to propose an adaptationist viewpoint that regards OCD as the dysfunction of a normal motivational system. A systems-design approach introduces the security motivation system (SMS) theory of OCD as a framework for research. Finally, a new perspective on psychopharmacological research for OCD is advanced, exploring three approaches: boosting infrastructure facilities of the brain, facilitating psychotherapeutic relearning, and targeting specific pathways of the SMS network to fix deficient SMS shut-down processes. SIGNIFICANCE STATEMENT: A significant proportion of patients with obsessive-compulsive disorder (OCD) do not achieve remission with current treatments, indicating the need for innovations in psychopharmacology for the disorder. OCD may be conceptualized as the dysfunction of a normal, special motivation system that evolved to manage the prospect of potential danger. This perspective, together with a wide-ranging review of the literature, suggests novel directions for psychopharmacological research, including boosting support systems of the brain, facilitating relearning that occurs in psychotherapy, and targeting specific pathways in the brain that provide deficient stopping processes in OCD.
Collapse
Affiliation(s)
- Henry Szechtman
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada (H.S.); SAMRC Unit on Risk Resilience in Mental Disorders, Department of Psychiatry, University of Cape Town, and Center of Excellence for Pharmaceutical Sciences, School of Pharmacy, North-West University (Potchefstroom Campus), Potchefstroom, South Africa (B.H.H.); Department of Psychology, University of Waterloo, Waterloo, Ontario, Canada (E.Z.W.); and Centro de Investigación en Reproducción Animal, CINVESTAV-Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico (K.L.H.)
| | - Brian H Harvey
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada (H.S.); SAMRC Unit on Risk Resilience in Mental Disorders, Department of Psychiatry, University of Cape Town, and Center of Excellence for Pharmaceutical Sciences, School of Pharmacy, North-West University (Potchefstroom Campus), Potchefstroom, South Africa (B.H.H.); Department of Psychology, University of Waterloo, Waterloo, Ontario, Canada (E.Z.W.); and Centro de Investigación en Reproducción Animal, CINVESTAV-Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico (K.L.H.)
| | - Erik Z Woody
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada (H.S.); SAMRC Unit on Risk Resilience in Mental Disorders, Department of Psychiatry, University of Cape Town, and Center of Excellence for Pharmaceutical Sciences, School of Pharmacy, North-West University (Potchefstroom Campus), Potchefstroom, South Africa (B.H.H.); Department of Psychology, University of Waterloo, Waterloo, Ontario, Canada (E.Z.W.); and Centro de Investigación en Reproducción Animal, CINVESTAV-Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico (K.L.H.)
| | - Kurt Leroy Hoffman
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada (H.S.); SAMRC Unit on Risk Resilience in Mental Disorders, Department of Psychiatry, University of Cape Town, and Center of Excellence for Pharmaceutical Sciences, School of Pharmacy, North-West University (Potchefstroom Campus), Potchefstroom, South Africa (B.H.H.); Department of Psychology, University of Waterloo, Waterloo, Ontario, Canada (E.Z.W.); and Centro de Investigación en Reproducción Animal, CINVESTAV-Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico (K.L.H.)
| |
Collapse
|
29
|
Wahl S, Engelhardt M, Schaupp P, Lappe C, Ivanov IV. The inner clock-Blue light sets the human rhythm. JOURNAL OF BIOPHOTONICS 2019; 12:e201900102. [PMID: 31433569 PMCID: PMC7065627 DOI: 10.1002/jbio.201900102] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 07/02/2019] [Accepted: 08/18/2019] [Indexed: 05/06/2023]
Abstract
Visible light synchronizes the human biological clock in the suprachiasmatic nuclei of the hypothalamus to the solar 24-hour cycle. Short wavelengths, perceived as blue color, are the strongest synchronizing agent for the circadian system that keeps most biological and psychological rhythms internally synchronized. Circadian rhythm is important for optimum function of organisms and circadian sleep-wake disruptions or chronic misalignment often may lead to psychiatric and neurodegenerative illness. The beneficial effect on circadian synchronization, sleep quality, mood, and cognitive performance depends not only on the light spectral composition but also on the timing of exposure and its intensity. Exposure to blue light during the day is important to suppress melatonin secretion, the hormone that is produced by the pineal gland and plays crucial role in circadian rhythm entrainment. While the exposure to blue is important for keeping organism's wellbeing, alertness, and cognitive performance during the day, chronic exposure to low-intensity blue light directly before bedtime, may have serious implications on sleep quality, circadian phase and cycle durations. This rises inevitably the need for solutions to improve wellbeing, alertness, and cognitive performance in today's modern society where exposure to blue light emitting devices is ever increasing.
Collapse
Affiliation(s)
- Siegfried Wahl
- Institute for Ophthalmic ResearchUniversity of TuebingenTuebingenGermany
- Carl Zeiss Vision International GmbHAalenGermany
| | - Moritz Engelhardt
- Institute for Ophthalmic ResearchUniversity of TuebingenTuebingenGermany
| | | | | | - Iliya V. Ivanov
- Institute for Ophthalmic ResearchUniversity of TuebingenTuebingenGermany
- Carl Zeiss Vision International GmbHAalenGermany
| |
Collapse
|
30
|
Circadian neurogenetics of mood disorders. Cell Tissue Res 2019; 377:81-94. [DOI: 10.1007/s00441-019-03033-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 04/09/2019] [Indexed: 02/07/2023]
|
31
|
2,3,7,8-Tetrachlorodibenzo-p-dioxin abolishes circadian regulation of hepatic metabolic activity in mice. Sci Rep 2019; 9:6514. [PMID: 31015483 PMCID: PMC6478849 DOI: 10.1038/s41598-019-42760-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 04/05/2019] [Indexed: 12/19/2022] Open
Abstract
Aryl hydrocarbon receptor (AhR) activation is reported to alter the hepatic expression of circadian clock regulators, however the impact on clock-controlled metabolism has not been thoroughly investigated. This study examines the effects of AhR activation on hepatic transcriptome and metabolome rhythmicity in male C57BL/6 mice orally gavaged with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) every 4 days for 28 days. TCDD diminished the rhythmicity of several core clock regulators (e.g. Arntl, Clock, Nr1d1, Per1, Cry1, Nfil3) in a dose-dependent manner, involving either a ≥ 3.3-fold suppression in amplitude or complete loss of oscillation. Accordingly, protein levels (ARNTL, REV-ERBα, NFIL3) and genomic binding (ARNTL) of select regulators were reduced and arrhythmic following treatment. As a result, the oscillating expression of 99.6% of 5,636 clock-controlled hepatic genes was abolished including genes associated with the metabolism of lipids, glucose/glycogen, and heme. For example, TCDD flattened expression of the rate-limiting enzymes in both gluconeogenesis (Pck1) and glycogenesis (Gys2), consistent with the depletion and loss of rhythmicity in hepatic glycogen levels. Examination of polar hepatic extracts by untargeted mass spectrometry revealed that virtually all oscillating metabolites lost rhythmicity following treatment. Collectively, these results suggest TCDD disrupted circadian regulation of hepatic metabolism, altering metabolic efficiency and energy storage.
Collapse
|
32
|
Abstract
Light, through its non-imaging forming effects, plays a dominant role on a myriad of physiological functions, including the human sleep–wake cycle. The non-image forming effects of light heavily rely on specific properties such as intensity, duration, timing, pattern, and wavelengths. Here, we address how specific properties of light influence sleep and wakefulness in humans through acute effects, e.g., on alertness, and/or effects on the circadian timing system. Of critical relevance, we discuss how different characteristics of light exposure across the 24-h day can lead to changes in sleep–wake timing, sleep propensity, sleep architecture, and sleep and wake electroencephalogram (EEG) power spectra. Ultimately, knowledge on how light affects sleep and wakefulness can improve light settings at home and at the workplace to improve health and well-being and optimize treatments of chronobiological disorders.
Collapse
|
33
|
Hardeland R. Aging, Melatonin, and the Pro- and Anti-Inflammatory Networks. Int J Mol Sci 2019; 20:ijms20051223. [PMID: 30862067 PMCID: PMC6429360 DOI: 10.3390/ijms20051223] [Citation(s) in RCA: 224] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 03/05/2019] [Accepted: 03/07/2019] [Indexed: 12/17/2022] Open
Abstract
Aging and various age-related diseases are associated with reductions in melatonin secretion, proinflammatory changes in the immune system, a deteriorating circadian system, and reductions in sirtuin-1 (SIRT1) activity. In non-tumor cells, several effects of melatonin are abolished by inhibiting SIRT1, indicating mediation by SIRT1. Melatonin is, in addition to its circadian and antioxidant roles, an immune stimulatory agent. However, it can act as either a pro- or anti-inflammatory regulator in a context-dependent way. Melatonin can stimulate the release of proinflammatory cytokines and other mediators, but also, under different conditions, it can suppress inflammation-promoting processes such as NO release, activation of cyclooxygenase-2, inflammasome NLRP3, gasdermin D, toll-like receptor-4 and mTOR signaling, and cytokine release by SASP (senescence-associated secretory phenotype), and amyloid-β toxicity. It also activates processes in an anti-inflammatory network, in which SIRT1 activation, upregulation of Nrf2 and downregulation of NF-κB, and release of the anti-inflammatory cytokines IL-4 and IL-10 are involved. A perhaps crucial action may be the promotion of macrophage or microglia polarization in favor of the anti-inflammatory phenotype M2. In addition, many factors of the pro- and anti-inflammatory networks are subject to regulation by microRNAs that either target mRNAs of the respective factors or upregulate them by targeting mRNAs of their inhibitor proteins.
Collapse
Affiliation(s)
- Rüdiger Hardeland
- Johann Friedrich Blumenbach Institute of Zoology and Anthropology, University of Göttingen, 37073 Göttingen, Germany.
| |
Collapse
|
34
|
Mechanisms of Communication in the Mammalian Circadian Timing System. Int J Mol Sci 2019; 20:ijms20020343. [PMID: 30650649 PMCID: PMC6359556 DOI: 10.3390/ijms20020343] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/08/2019] [Accepted: 01/10/2019] [Indexed: 12/17/2022] Open
Abstract
24-h rhythms in physiology and behaviour are organized by a body-wide network of endogenous circadian clocks. In mammals, a central pacemaker in the hypothalamic suprachiasmatic nucleus (SCN) integrates external light information to adapt cellular clocks in all tissues and organs to the external light-dark cycle. Together, central and peripheral clocks co-regulate physiological rhythms and functions. In this review, we outline the current knowledge about the routes of communication between the environment, the main pacemakers and the downstream clocks in the body, focusing on what we currently know and what we still need to understand about the communication mechanisms by which centrally and peripherally controlled timing signals coordinate physiological functions and behaviour. We highlight recent findings that shed new light on the internal organization and function of the SCN and neuroendocrine mechanisms mediating clock-to-clock coupling. These findings have implications for our understanding of circadian network entrainment and for potential manipulations of the circadian clock system in therapeutic settings.
Collapse
|
35
|
Rao RV. Ayurveda and the science of aging. J Ayurveda Integr Med 2018; 9:225-232. [PMID: 29276113 PMCID: PMC6148064 DOI: 10.1016/j.jaim.2017.10.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 10/13/2017] [Accepted: 10/16/2017] [Indexed: 02/01/2023] Open
Abstract
Since time immemorial, humanity has been concerned with developing and preserving youthful vigor, and extending longevity by stopping or delaying the aging process. By 2030, one in five of the world population will be over 65 years old. Longevity and old age are accompanied with a variety of health challenges and population studies indicate that the elderly will use between three to five times more healthcare services compared to the younger population. Modern medicine has made a great deal of progress in understanding the aging process and in controlling age-associated health issues including heart attacks, strokes, diabetes, cancer, senility, and arthritis. Thus, every individual is now looking forward to a youthful, productive lifespan of 100 or more years filled with unlimited health and opportunity. Research by aging experts is focused on ways to go against the natural order of the aging process in order to delay it. Interventions include among other things anti-aging pills, restricted food consumption and cloning body parts to stay young and delay biological aging. Ayurveda, one of the world's most authoritative mind-body-spirit medicinal systems, offers various concepts of the aging process. This system of medicine includes therapies for healthy aging so as to create an optimal health and lengthen an individual's healthspan by living in harmony with nature. This review will explore various aspects of aging and longevity by comparing the science of aging as defined by modern medicine with the Ayurvedic treatise of Jara and Vriddhavastha.
Collapse
Affiliation(s)
- Rammohan V Rao
- The Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945, USA; Faculty, California College of Ayurveda, 700 Zion Street, Nevada City, CA 95959, USA.
| |
Collapse
|
36
|
Masocha W, Kristensson K. Human African trypanosomiasis: How do the parasites enter and cause dysfunctions of the nervous system in murine models? Brain Res Bull 2018; 145:18-29. [PMID: 29870779 DOI: 10.1016/j.brainresbull.2018.05.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 05/24/2018] [Accepted: 05/30/2018] [Indexed: 12/27/2022]
Abstract
In this review we describe how Trypanosoma brucei brucei, a rodent pathogenic strain of African trypanosomes, can invade the nervous system, first by localization to the choroid plexus, the circumventricular organs (CVOs) and peripheral ganglia, which have fenestrated vessels, followed by crossing of the blood-brain barrier (BBB) into the white matter, hypothalamus, thalamus and basal ganglia. White blood cells (WBCs) pave the way for the trypanosome neuroinvasion. Experiments with immune deficient mice show that the invasion of WBCs is initiated by the toll-like receptor 9, followed by an augmentation phase that depends on the cytokine IFN-γ and the chemokine CXCL10. Nitric oxide (NO) derived from iNOS then prevents a break-down of the BBB and non-regulated passage of cells. This chain of events is relevant for design of better diagnostic tools to distinguish the different stages of the disease as well as for better understanding of the pathogenesis of the nervous system dysfunctions, which include circadian rhythm changes with sleep pattern disruption, pain syndromes, movement disorders and mental disturbances including dementia.
Collapse
Affiliation(s)
- Willias Masocha
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Kuwait University, Kuwait.
| | | |
Collapse
|
37
|
Cavalcanti-Ferreira P, Berk L, Daher N, Campus T, Araujo J, Petrofsky J, Lohman E. A nonparametric methodological analysis of rest-activity rhythm in type 2 diabetes. ACTA ACUST UNITED AC 2018; 11:281-289. [PMID: 30746047 PMCID: PMC6361299 DOI: 10.5935/1984-0063.20180044] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The purpose of this study was to analyze the rest-activity rhythm of type 2
diabetics mellitus (T2DM) and compare it to healthy controls using the
nonparametric analytic approach. Twenty-one diabetics and 21 healthy subjects
matched for gender and age were recruited to participate in the study. Data were
analyzed using the Independent t-test, Mann-Whitney U test, and Spearmans
correlation. T2DM subjects demonstrate lower interdaily stability (IS)
(p=.03), higher intradaily variability
(p=.046) and lower rhythm amplitude (p=.02)
when compared to healthy controls. Also, there was a positive correlation
between IS and most active 10 hr (M10) in the average of 24 hours pattern (r
=.44; p=.046) in the diabetes group and a negative correlation
between IV and M10 in the healthy group (r =-.57; p=.007).
These data together suggest that T2DM exhibits a dysfunction in the
rest-activity rhythm due to alterations in the circadian function as well as in
the homeostatic capacity to maintain sleep; mainly characterized by less
consistency across days of the daily circadian signal, higher rhythm
fragmentation and lower rhythm amplitude. Future approaches may be developed
considering the influence of circadian glucose variations throughout the day on
the coupling of the rest-activity rhythm to zeitgeber and rhythm
fragmentation.
Collapse
Affiliation(s)
- Paula Cavalcanti-Ferreira
- Loma Linda University, Physical Therapy - Loma Linda - CA - United States.,California State University, Northridge - CA - United States
| | - Lee Berk
- Loma Linda University, Physical Therapy - Loma Linda - CA - United States
| | - Noha Daher
- Loma Linda University, Physical Therapy - Loma Linda - CA - United States
| | - Tania Campus
- Universidade Federal do Rio Grande do Norte, Physical Therapy - Natal - RN - Brazil
| | - John Araujo
- Universidade Federal do Rio Grande do Norte, Physical Therapy - Natal - RN - Brazil
| | - Jerrold Petrofsky
- Loma Linda University, Physical Therapy - Loma Linda - CA - United States
| | - Everett Lohman
- Loma Linda University, Physical Therapy - Loma Linda - CA - United States
| |
Collapse
|
38
|
Acuña-Castroviejo D, Rahim I, Acuña-Fernández C, Fernández-Ortiz M, Solera-Marín J, Sayed RKA, Díaz-Casado ME, Rusanova I, López LC, Escames G. Melatonin, clock genes and mitochondria in sepsis. Cell Mol Life Sci 2017; 74:3965-3987. [PMID: 28785808 PMCID: PMC11107653 DOI: 10.1007/s00018-017-2610-1] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 08/03/2017] [Indexed: 12/22/2022]
Abstract
After the characterization of the central pacemaker in the suprachiasmatic nucleus, the expression of clock genes was identified in several peripheral tissues including the immune system. The hierarchical control from the central clock to peripheral clocks extends to other functions including endocrine, metabolic, immune, and mitochondrial responses. Increasing evidence links the disruption of the clock genes expression with multiple diseases and aging. Chronodisruption is associated with alterations of the immune system, immunosenescence, impairment of energy metabolism, and reduction of pineal and extrapineal melatonin production. Regarding sepsis, a condition coursing with an exaggerated response of innate immunity, experimental and clinical data showed an alteration of circadian rhythms that reflects the loss of the normal oscillation of the clock. Moreover, recent data point to that some mediators of the immune system affects the normal function of the clock. Under specific conditions, this control disappears reactivating the immune response. So, it seems that clock gene disruption favors the innate immune response, which in turn induces the expression of proinflammatory mediators, causing a further alteration of the clock. Here, the clock control of the mitochondrial function turns off, leading to a bioenergetic decay and formation of reactive oxygen species that, in turn, activate the inflammasome. This arm of the innate immunity is responsible for the huge increase of interleukin-1β and entrance into a vicious cycle that could lead to the death of the patient. The broken clock is recovered by melatonin administration, that is accompanied by the normalization of the innate immunity and mitochondrial homeostasis. Thus, this review emphasizes the connection between clock genes, innate immunity and mitochondria in health and sepsis, and the role of melatonin to maintain clock homeostasis.
Collapse
Affiliation(s)
- Darío Acuña-Castroviejo
- Departamento de Fisiología, Facultad de Medicina, Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Avenida del Conocimiento s/n, 18016, Granada, Spain.
- CIBERfes, Ibs.Granada, and UGC de Laboratorios Clínicos, Complejo Hospitalario de Granada, Granada, Spain.
| | - Ibtissem Rahim
- Departamento de Fisiología, Facultad de Medicina, Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Avenida del Conocimiento s/n, 18016, Granada, Spain
- Département de Biologie et Physiologie Cellulaire, Faculté des Sciences de la Nature et de la Vie, Université Blida 1, Blida, Algeria
| | - Carlos Acuña-Fernández
- Unidad of Anestesiología y Reanimación, Hospital Universitario de Canarias, San Cristóbal de La Laguna, Santa Cruz de Tenerife, Spain
| | - Marisol Fernández-Ortiz
- Departamento de Fisiología, Facultad de Medicina, Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Avenida del Conocimiento s/n, 18016, Granada, Spain
| | - Jorge Solera-Marín
- Unidad of Anestesiología y Reanimación, Hospital Universitario de Canarias, San Cristóbal de La Laguna, Santa Cruz de Tenerife, Spain
| | - Ramy K A Sayed
- Departamento de Fisiología, Facultad de Medicina, Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Avenida del Conocimiento s/n, 18016, Granada, Spain
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Sohag University, Sohâg, Egypt
| | - María E Díaz-Casado
- Departamento de Fisiología, Facultad de Medicina, Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Avenida del Conocimiento s/n, 18016, Granada, Spain
| | - Iryna Rusanova
- Departamento de Fisiología, Facultad de Medicina, Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Avenida del Conocimiento s/n, 18016, Granada, Spain
- CIBERfes, Ibs.Granada, and UGC de Laboratorios Clínicos, Complejo Hospitalario de Granada, Granada, Spain
| | - Luis C López
- Departamento de Fisiología, Facultad de Medicina, Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Avenida del Conocimiento s/n, 18016, Granada, Spain
- CIBERfes, Ibs.Granada, and UGC de Laboratorios Clínicos, Complejo Hospitalario de Granada, Granada, Spain
| | - Germaine Escames
- Departamento de Fisiología, Facultad de Medicina, Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Avenida del Conocimiento s/n, 18016, Granada, Spain
- CIBERfes, Ibs.Granada, and UGC de Laboratorios Clínicos, Complejo Hospitalario de Granada, Granada, Spain
| |
Collapse
|
39
|
Blancas-Velazquez A, la Fleur SE, Mendoza J. Effects of a free-choice high-fat high-sugar diet on brain PER2 and BMAL1 protein expression in mice. Appetite 2017; 117:263-269. [PMID: 28687372 DOI: 10.1016/j.appet.2017.07.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 06/30/2017] [Accepted: 07/02/2017] [Indexed: 01/04/2023]
Abstract
The suprachiasmatic nucleus (SCN) times the daily rhythms of behavioral processes including feeding. Beyond the SCN, the hypothalamic arcuate nucleus (ARC), involved in feeding regulation and metabolism, and the epithalamic lateral habenula (LHb), implicated in reward processing, show circadian rhythmic activity. These brain oscillators are functionally coupled to coordinate the daily rhythm of food intake. In rats, a free choice high-fat high-sugar (fcHFHS) diet leads to a rapid increase of calorie intake and body weight gain. Interestingly, under a fcHFHS condition, rats ingest a similar amount of sugar during day time (rest phase) as during night time (active phase), but keep the rhythmic intake of regular chow-food. The out of phase between feeding patterns of regular (chow) and highly rewarding food (sugar) may involve alterations of brain circadian oscillators regulating feeding. Here, we report that the fcHFHS diet is a successful model to induce calorie intake, body weight gain and fat tissue accumulation in mice, extending its effectiveness as previously reported in rats. Moreover, we observed that whereas in the SCN the day-night difference in the PER2 clock protein expression was similar between chow-fed and fcHFHS-fed animals, in the LHb, this day-night difference was altered in fcHFHS-exposed animals compared to control chow mice. These findings confirm previous observations in rats showing disrupted daily patterns of feeding behavior under a fcHFHS diet exposure, and extend our insights on the effects of the diet on circadian gene expression in brain clocks.
Collapse
Affiliation(s)
- Aurea Blancas-Velazquez
- Institute of Cellular and Integrative Neurosciences, CNRS UPR-3212, University of Strasbourg, France; Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Susanne E la Fleur
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Jorge Mendoza
- Institute of Cellular and Integrative Neurosciences, CNRS UPR-3212, University of Strasbourg, France.
| |
Collapse
|
40
|
Fernández Blanco A, Andrini L, Errecalde AL, Inda AM, García M. Circadian rhythm of VEGF expression in the liver of hepatectomized-tumor-bearing adult and young mice. BIOL RHYTHM RES 2017. [DOI: 10.1080/09291016.2017.1377983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Ayelén Fernández Blanco
- Facultad de Ciencias Médicas, Cátedra de Citología, Histología y Embriología “A”, Universidad Nacional de La Plata, Buenos Aires, Argentina
| | - Laura Andrini
- Facultad de Ciencias Médicas, Cátedra de Citología, Histología y Embriología “A”, Universidad Nacional de La Plata, Buenos Aires, Argentina
| | - Ana Lía Errecalde
- Facultad de Ciencias Médicas, Cátedra de Citología, Histología y Embriología “A”, Universidad Nacional de La Plata, Buenos Aires, Argentina
| | - Ana María Inda
- Facultad de Ciencias Médicas, Cátedra de Citología, Histología y Embriología “A”, Universidad Nacional de La Plata, Buenos Aires, Argentina
- CIC, Buenos Aires, Argentina
| | - Marcela García
- Facultad de Ciencias Médicas, Cátedra de Citología, Histología y Embriología “A”, Universidad Nacional de La Plata, Buenos Aires, Argentina
| |
Collapse
|
41
|
Buijs FN, León-Mercado L, Guzmán-Ruiz M, Guerrero-Vargas NN, Romo-Nava F, Buijs RM. The Circadian System: A Regulatory Feedback Network of Periphery and Brain. Physiology (Bethesda) 2017; 31:170-81. [PMID: 27053731 DOI: 10.1152/physiol.00037.2015] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Circadian rhythms are generated by the autonomous circadian clock, the suprachiasmatic nucleus (SCN), and clock genes that are present in all tissues. The SCN times these peripheral clocks, as well as behavioral and physiological processes. Recent studies show that frequent violations of conditions set by our biological clock, such as shift work, jet lag, sleep deprivation, or simply eating at the wrong time of the day, may have deleterious effects on health. This infringement, also known as circadian desynchronization, is associated with chronic diseases like diabetes, hypertension, cancer, and psychiatric disorders. In this review, we will evaluate evidence that these diseases stem from the need of the SCN for peripheral feedback to fine-tune its output and adjust physiological processes to the requirements of the moment. This feedback can vary from neuronal or hormonal signals from the liver to changes in blood pressure. Desynchronization renders the circadian network dysfunctional, resulting in a breakdown of many functions driven by the SCN, disrupting core clock rhythms in the periphery and disorganizing cellular processes that are normally driven by the synchrony between behavior and peripheral signals with neuronal and humoral output of the hypothalamus. Consequently, we propose that the loss of synchrony between the different elements of this circadian network as may occur during shiftwork and jet lag is the reason for the occurrence of health problems.
Collapse
Affiliation(s)
- Frederik N Buijs
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico; Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Luis León-Mercado
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico
| | - Mara Guzmán-Ruiz
- Departamento de Anatomía, Facultad de Medicina, Universidad Autónoma de México, Ciudad Universitaria, Mexico
| | - Natali N Guerrero-Vargas
- Departamento de Anatomía, Facultad de Medicina, Universidad Autónoma de México, Ciudad Universitaria, Mexico
| | - Francisco Romo-Nava
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico; Department of Psychiatry and Behavioral Neuroscience, Division of Bipolar Disorder Research, University of Cincinnati, Cincinnati, Ohio; and
| | - Ruud M Buijs
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico;
| |
Collapse
|
42
|
Porfirio MC, Gomes de Almeida JP, Stornelli M, Giovinazzo S, Purper-Ouakil D, Masi G. Can melatonin prevent or improve metabolic side effects during antipsychotic treatments? Neuropsychiatr Dis Treat 2017; 13:2167-2174. [PMID: 28860773 PMCID: PMC5560235 DOI: 10.2147/ndt.s127564] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In the last two decades, second-generation antipsychotics (SGAs) were more frequently used than typical antipsychotics for treating both psychotic and nonpsychotic psychiatric disorders in both children and adolescents, because of their lower risk of adverse neurological effects, that is, extrapyramidal symptoms. Recent studies have pointed out their effect on weight gain and increased visceral adiposity as they induce metabolic syndrome. Patients receiving SGAs often need to be treated with other substances to counteract metabolic side effects. In this paper, we point out the possible protective effect of add-on melatonin treatment in preventing, mitigating, or even reversing SGAs metabolic effects, improving quality of life and providing safer long-term treatments in pediatric patients. Melatonin is an endogenous indolamine secreted during darkness by the pineal gland; it plays a key role in regulating the circadian rhythm, generated by the suprachiasmatic nuclei (SCN) of the hypothalamus, and has many other biological functions, including chronobiotic, antioxidant and neuroprotective properties, anti-inflammatory and free radical scavenging effects, and diminishing oxidative injury and fat distribution. It has been hypothesized that SGAs cause adverse metabolic effects that may be restored by nightly administration of melatonin because of its influence on autonomic and hormonal outputs. Interestingly, atypical anti-psychotics (AAPs) can cause several sleep disorders, and circadian misalignment can influence hormones involved in the metabolic regulation, such as insulin, leptin, and ghrelin; furthermore, a relationship between obesity and sleep curtailment has been demonstrated, as well as sleep deprivation in rats has been associated with hyperphagia. Metabolic effects of melatonin, both central and peripheral, direct and indirect, target most metabolic disorders reported during and after SGA treatment in children, adolescents, and adults. Further systematic studies on psychiatric patients are needed to explore the effect of add-on melatonin on metabolic side effects of SGAs, independent of energy intake, diet, and exercise.
Collapse
Affiliation(s)
| | | | - Maddalena Stornelli
- Unit of Child Neurology and Psychiatry, "Tor Vergata" University of Rome, Italy
| | - Silvia Giovinazzo
- Unit of Child Neurology and Psychiatry, "Tor Vergata" University of Rome, Italy
| | - Diane Purper-Ouakil
- Unit of Child and Adolescent Psychiatry, Saint Eloi Hospital, Montpellier, France
| | - Gabriele Masi
- IRCCS Stella Maris, Scientific Institute of Child Neurology and Psychiatry, Calambrone, Pisa, Italy
| |
Collapse
|
43
|
Abstract
The biological clocks of the circadian timing system coordinate cellular and physiological processes and synchronizes these with daily cycles, feeding patterns also regulates circadian clocks. The clock genes and adipocytokines show circadian rhythmicity. Dysfunction of these genes are involved in the alteration of these adipokines during the development of obesity. Food availability promotes the stimuli associated with food intake which is a circadian oscillator outside of the suprachiasmatic nucleus (SCN). Its circadian rhythm is arranged with the predictable daily mealtimes. Food anticipatory activity is mediated by a self-sustained circadian timing and its principal component is food entrained oscillator. However, the hypothalamus has a crucial role in the regulation of energy balance rather than food intake. Fatty acids or their metabolites can modulate neuronal activity by brain nutrient-sensing neurons involved in the regulation of energy and glucose homeostasis. The timing of three-meal schedules indicates close association with the plasma levels of insulin and preceding food availability. Desynchronization between the central and peripheral clocks by altered timing of food intake and diet composition can lead to uncoupling of peripheral clocks from the central pacemaker and to the development of metabolic disorders. Metabolic dysfunction is associated with circadian disturbances at both central and peripheral levels and, eventual disruption of circadian clock functioning can lead to obesity. While CLOCK expression levels are increased with high fat diet-induced obesity, peroxisome proliferator-activated receptor (PPAR) alpha increases the transcriptional level of brain and muscle ARNT-like 1 (BMAL1) in obese subjects. Consequently, disruption of clock genes results in dyslipidemia, insulin resistance and obesity. Modifying the time of feeding alone can greatly affect body weight. Changes in the circadian clock are associated with temporal alterations in feeding behavior and increased weight gain. Thus, shift work is associated with increased risk for obesity, diabetes and cardio-vascular diseases as a result of unusual eating time and disruption of circadian rhythm.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- , Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
44
|
Williams WPT, McLin DE, Dressman MA, Neubauer DN. Comparative Review of Approved Melatonin Agonists for the Treatment of Circadian Rhythm Sleep-Wake Disorders. Pharmacotherapy 2016; 36:1028-41. [PMID: 27500861 PMCID: PMC5108473 DOI: 10.1002/phar.1822] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Circadian rhythm sleep‐wake disorders (CRSWDs) are characterized by persistent or recurrent patterns of sleep disturbance related primarily to alterations of the circadian rhythm system or the misalignment between the endogenous circadian rhythm and exogenous factors that affect the timing or duration of sleep. These disorders collectively represent a significant unmet medical need, with a total prevalence in the millions, a substantial negative impact on quality of life, and a lack of studied treatments for most of these disorders. Activation of the endogenous melatonin receptors appears to play an important role in setting the circadian clock in the suprachiasmatic nucleus of the hypothalamus. Therefore, melatonin agonists, which may be able to shift and/or stabilize the circadian phase, have been identified as potential therapeutic candidates for the treatment of CRSWDs. Currently, only one melatonin receptor agonist, tasimelteon, is approved for the treatment of a CRSWD: non–24‐hour sleep‐wake disorder (or non‐24). However, three additional commercially available melatonin receptor agonists—agomelatine, prolonged‐release melatonin, and ramelteon—have been investigated for potential use for treatment of CRSWDs. Data indicate that these melatonin receptor agonists have distinct pharmacologic profiles that may help clarify their clinical use in CRSWDs. We review the pharmacokinetic and pharmacodynamic properties of these melatonin agonists and summarize their efficacy profiles when used for the treatment of CRSWDs. Further studies are needed to determine the therapeutic potential of these melatonin agonists for most CRSWDs.
Collapse
Affiliation(s)
| | - Dewey E McLin
- Department of Medical Affairs, Vanda Pharmaceuticals, Inc., Washington, D.C
| | - Marlene A Dressman
- Department of Clinical Development, Vanda Pharmaceuticals, Inc., Washington, D.C
| | - David N Neubauer
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
45
|
When to eat? The influence of circadian rhythms on metabolic health: are animal studies providing the evidence? Nutr Res Rev 2016; 29:180-193. [PMID: 27364352 DOI: 10.1017/s095442241600010x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
As obesity and metabolic diseases rise, there is need to investigate physiological and behavioural aspects associated with their development. Circadian rhythms have a profound influence on metabolic processes, as they prepare the body to optimise energy use and storage. Moreover, food-related signals confer temporal order to organs involved in metabolic regulation. Therefore food intake should be synchronised with the suprachiasmatic nucleus (SCN) to elaborate efficient responses to environmental challenges. Human studies suggest that a loss of synchrony between mealtime and the SCN promotes obesity and metabolic disturbances. Animal research using different paradigms has been performed to characterise the effects of timing of food intake on metabolic profiles. Therefore the purpose of the present review is to critically examine the evidence of animal studies, to provide a state of the art on metabolic findings and to assess whether the paradigms used in rodent models give the evidence to support a 'best time' for food intake. First we analyse and compare the current findings of studies where mealtime has been shifted out of phase from the light-dark cycle. Then, we analyse studies restricting meal times to different moments within the active period. So far animal studies correlate well with human studies, demonstrating that restricting food intake to the active phase limits metabolic disturbances produced by high-energy diets and that eating during the inactive/sleep phase leads to a worse metabolic outcome. Based on the latter we discuss the missing elements and possible mechanisms leading to the metabolic consequences, as these are still lacking.
Collapse
|
46
|
Abstract
The circadian system regulates the timing and expression of nearly all biological processes, most notably, the sleep-wake cycle, and disruption of this system can result in adverse effects on both physical and mental health. The circadian rhythm sleep-wake disorders (CRSWDs) consist of 5 disorders that are due primarily to pathology of the circadian clock or to a misalignment of the timing of the endogenous circadian rhythm with the environment. This article outlines the nature of these disorders, the association of many of these disorders with psychiatric illness, and available treatment options.
Collapse
Affiliation(s)
- Sabra M Abbott
- Department of Neurology, Northwestern University Feinberg School of Medicine, 710 North Lake Shore Drive, Suite 500, Chicago, IL 60611, USA
| | - Kathryn J Reid
- Department of Neurology, Northwestern University Feinberg School of Medicine, 710 North Lake Shore Drive, Suite 500, Chicago, IL 60611, USA
| | - Phyllis C Zee
- Department of Neurology, Northwestern University Feinberg School of Medicine, 710 North Lake Shore Drive, Suite 500, Chicago, IL 60611, USA.
| |
Collapse
|
47
|
Emens JS, Burgess HJ. Effect of Light and Melatonin and Other Melatonin Receptor Agonists on Human Circadian Physiology. Sleep Med Clin 2015; 10:435-53. [PMID: 26568121 DOI: 10.1016/j.jsmc.2015.08.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Circadian (body clock) timing has a profound influence on mental health, physical health, and health behaviors. This review focuses on how light, melatonin, and other melatonin receptor agonist drugs can be used to shift circadian timing in patients with misaligned circadian rhythms. A brief overview of the human circadian system is provided, followed by a discussion of patient characteristics and safety considerations that can influence the treatment of choice. The important features of light treatment, light avoidance, exogenous melatonin, and other melatonin receptor agonists are reviewed, along with some of the practical aspects of light and melatonin treatment.
Collapse
Affiliation(s)
- Jonathan S Emens
- Department of Hospital and Specialty Medicine, Portland VA Medical Center, 3710 SW US Veterans Hospital Road, P3-PULM, Portland, OR 97239, USA; Departments of Psychiatry and Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Helen J Burgess
- Biological Rhythms Research Laboratory, Department of Behavioral Sciences, Rush University Medical Center, 1645 West Jackson Boulevard, Suite 425, Chicago, IL 60612, USA.
| |
Collapse
|
48
|
Garcia M, Andrini L, Martinez M, Inda A, Palma B, Miriuka S, Errecalde AL. Circadian rhythms of proliferation events in two mouse carcinomas. BIOL RHYTHM RES 2015. [DOI: 10.1080/09291016.2015.1034971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
49
|
Lazăr R, Solcan C, Creţu C, Lazăr M, Muntean C, Boişteanu P. Characterization of the relations between morphology and physiological status of the pineal gland in connection with the somatic development level in turkeys reared in Romania. ARQ BRAS MED VET ZOO 2015. [DOI: 10.1590/1678-4162-7111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This research started from the premises of the existence of some possible relationships between indole and pineal peptide hormones and the somatic development, with participation of hypothalamic-pituitary complex. Experimental factors, which were the subject of the present paper, influenced the dynamics of corporal mass and fodder consumption, leading to the occurrence of some important structural modifications at the level of pineal gland. The exposure of the individuals to continuous light (photic pinealectomy) produces increases in corporal mass, showing the involvement of the pineal gland in neuro-endocrine-metabolic reactions, which contributes to the maintenance of homeostatic balance, including somatic ones. Biological material was represented by a number of 50 individuals belonging to B.U.T. Big 6 hybrid, reared on soil, on a permanent litter, which could assure the expanding of knowledge area regarding the relation between rearing technology, modulation of some microclimate parameters and growing performances. Were also realised cytometric and hystometric muscular determinations.
Collapse
|
50
|
Hege A, Perko M, Johnson A, Yu CH, Sönmez S, Apostolopoulos Y. Surveying the Impact of Work Hours and Schedules on Commercial Motor Vehicle Driver Sleep. Saf Health Work 2015; 6:104-13. [PMID: 26106509 PMCID: PMC4476188 DOI: 10.1016/j.shaw.2015.02.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 02/02/2015] [Accepted: 02/03/2015] [Indexed: 12/01/2022] Open
Abstract
Background Given the long hours on the road involving multiple and interacting work stressors (i.e., delivery pressures, irregular shifts, ergonomic hazards), commercial drivers face a plethora of health and safety risks. Researchers goal was to determine whether and to what extent long-haul trucker work schedules influence sleep duration and quality. Methods Survey and biometric data collected from male long-haul truck drivers at a major truckstop in central North Carolina over a six month period. Results Daily hours worked (mean = 11 hours, 55 minutes) and frequency of working over government-mandated daily HOS regulations (23.8% “frequently or always”) were statistically significant predictors of sleep duration. Miles driven per week (mean = 2,812.61), irregular daily hours worked (63.8%), and frequency of working over the daily hour limit (23.8% “frequently or always”) were statistically significant predictors of sleep quality. Conclusion Implications of findings suggest a comprehensive review of the regulations and operational conditions for commercial motor vehicle drivers be undertaken.
Collapse
Affiliation(s)
- Adam Hege
- Department of Public Health Education, University of North Carolina at Greensboro, Greensboro, NS, USA
| | - Michael Perko
- Department of Public Health Education, University of North Carolina at Greensboro, Greensboro, NS, USA
| | - Amber Johnson
- Department of Public Health Education, University of North Carolina at Greensboro, Greensboro, NS, USA
| | - Chong Ho Yu
- Department of Psychology, Azusa Pacific University, Azusa, CA, USA
| | - Sevil Sönmez
- Department of Marketing, Hospitality, & Tourism, University of North Carolina at Greensboro, Greensboro, NS, USA
| | - Yorghos Apostolopoulos
- Department of Public Health Education, University of North Carolina at Greensboro, Greensboro, NS, USA ; Emory University School of Medicine, Atlanta, GA, USA ; Department of Health & Kinesiology, Texas A&M University, TX, USA
| |
Collapse
|