1
|
Duske J, D'Souza N, Mayer D, Dieterich DC, Fendt M. Orexinergic modulation of chronic jet lag-induced deficits in mouse cognitive flexibility. Neuropsychopharmacology 2025; 50:762-771. [PMID: 39478089 PMCID: PMC11914050 DOI: 10.1038/s41386-024-02017-8] [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: 07/09/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 03/19/2025]
Abstract
Cognitive flexibility and working memory are important executive functions mediated by the prefrontal cortex and can be impaired by circadian rhythm disturbances such as chronic jet lag (CJL) or shift work. In the present study, we used mice to investigate whether (1) simulated CJL impairs cognitive flexibility, (2) the orexin system is involved in such impairment, and (3) nasal administration of orexin A is able to reverse CJL-induced deficits in cognitive flexibility and working memory. Mice were exposed to either standard light-dark conditions or simulated CJL consisting of series of advance time shifts. Experiment (1) investigated the effects of a mild CJL protocol on cognitive flexibility using the attentional set shifting task. Experiment (2) used a stronger CJL protocol and examined CJL effects on the orexin system utilizing c-Fos and orexin immunohistochemistry. Experiment (3) tested whether nasal orexin application can rescue CJL-induced deficits in cognitive flexibility and working memory, the latter by measuring spontaneous alternation in the Y-maze. The present data show that CJL (1) impairs cognitive flexibility and (2) reduces the activity of orexin neurons in the lateral hypothalamus. (3) Nasal administration of orexin A rescued CJL-induced deficits in working memory and cognitive flexibility. These findings suggest that executive function impairments by circadian rhythm disturbances such as CJL are caused by dysregulation of orexinergic input to the prefrontal cortex. Compensation of decreased orexinergic input by nasal administration of orexin A could be a potential therapy for CJL- or shift work-induced human deficits in executive functions.
Collapse
Affiliation(s)
- Julius Duske
- Institute for Pharmacology and Toxicology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Nicole D'Souza
- Institute for Pharmacology and Toxicology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
- Institute of Neurophysiology, Goethe University, Frankfurt, Germany
| | - Dana Mayer
- Institute for Pharmacology and Toxicology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Daniela C Dieterich
- Institute for Pharmacology and Toxicology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
- Center of Behavioural Brain Sciences, Otto-von-Guericke University, Magdeburg, Germany
| | - Markus Fendt
- Institute for Pharmacology and Toxicology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany.
- Center of Behavioural Brain Sciences, Otto-von-Guericke University, Magdeburg, Germany.
| |
Collapse
|
2
|
Pugliane KC, Castelo-Branco R, Araújo KKBC, Pereira JC, Leal JCDO, Koike BDV, Fontenele-Araujo J, da Silva CA, Barbosa FF. Dissociation of circadian rhythms in adolescent rats affects object recognition and spatial recognition memories. Physiol Behav 2025; 292:114824. [PMID: 39880271 DOI: 10.1016/j.physbeh.2025.114824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/15/2025] [Accepted: 01/25/2025] [Indexed: 01/31/2025]
Abstract
The T22 protocol is an animal model of forced internal desynchronization, in which rats are exposed to an 11:11 light-dark (LD) cycle. This non-invasive protocol induces the dissociation of circadian rhythms in adult rats, making it possible to study the effects of circadian disruption on physiological and behavioral processes such as learning, memory, and emotional responses. However, the effects of circadian dissociation during other developmental stages, such as adolescence, remain unexplored. Adolescence is a period marked by significant changes in sleep patterns and increased exposure to bright light at night, making it essential to investigate how circadian dissociation may affect this phase of development. This study aimed to evaluate the circadian rhythmicity, cognitive performance and anxiety-like behavior in adolescent Wistar rats under the alignment (aligned T22 group) or misalignment (misaligned T22 group) phases of the T22 cycle. A third group of adolescent rats was maintained in a normal 12:12 LD cycle during the experiment and was used as control group (T24 group). Compared to the control group, adolescent rats under both phases of the T22 cycle exhibited a dissociated circadian rhythm of the locomotor activity and deficits in object recognition memory tasks, without impairments in tasks related to emotional responses. These findings indicate that forced desynchronization impairs recognition memory in adolescent rats, suggesting potential cognitive consequences of internal desynchronization during this critical developmental phase, with relevant implications for public health discussions.
Collapse
Affiliation(s)
- Karen Cristina Pugliane
- Memory and Cognition Studies Laboratory, Department of Psychology, Federal University of Paraiba, João Pessoa, PB, Brazil; Neurobiology and Biologic Rhythmicity Laboratory, Department of Physiology and Behavior, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Rochele Castelo-Branco
- Memory and Cognition Studies Laboratory, Department of Psychology, Federal University of Paraiba, João Pessoa, PB, Brazil
| | | | - Jeane Constantino Pereira
- Memory and Cognition Studies Laboratory, Department of Psychology, Federal University of Paraiba, João Pessoa, PB, Brazil
| | - Júlio César de Oliveira Leal
- Memory and Cognition Studies Laboratory, Department of Psychology, Federal University of Paraiba, João Pessoa, PB, Brazil; Neurobiology and Biologic Rhythmicity Laboratory, Department of Physiology and Behavior, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | | | - John Fontenele-Araujo
- Neurobiology and Biologic Rhythmicity Laboratory, Department of Physiology and Behavior, Federal University of Rio Grande do Norte, Natal, RN, Brazil.
| | - Crhistiane Andressa da Silva
- Memory and Cognition Studies Laboratory, Department of Psychology, Federal University of Paraiba, João Pessoa, PB, Brazil
| | - Flavio Freitas Barbosa
- Memory and Cognition Studies Laboratory, Department of Psychology, Federal University of Paraiba, João Pessoa, PB, Brazil.
| |
Collapse
|
3
|
Chaikin CA, Thakkar AV, Steffeck AWT, Pfrender EM, Hung K, Zhu P, Waldeck NJ, Nozawa R, Song W, Futtner CR, Quattrocelli M, Bass J, Ben-Sahra I, Peek CB. Control of circadian muscle glucose metabolism through the BMAL1-HIF axis in obesity. Proc Natl Acad Sci U S A 2025; 122:e2424046122. [PMID: 40127275 DOI: 10.1073/pnas.2424046122] [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: 11/18/2024] [Accepted: 02/24/2025] [Indexed: 03/26/2025] Open
Abstract
Disruptions of circadian rhythms are widespread in modern society and lead to accelerated and worsened symptoms of metabolic syndrome. In healthy mice, the circadian clock factor BMAL1 is required for skeletal muscle function and metabolism. However, the importance of muscle BMAL1 in the development of metabolic diseases, such as diet-induced obesity (DIO), remains unclear. Here, we demonstrate that skeletal muscle-specific BMAL1-deficient mice exhibit worsened glucose tolerance upon high-fat diet feeding, despite no evidence of increased weight gain. Metabolite profiling from Bmal1-deficient muscles revealed impaired glucose utilization specifically at early steps in glycolysis that dictate the switch between anabolic and catabolic glucose fate. We provide evidence that this is due to abnormal control of the nutrient stress-responsive hypoxia-inducible factor (HIF) pathway. Genetic HIF1α stabilization in muscle Bmal1-deficient mice restores glucose tolerance and expression of 217/736 dysregulated genes during DIO, including glycolytic enzymes. Together, these data indicate that during DIO, skeletal muscle BMAL1 is an important regulator of HIF-driven glycolysis and metabolic flexibility, which influences the development of high-fat-diet-induced glucose intolerance.
Collapse
Affiliation(s)
- Claire A Chaikin
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Abhishek V Thakkar
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Adam W T Steffeck
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Eric M Pfrender
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Kaitlyn Hung
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Pei Zhu
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Nathan J Waldeck
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Rino Nozawa
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Weimin Song
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Christopher R Futtner
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Mattia Quattrocelli
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Joseph Bass
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Issam Ben-Sahra
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Clara B Peek
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| |
Collapse
|
4
|
Delorme TC, Arcego DM, Penichet D, O'Toole N, Huebener N, Silveira PP, Srivastava LK, Cermakian N. Large-scale effects of prenatal inflammation and early life circadian disruption in mice: Implications for neurodevelopmental disorders. Brain Behav Immun 2025:S0889-1591(25)00108-4. [PMID: 40118225 DOI: 10.1016/j.bbi.2025.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 02/17/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025] Open
Abstract
Around 80 % of individuals with neurodevelopmental disorders such as schizophrenia and autism spectrum disorders experience disruptions in sleep/circadian rhythms. We explored whether environmental circadian disruption interacts with prenatal infection, a risk factor for neurodevelopmental disorders, to induce sex-specific deficits in mice. A maternal immune activation (MIA) protocol was used by injecting pregnant mice with a viral mimic poly IC or saline at E9.5. Juvenile/adolescent male and female offspring (3-7 weeks old) were then subjected to a standard light:dark cycle (12:12LD) or to constant light (LL). Significant interactions between treatment (MIA, control) and lighting (12:12LD, LL) were evident in behaviors related to cognition, anxiety, and sociability. This pattern persisted in our RNA sequencing analysis of the dorsal hippocampus, where poly IC exposure resulted in numerous differentially expressed genes (DEGs) in males, while exposure to both poly IC and LL led to a marked reduction in DEGs. Through WGCNA analysis, many significant gene modules were found to be positively associated with poly IC (vs. saline) and LL (vs. LD) in males (fewer in females). Many of the identified hub-bottleneck genes were homologous to human genes associated with sleep/circadian rhythms and neurodevelopmental disorders as revealed by GWA studies. The MIA- and LL-associated modules were enriched in microglia gene signatures, which was paralleled by trends of effects of each of the factors on microglia morphology. In conclusion, in a mouse model of prenatal infection, circadian disruption induced by LL during adolescence acts as a modulator of the effects of MIA at behavioral, cellular, and molecular levels.
Collapse
Affiliation(s)
- Tara C Delorme
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada; Integrated Program in Neuroscience, McGill University, Montréal, Québec H3A 2B4, Canada
| | - Danusa M Arcego
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada; Department of Psychiatry, McGill University, Montréal, Québec H3A 1A1, Canada
| | - Danae Penichet
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada; Integrated Program in Neuroscience, McGill University, Montréal, Québec H3A 2B4, Canada
| | - Nicholas O'Toole
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada
| | - Nikki Huebener
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada
| | - Patrícia P Silveira
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada; Department of Psychiatry, McGill University, Montréal, Québec H3A 1A1, Canada
| | - Lalit K Srivastava
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada; Department of Psychiatry, McGill University, Montréal, Québec H3A 1A1, Canada.
| | - Nicolas Cermakian
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada; Department of Psychiatry, McGill University, Montréal, Québec H3A 1A1, Canada.
| |
Collapse
|
5
|
Huang H, Hou J, Liao Y, Yu J, Xi B. Exposure to nanoplastics exacerbates light pollution hazards to mammalian. ENVIRONMENT INTERNATIONAL 2025; 197:109338. [PMID: 39983414 DOI: 10.1016/j.envint.2025.109338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 01/13/2025] [Accepted: 02/15/2025] [Indexed: 02/23/2025]
Abstract
Environmental light pollution adversely affects brain function, disturbing circadian rhythms and negatively impacting human health. Nanoplastics (NPs) pollution is pervasive in the human environment, and their minuscule size facilitates entry into the body, particularly invading brain and compromising its functionality. However, whether NPs infiltrate rhythm-regulated brain regions and disrupt circadian rhythms in organisms remains unclear. Our study demonstrates that exposure to NPs in mice perturbs normal circadian rhythms. Specifically, NPs invade the suprachiasmatic nucleus (SCN), affecting the circadian clock genes network and altering the regular oscillations of core clock genes. Exposure to NPs renders the intrinsic rhythms more susceptible to disruption by light pollution, resulting in more pronounced disorder to metabolism, immune regulation, and brain function. This work is the first to investigate the combined effects of ambient light pollution and NPs pollution on mammalian health, and our findings suggest that NPs amplify the health impacts of light pollution. These findings also highlight that efforts to mitigate human health risks from environmental pollutants should begin to consider the synergistic effects of various classes of pollutants.
Collapse
Affiliation(s)
- Haipeng Huang
- State Key Laboratory of Environment Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China; Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China; Research Unit of Mitochondria in Brain Diseases, Chinese Academy of Medical Sciences, PKU-Nanjing Institute of Translational Medicine, Nanjing 210061, China
| | - Jiaqi Hou
- State Key Laboratory of Environment Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China.
| | - Yilie Liao
- Duke-NUS Medical School, National University of Singapore, Singapore 169857, Republic of Singapore
| | - Jing Yu
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China; Research Unit of Mitochondria in Brain Diseases, Chinese Academy of Medical Sciences, PKU-Nanjing Institute of Translational Medicine, Nanjing 210061, China
| | - Beidou Xi
- State Key Laboratory of Environment Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China.
| |
Collapse
|
6
|
Balsamo F, Meneo D, Berretta E, Baglioni C, Gelfo F. Could sleep be a brain/cognitive/neural reserve-builder factor? A systematic review on the cognitive effects of sleep modulation in animal models. Neurosci Biobehav Rev 2025; 169:106015. [PMID: 39828234 DOI: 10.1016/j.neubiorev.2025.106015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
The brain/cognitive/neural reserve concept suggests that lifelong experiences, from early life through adulthood, make the brain more resilient to neuronal damage. Modifiable lifestyle factors, such as sleep, can support the development and enhance such a reserve, helping to counteract age- or disease-related brain changes and their impact on cognition. Sleep plays a crucial role in cognitive functioning, and disruptions or disorders may increase neurodegenerative risks. This systematic review aims to explore how functional and disturbed sleep impacts cognitive functions and neuromorphological mechanisms in rodents, aiming to better understand its role in brain/cognitive/neural reserve development. This systematic review, registered on PROSPERO (ID: CRD42023423901) and conducted according to PRISMA-P guidelines, searched PubMed, Scopus, Web of Science, and Embase databases for studies up to June 2022, with terms related to sleep, rodents, and cognitive functions. Of the 28,666 articles identified, 142 met the inclusion criteria. Main results showed significant cognitive decline after sleep deprivation, especially in memory performance. These findings supports the importance of sleep as a critical factor in modulating brain/cognitive/neural reserve.
Collapse
Affiliation(s)
- Francesca Balsamo
- Department of Human Sciences, Guglielmo Marconi University, Rome 00193, Italy; IRCCS Fondazione Santa Lucia, Rome 00179, Italy.
| | - Debora Meneo
- Department of Human Sciences, Guglielmo Marconi University, Rome 00193, Italy
| | | | - Chiara Baglioni
- Department of Human Sciences, Guglielmo Marconi University, Rome 00193, Italy; Department of Clinical Psychology and Psychophysiology/Sleep, Medicine, Centre for Mental Disorders, University Medical Centre, Freiburg, Germany
| | - Francesca Gelfo
- Department of Human Sciences, Guglielmo Marconi University, Rome 00193, Italy; IRCCS Fondazione Santa Lucia, Rome 00179, Italy.
| |
Collapse
|
7
|
Lu H, Ni X, Man Chan SS, Cheng CPW, Chan W, Lam LCW. Pre-treatment subjective sleep quality as a predictive biomarker of tDCS effects in preclinical Alzheimer's disease patients: Secondary analysis of a randomised clinical trial. PLoS One 2025; 20:e0317700. [PMID: 39874376 PMCID: PMC11774347 DOI: 10.1371/journal.pone.0317700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 12/30/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Despite transcranial direct current stimulation (tDCS) has demonstrated encouraging potential for modulating the circadian rhythm, little is known about how well and sustainably tDCS might improve the subjective sleep quality in older adults. This study sought to determine how tDCS affected sleep quality and cognition, as well as how well pre-treatment sleep quality predicted tDCS effects on domain-specific cognitive functions in patients with mild neurocognitive disorder due to Alzheimer's disease (NCD-AD). METHODS This clinical trial aimed to compare the effectiveness of tDCS and cognitive training in mild NCD-AD patients (n = 201). Over the course of four weeks, patients were randomized to receive either tDCS plus working memory training, or sham tDCS plus working memory training, or tDCS plus controlled cognitive training. The Pittsburgh Sleep Quality Index (PSQI) was used to measured subjective sleep quality. The Alzheimer's disease assessment scale-cognitive subscale (ADAS-Cog) was used to evaluate domain-specific cognitive functions. RESULTS Recurrent tDCS treatments enhanced subjective sleep quality and cognition considerably. The poor sleepers (i.e., PSQI > 5) who received tDCS treatment had more cognitive benefits (p = 0.031, Cohen's d = 0.605) and sleep improvements (p < 0.001, Cohen's d = 1.209) in comparison to cognitive training. Pre-treatment subjective sleep quality was linked to tDCS-induced improvement in memory function. CONCLUSION During the course of two months, repeated tDCS could considerably enhance subjective sleep quality. For the cognitive benefits of the treatments, the status of pre-treatment subjective sleep quality is crucial. More thorough research is necessary to explore an efficient approach to managing comorbidities for preclinical AD patients.
Collapse
Affiliation(s)
- Hanna Lu
- Department of Psychiatry, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xi Ni
- Department of Psychiatry, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
| | - Sandra Sau Man Chan
- Department of Psychiatry, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
| | - Calvin Pak Wing Cheng
- Department of Psychiatry, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Waichi Chan
- Department of Psychiatry, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Linda Chiu Wa Lam
- Department of Psychiatry, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
| |
Collapse
|
8
|
Xu J, Li C, Fan R, Yin J, Xie L, Peng X, Tao J, Xu W, Zhang S, Shi X, Dong K, Yu X, Chen X, Yang Y. BMAL1 ameliorates type 2 diabetes-induced cognitive impairment via AREG upregulation and PI3K/Akt/GSK-3β pathway activation. Cell Commun Signal 2025; 23:7. [PMID: 39762888 PMCID: PMC11705844 DOI: 10.1186/s12964-024-02019-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 12/27/2024] [Indexed: 01/11/2025] Open
Abstract
Cognitive impairment is a significant complication of type 2 diabetes mellitus (T2DM). However, the mechanisms underlying the development of cognitive dysfunction in individuals with T2DM remain elusive. Herein, we discussed the role of Bmal1, a core circadian rhythm-regulating gene, in the process of T2DM-associated cognitive dysfunction. We identified a marked decrease in BMAL1 levels in the hippocampus of db/db mice, followed by gain- and loss-of-function studies to explore the impact of BMAL1 on cognitive function. Our findings indicated that BMAL1 downregulation led to cognitive deficits, characterized by tau hyperphosphorylation and accumulated amyloid plaque. Conversely, BMAL1 overexpression mitigated these Alzheimer-like pathologies. Further investigation revealed that BMAL1 directly activated the transcription of Areg, thereby activating the PI3K/Akt/GSK-3β pathway and ameliorating cognitive dysfunction. Moreover, these effects of BMAL1 were attenuated by LY294002, a PI3K inhibitor. Collectively, these results underscore the significant role of BMAL1 in T2DM-associated cognitive impairment, proposing a novel intervention strategy for individuals exposed to risk factors of T2DM.
Collapse
Affiliation(s)
- Jialu Xu
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| | - Chunyu Li
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| | - Rongping Fan
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| | - Jiaxin Yin
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| | - Lei Xie
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
- Department of Endocrinology and Metabolism, GuiQian International General Hospital, Guiyang, China
| | - Xuemin Peng
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| | - Jing Tao
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| | - Weijie Xu
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| | - Shujun Zhang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| | - Xiaoli Shi
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| | - Kun Dong
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| | - Xuefeng Yu
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| | - Xi Chen
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China.
| | - Yan Yang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China.
| |
Collapse
|
9
|
Crespo MT, Trebucq LL, Senna CA, Hokama G, Paladino N, Agostino PV, Chiesa JJ. Circadian disruption of feeding-fasting rhythm and its consequences for metabolic, immune, cancer, and cognitive processes. Biomed J 2025:100827. [PMID: 39756653 DOI: 10.1016/j.bj.2025.100827] [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: 06/28/2024] [Revised: 12/30/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025] Open
Abstract
The circadian system is composed by a central hypothalamic clock at the suprachiasmatic nuclei (SCN) that communicates with peripheral circadian oscillators for daily coordination of behavior and physiology. The SCN entrain to the environmental 24-h light-dark (LD) cycle and drive daily rhythms of internal synchronizers such as core body temperature, hypothalamic-hypophysary hormones, sympathetic/parasympathetic activity, as well as behavioral and feeding-fasting rhythms, which supply signals setting core molecular clocks at central and peripheral tissues. Steady phase relationships between the SCN and peripheral oscillators keep homeostatic processes such as microbiota/microbiome composition/activity, metabolic supply/demand, energy balance, immunoinflammatory process, sleep amount and quality, psychophysiological stress, etc. Indeed, the risk of health alterations increase when these phase relationships are chronically changed prompting circadian disruption (CD), as occurring after sudden LD cycle changes (so-called jet-lag), or due to changes of activity/feeding-rest/fasting rhythm with respect to LD cycles (as humans subjected to nightwork, or restricting food access at rest in mice). Typical pathologies observed in animal models of CD and epidemiological studies include metabolic syndrome, type-2 diabetes, obesity, chronic inflammation, cancer, sleep disruption, decrease in physical and cognitive performance, and mood, among others. The present review discusses different aspects of such physiological dysregulations observed in animal models of CD having altered feeding-fasting rhythms, with potential translation to human health.
Collapse
Affiliation(s)
- Manuel Tomás Crespo
- ٰLaboratorio de Cronobiología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes/CONICET. Buenos Aires, Argentina
| | - Laura Lucía Trebucq
- ٰLaboratorio de Cronobiología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes/CONICET. Buenos Aires, Argentina
| | - Camila Agustina Senna
- ٰLaboratorio de Cronobiología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes/CONICET. Buenos Aires, Argentina
| | - Guido Hokama
- ٰLaboratorio de Cronobiología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes/CONICET. Buenos Aires, Argentina
| | - Natalia Paladino
- ٰLaboratorio de Cronobiología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes/CONICET. Buenos Aires, Argentina
| | - Patricia Verónica Agostino
- ٰLaboratorio de Cronobiología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes/CONICET. Buenos Aires, Argentina
| | - Juan José Chiesa
- ٰLaboratorio de Cronobiología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes/CONICET. Buenos Aires, Argentina.
| |
Collapse
|
10
|
Xu W, Li L, Cao Z, Ye J, Gu X. Circadian Rhythms and Lung Cancer in the Context of Aging: A Review of Current Evidence. Aging Dis 2025:AD.2024.1188. [PMID: 39812541 DOI: 10.14336/ad.2024.1188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 01/01/2025] [Indexed: 01/16/2025] Open
Abstract
Circadian rhythm is the internal homeostatic physiological clock that regulates the 24-hour sleep/wake cycle. This biological clock helps to adapt to environmental changes such as light, dark, temperature, and behaviors. Aging, on the other hand, is a process of physiological changes that results in a progressive decline in cells, tissues, and other vital systems of the body. Both aging and the circadian clock are highly interlinked phenomena with a bidirectional relationship. The process of aging leads to circadian disruptions while dysfunctional circadian rhythms promote age-related complications. Both processes involve diverse physiological, molecular, and cellular changes such as modifications in the DNA repair mechanisms, mechanisms, ROS generation, apoptosis, and cell proliferation. This review aims to examine the role of aging and circadian rhythms in the context of lung cancer. This will also review the existing literature on the role of circadian disruptions in the process of aging and vice versa. Various molecular pathways and genes such as BMAL1, SIRT1, HLF, and PER1 and their implications in aging, circadian rhythms, and lung cancer will also be discussed.
Collapse
Affiliation(s)
- Wenhui Xu
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, China
| | - Lei Li
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, China
| | - Zhendong Cao
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, China
| | - Jinghong Ye
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, China
| | - Xuyu Gu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
11
|
Zeng G, Liu X, Zheng Z, Zhao J, Zhuo W, Bai Z, Lin E, Cai S, Cai C, Li P, Zou B, Li J. Knockdown of RASD1 improves MASLD progression by inhibiting the PI3K/AKT/mTOR pathway. Lipids Health Dis 2024; 23:424. [PMID: 39731125 DOI: 10.1186/s12944-024-02419-z] [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: 10/29/2024] [Accepted: 12/22/2024] [Indexed: 12/29/2024] Open
Abstract
BACKGROUND There is still no reliable therapeutic targets and effective pharmacotherapy for metabolic dysfunction-associated steatotic liver disease (MASLD). RASD1 is short for Ras-related dexamethasone-induced 1, a pivotal factor in various metabolism processes of Human. However, the role of RASD1 remains poorly illustrated in MASLD. Therefore, we designed a study to elucidate how RASD1 could impact on MASLD as well as the mechanisms involved. METHODS The expression level of RASD1 was validated in MASLD. Lipid metabolism and its underlying mechanism were investigated in hepatocytes and mice with either overexpression or knockdown of RASD1. RESULTS Hepatic RASD1 expression was upregulated in MASLD. Lipid deposition was significantly reduced in RASD1-knockdown hepatocytes and mice, accompanied by a marked downregulation of key genes in the signaling pathway of de novo lipogenesis. Conversely, RASD1 overexpression in hepatocytes had the opposite effect. Mechanistically, RASD1 regulated lipid metabolism in MASLD through the PI3K/AKT/mTOR signaling pathway. CONCLUSIONS We discovered a novel role of RASD1 in MASLD by regulating lipogenesis via the PI3K/AKT/mTOR pathway, thereby identifying a potential treatment target for MASLD.
Collapse
Affiliation(s)
- Guifang Zeng
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China.
| | - Xialei Liu
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China
| | - Zhouying Zheng
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China
| | - Jiali Zhao
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China
| | - Wenfeng Zhuo
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China
| | - Zirui Bai
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China
| | - En Lin
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China
| | - Shanglin Cai
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China
| | - Chaonong Cai
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China
| | - Peiping Li
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China.
| | - Baojia Zou
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China.
| | - Jian Li
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China.
| |
Collapse
|
12
|
Woodie LN, Alberto AJ, Krusen BM, Melink LC, Lazar MA. Genetic synchronization of the brain and liver molecular clocks defend against chrono-metabolic disease. Proc Natl Acad Sci U S A 2024; 121:e2417678121. [PMID: 39665757 DOI: 10.1073/pnas.2417678121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/12/2024] [Indexed: 12/13/2024] Open
Abstract
Nearly every cell of the body contains a circadian clock mechanism that is synchronized with the light-entrained clock in the suprachiasmatic nucleus (SCN). Desynchrony between the SCN and the external environment leads to metabolic dysfunction in shift workers. Similarly, mice with markedly shortened endogenous period due to the deletion of circadian REV-ERBα/β nuclear receptors in the SCN (SCN DKO) exhibit increased sensitivity to diet-induced obesity (DIO) on a 24 h light:dark cycle while mice with REV-ERBs deleted in hepatocytes (HepDKO) display exacerbated hepatosteatosis in response to a high-fat diet. Here, we show that inducing deletion of hepatocyte REV-ERBs in SCN DKO mice (Hep-SCN DDKO) rescued the exacerbated DIO and hepatic triglyceride accumulation, without affecting the shortened behavioral period. These findings suggest that metabolic disturbances due to environmental desynchrony with the central clock are due to effects on peripheral clocks which can be mitigated by matching peripheral and central clock periods even in a desynchronous environment. Thus, maintaining synchrony within an organism, rather than between endogenous and exogenous clocks, may be a viable target for the treatment of metabolic disorders associated with circadian disruption.
Collapse
Affiliation(s)
- Lauren N Woodie
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
| | - Ahren J Alberto
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
| | - Brianna M Krusen
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
| | - Lily C Melink
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
| | - Mitchell A Lazar
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
13
|
DePoy LM, Vadnie CA, Petersen KA, Scott MR, Zong W, Yin R, Matthaei RC, Anaya FJ, Kampe CI, Tseng GC, McClung CA. Adolescent circadian rhythm disruption increases reward and risk-taking. Front Neurosci 2024; 18:1478508. [PMID: 39737435 PMCID: PMC11683121 DOI: 10.3389/fnins.2024.1478508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 11/11/2024] [Indexed: 01/01/2025] Open
Abstract
Introduction Circadian rhythm disturbances have long been associated with the development of psychiatric disorders, including mood and substance use disorders. Adolescence is a particularly vulnerable time for the onset of psychiatric disorders and for circadian rhythm and sleep disruptions. Preclinical studies have found that circadian rhythm disruption (CRD) impacts the brain and behavior, but this research is largely focused on adult disruptions. Here, we hypothesized that adolescent CRD would have a greater effect on psychiatric-related behaviors, relative to adult disruption. Methods We determined the long-term behavioral and neurobiological effects of CRD during early adolescence by exposing mice to 12 h shifts in the light/dark cycle. Adult mice were exposed to the same CRD paradigm. Behavior testing began approximately 4 weeks later for both groups. To identify possible mechanisms, we also measured gene expression in brain regions relevant to circadian rhythms, mood and reward. Results CRD during early adolescence, but not adulthood, persistently increased exploratory drive (risk-taking behavior) and cocaine preference when tested later in life. Interestingly, we found sex differences when intravenous cocaine self-administration was tested. While female mice with a history of adolescent CRD had a greater propensity to self-administer cocaine, as well as increased motivation and cue-induced reinstatement, male adolescent CRD mice had reduced motivation and extinction responding. Importantly, we found that transcripts in the SCN were affected by adolescent CRD and these were largely distinct across sex. Conclusion Overall, adolescent CRD in mice caused persistent increases in risky behavior, cocaine reward and cocaine self-administration, which suggests that CRD during adolescence may predispose individuals toward substance use disorders. Future research is required to elucidate how adolescent CRD affects behaviors relevant to mood-and substance use-related disorders across the 24-h day, as well as to identify intervention strategies to alleviate disruption during adolescence and novel therapeutic approaches once symptoms have begun.
Collapse
Affiliation(s)
- Lauren M. DePoy
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
| | - Chelsea A. Vadnie
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
- David O. Robbins Neuroscience Program, Department of Psychology, Ohio Wesleyan University, Delaware, OH, United States
| | - Kaitlyn A. Petersen
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
| | - Madeline R. Scott
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
| | - Wei Zong
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, United States
| | - RuoFei Yin
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ross C. Matthaei
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | | | - Callie I. Kampe
- David O. Robbins Neuroscience Program, Department of Psychology, Ohio Wesleyan University, Delaware, OH, United States
| | - George C. Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Colleen A. McClung
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
- David O. Robbins Neuroscience Program, Department of Psychology, Ohio Wesleyan University, Delaware, OH, United States
| |
Collapse
|
14
|
Ma T, Matsuo R, Kurogi K, Miyamoto S, Morita T, Shinozuka M, Taniguchi F, Ikegami K, Yasuo S. Sex-dependent effects of chronic jet lag on circadian rhythm and metabolism in mice. Biol Sex Differ 2024; 15:102. [PMID: 39639385 PMCID: PMC11619446 DOI: 10.1186/s13293-024-00679-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND The circadian clock integrates external environmental changes into the internal physiology of organisms. Perturbed circadian clocks due to misaligned light cycles increase the risk of diseases, including metabolic disorders. However, the effects of sex differences in this context remain unclear. METHODS Circadian misalignment was induced by a chronic jet lag (CJL) shift schedule (light-on time advanced by 6 h every 2 days) in C57BL/6N male and female mice. Core body temperature and activity rhythms were recorded using a nano tag, and the gene expression rhythms of clock and clock-controlled genes in the liver and adrenal glands were analyzed using qPCR. Glucose metabolism and insulin response were evaluated using glucose tolerance, insulin sensitivity, and glucose response assays. Castration and testosterone replacement were performed to assess the fundamental role of testosterone in male phenotypes under CJL. RESULTS Under CJL treatment, male mice exhibited increased weight gain, whereas females exhibited decreased weight gain compared to that of the respective controls. CJL treatment induced a lower robustness of circadian rhythms in core body temperature and a weaker rhythm of clock gene expression in the liver and adrenal glands in females, but not in males. Only male mice exhibited glucose intolerance under CJL conditions, without the development of insulin resistance. Castrated mice without testosterone exhibited decreased weight gain and reduced robustness of body temperature rhythm, as observed in intact females. Testosterone replacement in castrated mice recovered the CJL-induced weight gain, robustness of temperature rhythm, and glucose intolerance observed in intact males. CONCLUSIONS Significant sex-based differences were observed in circadian clock organization and metabolism under CJL. Testosterone plays a crucial role in maintaining the circadian clock and regulating CJL metabolism in males.
Collapse
Affiliation(s)
- Tiantian Ma
- Laboratory of Regulation in Metabolism and Behavior, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Ryohei Matsuo
- Laboratory of Regulation in Metabolism and Behavior, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Kaito Kurogi
- Laboratory of Regulation in Metabolism and Behavior, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Shunsuke Miyamoto
- Laboratory of Regulation in Metabolism and Behavior, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Tatsumi Morita
- Laboratory of Regulation in Metabolism and Behavior, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Marina Shinozuka
- Laboratory of Regulation in Metabolism and Behavior, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Fuka Taniguchi
- Laboratory of Regulation in Metabolism and Behavior, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Keisuke Ikegami
- Laboratory of Regulation in Metabolism and Behavior, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Shinobu Yasuo
- Laboratory of Regulation in Metabolism and Behavior, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan.
| |
Collapse
|
15
|
Kawata A, Kaneda Y, Matsunaga D, Nakagawa H, Togo F, Yasumatsu M, Ishiwata T. Influence of extreme light/dark cycles on monoamine levels, physiological indices, and emotional behaviors in rats. Chronobiol Int 2024; 41:1516-1532. [PMID: 39618305 DOI: 10.1080/07420528.2024.2434173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 11/18/2024] [Accepted: 11/20/2024] [Indexed: 12/12/2024]
Abstract
Aberrant light/dark (LD) cycles are prevalent in modern society due to electric light usage, leading to mood disorders from circadian disruption or misalignment. However, research on the physiological and behavioral effects of LD variations on brain neurotransmitters is limited. We investigated the effects of extreme LD cycles on body weight (BW), core body temperature (Tcore), locomotor activity (ACT), emotional behaviors, and monoamine levels (noradrenaline [NA], dopamine [DA], and serotonin [5-HT]) in male Wistar rats that were exposed to 1 month of either long light phase (20 L:4D), long dark phase (4 L:20D), or normal (12 L:12D) LD cycles. The 20 L:4D rats exhibited blunted rhythms, with decreased amplitude and advanced/delayed acrophase in Tcore and ACT, alongside increased BW. The 4 L:20D rats showed circadian misalignment, with increased/decreased amplitude in Tcore or ACT and delayed acrophase in Tcore and ACT, also gaining BW. In the 20 L:4D group, NA and 5-HT levels decreased in the suprachiasmatic nucleus and amygdala, respectively, while the 4 L:20D group had increased DA and 5-HT levels in the caudate putamen and dorsomedial hypothalamus, respectively. Open field and social interaction tests indicated anxiety-like behaviors in both test groups. Overall, each extreme LD cycle affected Tcore, ACT amplitude, acrophase, and monoamine levels differently, inducing anxiogenic responses.
Collapse
Affiliation(s)
- Akira Kawata
- Graduate School of Community and Human Services, Rikkyo University, Saitama, Japan
| | - Yuta Kaneda
- Graduate School of Community and Human Services, Rikkyo University, Saitama, Japan
| | - Daisuke Matsunaga
- Department of Health-Promotion and Sports Science, Osaka Electro-Communication University, Osaka, Japan
| | - Hikaru Nakagawa
- College of Sport and Wellness, Rikkyo University, Saitama, Japan
| | - Fumiharu Togo
- Department of Physical and Health Education, Graduate School of Education, The University of Tokyo, Tokyo, Japan
| | - Mikinobu Yasumatsu
- Graduate School of Community and Human Services, Rikkyo University, Saitama, Japan
- College of Sport and Wellness, Rikkyo University, Saitama, Japan
| | - Takayuki Ishiwata
- Graduate School of Community and Human Services, Rikkyo University, Saitama, Japan
- College of Sport and Wellness, Rikkyo University, Saitama, Japan
| |
Collapse
|
16
|
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
|
17
|
Deng Q, Li Y, Sun Z, Gao X, Zhou J, Ma G, Qu WM, Li R. Sleep disturbance in rodent models and its sex-specific implications. Neurosci Biobehav Rev 2024; 164:105810. [PMID: 39009293 DOI: 10.1016/j.neubiorev.2024.105810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/17/2024]
Abstract
Sleep disturbances, encompassing altered sleep physiology or disorders like insomnia and sleep apnea, profoundly impact physiological functions and elevate disease risk. Despite extensive research, the underlying mechanisms and sex-specific differences in sleep disorders remain elusive. While polysomnography serves as a cornerstone for human sleep studies, animal models provide invaluable insights into sleep mechanisms. However, the availability of animal models of sleep disorders is limited, with each model often representing a specific sleep issue or mechanism. Therefore, selecting appropriate animal models for sleep research is critical. Given the significant sex differences in sleep patterns and disorders, incorporating both male and female subjects in studies is essential for uncovering sex-specific mechanisms with clinical relevance. This review provides a comprehensive overview of various rodent models of sleep disturbance, including sleep deprivation, sleep fragmentation, and circadian rhythm dysfunction. We evaluate the advantages and disadvantages of each model and discuss sex differences in sleep and sleep disorders, along with potential mechanisms. We aim to advance our understanding of sleep disorders and facilitate sex-specific interventions.
Collapse
Affiliation(s)
- Qi Deng
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Yuhong Li
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Zuoli Sun
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Xiang Gao
- Shanxi Bethune Hospital, Shanxi, China
| | | | - Guangwei Ma
- Peking University Sixth Hospital, Beijing, China
| | - Wei-Min Qu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China; Department of Pharmacology, School of Basic Medical Sciences, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Rena Li
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
18
|
Burns JN, Jenkins AK, Yin R, Zong W, Vadnie CA, DePoy LM, Petersen KA, Tsyglakova M, Scott MR, Tseng GC, Huang YH, McClung CA. Molecular and cellular rhythms in excitatory and inhibitory neurons in the mouse prefrontal cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.05.601880. [PMID: 39005410 PMCID: PMC11245095 DOI: 10.1101/2024.07.05.601880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Previous studies have shown that there are rhythms in gene expression in the mouse prefrontal cortex (PFC); however, the contribution of different cell types and potential variation by sex has not yet been determined. Of particular interest are excitatory pyramidal cells and inhibitory parvalbumin (PV) interneurons, as interactions between these cell types are essential for regulating the excitation/inhibition balance and controlling many of the cognitive functions regulated by the PFC. In this study, we identify cell-type specific rhythms in the translatome of PV and pyramidal cells in the mouse PFC and assess diurnal rhythms in PV cell electrophysiological properties. We find that while core molecular clock genes are conserved and synchronized between cell types, pyramidal cells have nearly twice as many rhythmic transcripts as PV cells (35% vs. 18%). Rhythmic transcripts in pyramidal cells also show a high degree of overlap between sexes, both in terms of which transcripts are rhythmic and in the biological processes associated with them. Conversely, in PV cells, rhythmic transcripts from males and females are largely distinct. Moreover, we find sex-specific effects of phase on action potential properties in PV cells that are eliminated by environmental circadian disruption. Together, this study demonstrates that rhythms in gene expression and electrophysiological properties in the mouse PFC vary by both cell type and sex. Moreover, the biological processes associated with these rhythmic transcripts may provide insight into the unique functions of rhythms in these cells, as well as their selective vulnerabilities to circadian disruption.
Collapse
Affiliation(s)
- Jennifer N. Burns
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - Aaron K. Jenkins
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - RuoFei Yin
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15261
| | - Wei Zong
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15261
| | - Chelsea A. Vadnie
- David O. Robbins Neuroscience Program, Department of Psychology, Ohio Wesleyan University, Delaware, OH 43015
| | - Lauren M. DePoy
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - Kaitlyn A Petersen
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - Mariya Tsyglakova
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - Madeline R. Scott
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - George C. Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15261
| | - Yanhua H. Huang
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - Colleen A. McClung
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| |
Collapse
|
19
|
Gao X, Sun H, Wei Y, Niu J, Hao S, Sun H, Tang G, Qi C, Ge J. Protective effect of melatonin against metabolic disorders and neuropsychiatric injuries in type 2 diabetes mellitus mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 131:155805. [PMID: 38851097 DOI: 10.1016/j.phymed.2024.155805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/11/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is a metabolic disease characterized by hyperglycemia and progressive cognitive dysfunction, and our clinical investigation revealed that the plasma concentration of melatonin (Mlt) decreased and was closely related to cognition in T2DM patients. However, although many studies have suggested that Mlt has a certain protective effect on glucose and lipid metabolism disorders and neuropsychiatric injury, the underlying mechanism of Mlt against T2DM-related metabolic and cognitive impairments remains unclear. PURPOSE The aim of the present study was to investigate the therapeutic effect of Mlt on metabolic disorders and Alzheimer's disease (AD)-like neuropsychiatric injuries in T2DM mice and to explore the possible underlying molecular mechanism involved. METHODS A T2DM mouse model was established by a combination of a high-fat diet (HFD) and streptozotocin (STZ, 100 mg/kg, i.p.), and Mlt (5, 10 or 20 mg/kg) was intragastrically administered for six consecutive weeks. The serum levels of glycolipid metabolism indicators were measured, behavioral performance was tested, and the protein expression of key molecules involved in the regulation of synaptic plasticity, circadian rhythms, and neuroinflammation in the hippocampus was detected. Moreover, the fluorescence intensities of glial fibrillary acidic protein (GFAP), ionized calcium binding adapter molecule 1 (IBA-1), amyloid β-protein (Aβ) and phosphorylated Tau (p-Tau) in the hippocampus were also observed. RESULTS Treatment with Mlt not only improved T2DM-related metabolic disorders, as indicated by increased serum concentrations of fasting blood glucose (FBG), glycosylated hemoglobin (HbAlc), insulin (INS), total cholesterol (TC) and triglyceride (TG), improved glucose tolerance and liver and pancreas function but also alleviated AD-like neuropsychiatric injuries in a HFD/STZ-induced mouse model, as indicated by decreased immobility time in the tail suspension test (TST) and forced swimming test (FST), increased preference indices of novel objects or novel arms in the novel object recognition test (NOR) and Y-maze test (Y-maze), and improved platform positioning capability in the Morris water maze (MWM) test. Moreover, treatment with Mlt also improved the hyperactivation of astrocytes and microglia in the hippocampus of mice, accompanied by reduced expression of interleukin 1β (IL-1β), interleukin 6 (IL-6), tumor necrosis factor (TNF-α), Aβ, and p-Tau and increased expression of brain-derived neurotrophic factor (BDNF), Synapsin I, Synaptotagmin I, melatonin receptor 1B (MT1B), brain muscle arnt-like protein 1 (Bmal1), circadian locomotor output cycles kaput (Clock), period 2 (Per2), and cryptochrome 2 (Cry2). CONCLUSION Mlt alleviated T2DM-related metabolic disorders and AD-like neuropsychiatric injuries in a HFD/STZ-induced mouse model, possibly through a mechanism involving the regulation of glial activation and associated neuroinflammation and the balancing of synaptic plasticity and circadian rhythms in the hippocampus.
Collapse
Affiliation(s)
- Xinran Gao
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, PR China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, PR China; Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, PR China
| | - Huaizhi Sun
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, PR China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, PR China; Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, PR China
| | - Yadong Wei
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, PR China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, PR China; Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, PR China
| | - Jiachun Niu
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, PR China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, PR China; Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, PR China
| | - Shengwei Hao
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, PR China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, PR China; Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, PR China
| | - Huimin Sun
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, PR China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, PR China; Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, PR China
| | - Guozhang Tang
- School of 1st Clinic Medicine, Anhui Medical University, 81 Meishan Road, Hefei 230032, PR China
| | - Congcong Qi
- Department of Laboratory Animal Science, Fudan University, Shanghai, PR China.
| | - Jinfang Ge
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, PR China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, PR China; Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, PR China.
| |
Collapse
|
20
|
McHill AW, Butler MP. Eating Around the Clock: Circadian Rhythms of Eating and Metabolism. Annu Rev Nutr 2024; 44:25-50. [PMID: 38848598 PMCID: PMC11849495 DOI: 10.1146/annurev-nutr-062122-014528] [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] [Indexed: 06/09/2024]
Abstract
The time of day that we eat is increasingly recognized as contributing as importantly to overall health as the amount or quality of the food we eat. The endogenous circadian clock has evolved to promote intake at optimal times when an organism is intended to be awake and active, but electric lights and abundant food allow eating around the clock with deleterious health outcomes. In this review, we highlight literature pertaining to the effects of food timing on health, beginning with animal models and then translation into human experiments. We emphasize the pitfalls and opportunities that technological advances bring in bettering understanding of eating behaviors and their association with health and disease. There is great promise for restricting the timing of food intake both in clinical interventions and in public health campaigns for improving health via nonpharmacological therapies.
Collapse
Affiliation(s)
- Andrew W McHill
- Sleep, Chronobiology, and Health Laboratory, School of Nursing, Oregon Health & Science University, Portland, Oregon, USA
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, Oregon, USA
| | - Matthew P Butler
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA;
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
21
|
Rodríguez RM, Colom-Pellicer M, Hernández-Baixauli J, Calvo E, Suárez M, Arola-Arnal A, Torres-Fuentes C, Aragonès G, Mulero M. Grape Seed Proanthocyanidin Extract Attenuates Cafeteria-Diet-Induced Liver Metabolic Disturbances in Rats: Influence of Photoperiod. Int J Mol Sci 2024; 25:7713. [PMID: 39062955 PMCID: PMC11276873 DOI: 10.3390/ijms25147713] [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: 05/17/2024] [Revised: 07/04/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
This study investigated the influence of photoperiod (day length) on the efficacy of grape seed proanthocyanidin extract (GSPE) in mitigating metabolic disorders in obese rats fed a cafeteria diet. Rats were exposed to standard (L12), long (L18), or short (L6) photoperiods and treated with GSPE or vehicle. In the standard photoperiod, GSPE reduced body weight gain (50.5%), total cholesterol (37%), and triglycerides (34.8%), while increasing the expression of hepatic metabolic genes. In the long photoperiod, GSPE tended to decrease body weight gain, increased testosterone levels (68.3%), decreased liver weight (12.4%), and decreased reverse serum amino acids. In the short photoperiod, GSPE reduced glycemia (~10%) and lowered triglyceride levels (38.5%), with effects modified by diet. The standard photoperiod showed the greatest efficacy against metabolic syndrome-associated diseases. The study showed how day length affects GSPE's benefits and underscores considering biological rhythms in metabolic disease therapies.
Collapse
Affiliation(s)
- Romina M. Rodríguez
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili (URV), Campus de Sescelades, 43007 Tarragona, Spain; (R.M.R.); (M.C.-P.); (E.C.); (M.S.); (A.A.-A.); (C.T.-F.); (G.A.)
| | - Marina Colom-Pellicer
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili (URV), Campus de Sescelades, 43007 Tarragona, Spain; (R.M.R.); (M.C.-P.); (E.C.); (M.S.); (A.A.-A.); (C.T.-F.); (G.A.)
| | - Julia Hernández-Baixauli
- Laboratory of Metabolism and Obesity, Vall d’Hebron-Institut de Recerca, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain;
| | - Enrique Calvo
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili (URV), Campus de Sescelades, 43007 Tarragona, Spain; (R.M.R.); (M.C.-P.); (E.C.); (M.S.); (A.A.-A.); (C.T.-F.); (G.A.)
- Center of Environmental, Food and Toxicological Technology-TecnATox, Rovira i Virgili University, 43201 Reus, Spain
| | - Manuel Suárez
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili (URV), Campus de Sescelades, 43007 Tarragona, Spain; (R.M.R.); (M.C.-P.); (E.C.); (M.S.); (A.A.-A.); (C.T.-F.); (G.A.)
- Center of Environmental, Food and Toxicological Technology-TecnATox, Rovira i Virgili University, 43201 Reus, Spain
| | - Anna Arola-Arnal
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili (URV), Campus de Sescelades, 43007 Tarragona, Spain; (R.M.R.); (M.C.-P.); (E.C.); (M.S.); (A.A.-A.); (C.T.-F.); (G.A.)
- Center of Environmental, Food and Toxicological Technology-TecnATox, Rovira i Virgili University, 43201 Reus, Spain
| | - Cristina Torres-Fuentes
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili (URV), Campus de Sescelades, 43007 Tarragona, Spain; (R.M.R.); (M.C.-P.); (E.C.); (M.S.); (A.A.-A.); (C.T.-F.); (G.A.)
- Center of Environmental, Food and Toxicological Technology-TecnATox, Rovira i Virgili University, 43201 Reus, Spain
| | - Gerard Aragonès
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili (URV), Campus de Sescelades, 43007 Tarragona, Spain; (R.M.R.); (M.C.-P.); (E.C.); (M.S.); (A.A.-A.); (C.T.-F.); (G.A.)
- Center of Environmental, Food and Toxicological Technology-TecnATox, Rovira i Virgili University, 43201 Reus, Spain
| | - Miquel Mulero
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili (URV), Campus de Sescelades, 43007 Tarragona, Spain; (R.M.R.); (M.C.-P.); (E.C.); (M.S.); (A.A.-A.); (C.T.-F.); (G.A.)
- Center of Environmental, Food and Toxicological Technology-TecnATox, Rovira i Virgili University, 43201 Reus, Spain
| |
Collapse
|
22
|
Tonet NS, da Silva Marçal DF, da Silva FN, Brunetta HS, Mori MADS, Dos Santos GJ, Moreira ELG, Rafacho A. Moderate chronic sleep perturbation impairs glucose and lipid homeostasis in rats. Sleep 2024; 47:zsae118. [PMID: 38788154 DOI: 10.1093/sleep/zsae118] [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: 01/27/2024] [Revised: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
STUDY OBJECTIVES Sleep deprivation is a potential risk factor for metabolic diseases, including obesity and type 2 diabetes. We evaluated the impacts of moderate chronic sleep deprivation on glucose and lipid homeostasis in adult rats. METHODS Wistar rats (both sexes) were sleep-perturbed daily for 2 hours at the early (06:00-08:00) and the late light cycle (16:00-18:00) five days a week (except weekends) for 4 weeks. RESULTS Sleep perturbation (SP) resulted in reduced body weight gain in both sexes, associated with altered food intake and reduced adiposity. SP did not alter the short- or long-term memories or cause anxiogenic behavior. No major changes were observed in the plasma insulin, leptin, triacylglycerol, non-esterified fatty acids, and blood glucose upon SP. After SP, females exhibited a transitory glucose intolerance, while males became glucose intolerant at the end of the experimental period. Male rats also developed higher insulin sensitivity at the end of the SP protocol. Morphometric analyses revealed no changes in hepatic glycogen deposition, pancreatic islet mass, islet-cell distribution, or adrenal cortex thickness in SP rats from both sexes, except for lower adipocyte size compared with controls. We did not find homogeneous changes in the relative expression of circadian and metabolic genes in muscle or hepatic tissues from the SP rats. CONCLUSIONS Moderate chronic SP reduces visceral adiposity and causes glucose intolerance with a more pronounced impact on male rats, reinforcing the metabolic risks of exposure to sleep disturbances.
Collapse
Affiliation(s)
- Natália Stinghen Tonet
- Laboratory of Investigation in Chronic Diseases (LIDoC), Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
- Graduate Program in Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Danilo Francisco da Silva Marçal
- Laboratory of Investigation in Chronic Diseases (LIDoC), Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Flavia Natividade da Silva
- Laboratory of Investigation in Chronic Diseases (LIDoC), Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Henver Simionato Brunetta
- Department of Biochemistry and Tissue Biology, Institute of Biology, Universidade Estadual de Campinas, Campinas, Brazil
| | - Marcelo Alves da Silva Mori
- Department of Biochemistry and Tissue Biology, Institute of Biology, Universidade Estadual de Campinas, Campinas, Brazil
| | - Gustavo Jorge Dos Santos
- Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Eduardo Luiz Gasnhar Moreira
- Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Alex Rafacho
- Laboratory of Investigation in Chronic Diseases (LIDoC), Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
- Graduate Program in Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
- Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| |
Collapse
|
23
|
Duong HA, Baba K, DeBruyne JP, Davidson AJ, Ehlen C, Powell M, Tosini G. Environmental circadian disruption re-writes liver circadian proteomes. Nat Commun 2024; 15:5537. [PMID: 38956413 PMCID: PMC11220080 DOI: 10.1038/s41467-024-49852-3] [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: 02/20/2024] [Accepted: 06/21/2024] [Indexed: 07/04/2024] Open
Abstract
Circadian gene expression is fundamental to the establishment and functions of the circadian clock, a cell-autonomous and evolutionary-conserved timing system. Yet, how it is affected by environmental-circadian disruption (ECD) such as shiftwork and jetlag are ill-defined. Here, we provided a comprehensive and comparative description of male liver circadian gene expression, encompassing transcriptomes, whole-cell proteomes and nuclear proteomes, under normal and after ECD conditions. Under both conditions, post-translation, rather than transcription, is the dominant contributor to circadian functional outputs. After ECD, post-transcriptional and post-translational processes are the major contributors to whole-cell or nuclear circadian proteome, respectively. Furthermore, ECD re-writes the rhythmicity of 64% transcriptome, 98% whole-cell proteome and 95% nuclear proteome. The re-writing, which is associated with changes of circadian regulatory cis-elements, RNA-processing and protein localization, diminishes circadian regulation of fat and carbohydrate metabolism and persists after one week of ECD-recovery.
Collapse
Affiliation(s)
- Hao A Duong
- Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, GA, 30310, USA.
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, 30310, USA.
| | - Kenkichi Baba
- Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Jason P DeBruyne
- Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Alec J Davidson
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Christopher Ehlen
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Michael Powell
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Gianluca Tosini
- Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| |
Collapse
|
24
|
Kyung M, Park S, Park CG, Hong O. Association between Sleep Duration, Social Jetlag, and the Metabolic Syndrome by Shift Works. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2024; 21:668. [PMID: 38928916 PMCID: PMC11204024 DOI: 10.3390/ijerph21060668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/15/2024] [Accepted: 05/19/2024] [Indexed: 06/28/2024]
Abstract
Lifestyle factors, including sleep characteristics, have been implicated in the development of metabolic syndrome, particularly among shift workers. This study aimed to explore the relationship between shift work, sleep duration, social jetlag, and the risk of metabolic syndrome among U.S. workers and the moderating effect of sleep duration and social jetlag on this relationship. Data from the National Health and Nutrition Examination Survey (NHANES) in 2017-2020 March were analyzed. Poisson regression models were employed to examine associations. Among 4136 U.S. workers, 53.3% had metabolic syndrome, with a higher proportion of shift workers (63.8% vs. 56.7%, p = 0.001) and those sleeping less than 6 h or more than 9 h per week (22.3% vs. 19.1%, p = 0.044) in the affected group. Shift workers were initially found to have an increased risk of metabolic syndrome (Coef. = 0.03, 95% CI: 0.02, 0.16); however, this association was mitigated when accounting for the interaction with social jetlag. Specifically, 1 to <2 h of social jetlag interacted significantly, increasing metabolic risk (Coef. = 0.15, 95% CI: 0.09, 0.22), whereas 1 to <2 h alone showed a protective effect (Coef. = -0.11, 95% CI: -0.17, -0.06). These findings suggest that optimizing sleep schedules and addressing social jetlag may be crucial in mitigating metabolic syndrome risks among shift workers.
Collapse
Affiliation(s)
- Minjung Kyung
- School of Nursing, College of Nursing, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Sungwon Park
- School of Nursing, University of Michigan, 400 North Ingalls Street, Ann Arbor, MI 48109, USA;
| | - Chang Gi Park
- College of Nursing, University of Illinois at Chicago, 845 S. Damen Ave., MC 802, Chicago, IL 60612, USA;
| | - OiSaeng Hong
- School of Nursing, University of California, San Francisco, 2 Koret Way, San Francisco, CA 94143, USA;
| |
Collapse
|
25
|
Ma D, Qu Y, Wu T, Liu X, Cai L, Wang Y. Excessive fat expenditure in MCT-induced heart failure rats is associated with BMAL1/REV-ERBα circadian rhythmic loop disruption. Sci Rep 2024; 14:8128. [PMID: 38584196 PMCID: PMC10999456 DOI: 10.1038/s41598-024-58577-8] [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: 11/22/2023] [Accepted: 04/01/2024] [Indexed: 04/09/2024] Open
Abstract
Fat loss predicts adverse outcomes in advanced heart failure (HF). Disrupted circadian clocks are a primary cause of lipid metabolic issues, but it's unclear if this disruption affects fat expenditure in HF. To address this issue, we investigated the effects of disruption of the BMAL1/REV-ERBα circadian rhythmic loop on adipose tissue metabolism in HF.50 Wistar rats were initially divided into control (n = 10) and model (n = 40) groups. The model rats were induced with HF via monocrotaline (MCT) injections, while the control group received equivalent solvent injections. After establishing the HF model, the model group was further subdivided into four groups: normal rhythm (LD), inverted rhythm (DL), lentivirus vector carrying Bmal1 short hairpin RNA (LV-Bmal1 shRNA), and empty lentivirus vector control (LV-Control shRNA) groups, each with 10 rats. The DL subgroup was exposed to a reversed light-dark cycle of 8 h: 16 h (dark: light), while the rest adhered to normal light-dark conditions (light: dark 12 h: 12 h). Histological analyses were conducted using H&E, Oil Red O, and Picrosirius red stains to examine adipose and liver tissues. Immunohistochemical staining, RT-qPCR, and Western blotting were performed to detect markers of lipolysis, lipogenesis, and beiging of white adipose tissue (WAT), while thermogenesis indicators were detected in brown adipose tissue (BAT). The LD group rats exhibited decreased levels of BMAL1 protein, increased levels of REV-ERBα protein, and disrupted circadian circuits in adipose tissue compared to controls. Additionally, HF rats showed reduced adipose mass and increased ectopic lipid deposition, along with smaller adipocytes containing lower lipid content and fibrotic adipose tissue. In the LD group WAT, expression of ATGL, HSL, PKA, and p-PKA proteins increased, alongside elevated mRNA levels of lipase genes (Hsl, Atgl, Peripilin) and FFA β-oxidation genes (Cpt1, acyl-CoA). Conversely, lipogenic gene expression (Scd1, Fas, Mgat, Dgat2) decreased, while beige adipocyte markers (Cd137, Tbx-1, Ucp-1, Zic-1) and UCP-1 protein expression increased. In BAT, HF rats exhibited elevated levels of PKA, p-PKA, and UCP-1 proteins, along with increased expression of thermogenic genes (Ucp-1, Pparγ, Pgc-1α) and lipid transportation genes (Cd36, Fatp-1, Cpt-1). Plasma NT-proBNP levels were higher in LD rats, accompanied by elevated NE and IL-6 levels in adipose tissue. Remarkably, morphologically, the adipocytes in the DL and LV-Bmal1 shRNA groups showed reduced size and lower lipid content, while lipid deposition in the liver was more pronounced in these groups compared to the LD group. At the gene/protein level, the BMAL1/REV-ERBα circadian loop exhibited severe disruption in LV-Bmal1 shRNA rats compared to LD rats. Additionally, there was increased expression of lipase genes, FFA β oxidation genes, and beige adipocyte markers in WAT, as well as higher expression of thermogenic genes and lipid transportation genes in BAT. Furthermore, plasma NT-proBNP levels and adipose tissue levels of NE and IL-6 were elevated in LV-Bmal1 shRNA rats compared with LD rats. The present study demonstrates that disruption of the BMAL1/REV-ERBα circadian rhythmic loop is associated with fat expenditure in HF. This result suggests that restoring circadian rhythms in adipose tissue may help counteract disorders of adipose metabolism and reduce fat loss in HF.
Collapse
Affiliation(s)
- Dufang Ma
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, China
- Department of Cardiology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, No. 16369 Jingshi Road, Lixia District, Jinan, 250014, Shandong, China
| | - Yiwei Qu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, China
| | - Tao Wu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, China
| | - Xue Liu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, China
| | - Lu Cai
- Department of Cardiology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, No. 16369 Jingshi Road, Lixia District, Jinan, 250014, Shandong, China
| | - Yong Wang
- Department of Cardiology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, No. 16369 Jingshi Road, Lixia District, Jinan, 250014, Shandong, China.
| |
Collapse
|
26
|
Chawla S, Oster H, Duffield GE, Maronde E, Guido ME, Chabot C, Dkhissi-Benyahya O, Provencio I, Goel N, Youngstedt SD, Zi-Ching Mak N, Caba M, Nikhat A, Chakrabarti S, Wang L, Davis SJ. Reflections on Several Landmark Advances in Circadian Biology. J Circadian Rhythms 2024; 22:1. [PMID: 38617711 PMCID: PMC11011952 DOI: 10.5334/jcr.236] [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: 01/12/2024] [Accepted: 01/17/2024] [Indexed: 04/16/2024] Open
Abstract
Circadian Biology intersects with diverse scientific domains, intricately woven into the fabric of organismal physiology and behavior. The rhythmic orchestration of life by the circadian clock serves as a focal point for researchers across disciplines. This retrospective examination delves into several of the scientific milestones that have fundamentally shaped our contemporary understanding of circadian rhythms. From deciphering the complexities of clock genes at a cellular level to exploring the nuances of coupled oscillators in whole organism responses to stimuli. The field has undergone significant evolution lately guided by genetics approaches. Our exploration here considers key moments in the circadian-research landscape, elucidating the trajectory of this discipline with a keen eye on scientific advancements and paradigm shifts.
Collapse
Affiliation(s)
| | - Henrik Oster
- Institute of Neurobiology, Center for Brain, Behavior & Metabolism (CBBM), University of Luebeck, 23562 Luebeck, DE
| | - Giles E. Duffield
- Department of Biological Sciences and Eck Institute for Global Health, Galvin Life Science Center, University of Notre Dame, Notre Dame, IN 46556, US
| | - Erik Maronde
- Institut für Anatomie II, Dr. Senckenbergische Anatomie, Goethe-Universität Frankfurt, Theodor-Stern-Kai-7, 60590 Frankfurt, DE
| | - Mario E. Guido
- CIQUIBIC-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, AR
- Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, AR
| | - Christopher Chabot
- Department of Biological Sciences, Plymouth State University, Plymouth, NH 03264, US
| | - Ouria Dkhissi-Benyahya
- Inserm, Stem Cell and Brain Research Institute U1208, Univ Lyon, UniversitéClaude Bernard Lyon 1, 18 Avenue du Doyen Lépine, 69500, Bron, FR
| | - Ignacio Provencio
- Department of Biology and Department of Ophthalmology, University of Virginia, Charlottesville, VA, US
| | - Namni Goel
- Biological Rhythms Research Laboratory, Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, IL, US
| | - Shawn D. Youngstedt
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ, US
- Department of Medicine, University of Arizona, Tucson, AZ, US
| | | | - Mario Caba
- Centro de Investigaciones Biomédicas, Universidad Veracruzana, Xalapa, Ver., MX
| | - Anjoom Nikhat
- Simons Centre for the Study of Living Machines, National Centre for Biological Sciences, Bangalore, Karnataka 560065, IN
| | - Shaon Chakrabarti
- Simons Centre for the Study of Living Machines, National Centre for Biological Sciences, Bangalore, Karnataka 560065, IN
| | - Lei Wang
- Key Laboratory of Plant Molecular Physiology, CAS Center for Excellence in Molecular Plant Sciences, China National Botanical Garden, Beijing 100093, CN
| | - Seth J. Davis
- Department of Biology, University of York, York YO105DD, UK
- State Key Laboratory of Crop Stress Biology, School of Life Sciences, Henan University, Kaifeng 475004, CN
| |
Collapse
|
27
|
Dial MB, Malek EM, Cooper AR, Neblina GA, Vasileva NI, Hines DJ, McGinnis GR. Social jet lag impairs exercise volume and attenuates physiological and metabolic adaptations to voluntary exercise training. J Appl Physiol (1985) 2024; 136:996-1006. [PMID: 38450426 PMCID: PMC11305643 DOI: 10.1152/japplphysiol.00632.2023] [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: 09/06/2023] [Revised: 01/31/2024] [Accepted: 02/26/2024] [Indexed: 03/08/2024] Open
Abstract
Social jet lag (SJL) is a misalignment between sleep and wake times on workdays and free days. SJL leads to chronic circadian rhythm disruption and may affect nearly 70% of the general population, leading to increased risk for cardiometabolic diseases. This study investigated the effects of SJL on metabolic health, exercise performance, and exercise-induced skeletal muscle adaptations in mice. Ten-week-old C57BL/6J mice (n = 40) were allocated to four groups: control sedentary (CON-SED), control exercise (CON-EX), social jet lag sedentary (SJL-SED), and social jet lag exercise (SJL-EX). CON mice were housed under a 12:12-h light-dark cycle. SJL was simulated by implementing a 4-h phase delay for 3 days to simulate "weekends," followed by a 4-h phase advance back to "weekdays," for 6 wk. EX mice had free access to a running wheel. Graded exercise tests (GXTs) and glucose tolerance tests (GTTs) were performed at baseline and after intervention to monitor the effects of exercise and social jet lag on cardiorespiratory and metabolic health, respectively. SJL led to alterations in activity and running patterns and clock gene expression in skeletal muscle and decreased average running distance (P < 0.05). SJL-SED mice gained significantly more weight compared with CON-SED and SJL-EX mice (P < 0.01). SJL impaired fasting blood glucose and glucose tolerance compared with CON mice (P < 0.05), which was partially restored by exercise in SJL-EX mice. SJL also blunted improvements in exercise performance and mitochondrial content in the quadriceps. These data suggest that SJL blunted some cardiometabolic adaptations to exercise and that proper circadian hygiene is necessary for maintaining health and performance.NEW & NOTEWORTHY In mice, disrupting circadian rhythms with social jet lag for 6 wk caused significant weight gain, higher fasting blood glucose, and impaired glucose tolerance compared with control. Voluntary exercise in mice experiencing social jet lag prevented weight gain, though the mice still experienced increased fasting blood glucose and impaired exercise performance compared with trained mice not experiencing social jet lag. Social jet lag seems to be a potent circadian rhythm disruptor that impacts exercise-induced training adaptations.
Collapse
Affiliation(s)
- Michael B Dial
- Department of Kinesiology and Nutrition Sciences, School of Integrated Health Sciences, University of Nevada, Las Vegas, Nevada, United States
| | - Elias M Malek
- Department of Kinesiology and Nutrition Sciences, School of Integrated Health Sciences, University of Nevada, Las Vegas, Nevada, United States
| | - Austin R Cooper
- Department of Kinesiology and Nutrition Sciences, School of Integrated Health Sciences, University of Nevada, Las Vegas, Nevada, United States
| | - Greco A Neblina
- Department of Kinesiology and Nutrition Sciences, School of Integrated Health Sciences, University of Nevada, Las Vegas, Nevada, United States
| | - Nikoleta I Vasileva
- Department of Kinesiology and Nutrition Sciences, School of Integrated Health Sciences, University of Nevada, Las Vegas, Nevada, United States
| | - Dustin J Hines
- Department of Psychology, Psychological and Brain Sciences and Interdisciplinary Neuroscience Programs, University of Nevada, Las Vegas, Nevada, United States
| | - Graham R McGinnis
- Department of Kinesiology and Nutrition Sciences, School of Integrated Health Sciences, University of Nevada, Las Vegas, Nevada, United States
| |
Collapse
|
28
|
Licastro E, Pignataro G, Iliff JJ, Xiang Y, Lo EH, Hayakawa K, Esposito E. Glymphatic and lymphatic communication with systemic responses during physiological and pathological conditions in the central nervous system. Commun Biol 2024; 7:229. [PMID: 38402351 PMCID: PMC10894274 DOI: 10.1038/s42003-024-05911-5] [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: 07/06/2023] [Accepted: 02/12/2024] [Indexed: 02/26/2024] Open
Abstract
Crosstalk between central nervous system (CNS) and systemic responses is important in many pathological conditions, including stroke, neurodegeneration, schizophrenia, epilepsy, etc. Accumulating evidence suggest that signals for central-systemic crosstalk may utilize glymphatic and lymphatic pathways. The glymphatic system is functionally connected to the meningeal lymphatic system, and together these pathways may be involved in the distribution of soluble proteins and clearance of metabolites and waste products from the CNS. Lymphatic vessels in the dura and meninges transport cerebrospinal fluid, in part collected from the glymphatic system, to the cervical lymph nodes, where solutes coming from the brain (i.e., VEGFC, oligomeric α-syn, β-amyloid) might activate a systemic inflammatory response. There is also an element of time since the immune system is strongly regulated by circadian rhythms, and both glymphatic and lymphatic dynamics have been shown to change during the day and night. Understanding the mechanisms regulating the brain-cervical lymph node (CLN) signaling and how it might be affected by diurnal or circadian rhythms is fundamental to find specific targets and timing for therapeutic interventions.
Collapse
Affiliation(s)
- Ester Licastro
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University "Federico II", Naples, Italy
| | - Giuseppe Pignataro
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University "Federico II", Naples, Italy
| | - Jeffrey J Iliff
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Yanxiao Xiang
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Pharmacy, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Eng H Lo
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Consortium International pour la Recherche Circadienne sur l'AVC (CIRCA), Radcliffe Department of Medicine, University of Oxford, Headington, Oxford, UK
| | - Kazuhide Hayakawa
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.
| | - Elga Esposito
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.
- Consortium International pour la Recherche Circadienne sur l'AVC (CIRCA), Radcliffe Department of Medicine, University of Oxford, Headington, Oxford, UK.
| |
Collapse
|
29
|
Zhong Z, Tan X, An X, Li J, Cai J, Jiang Y, Taufique SKT, Li B, Shi Q, Zhao M, Wang Y, Luo Q, Wang H. Administration of blue light in the morning and no blue-ray light in the evening improves the circadian functions of non-24-hour shift workers. Chronobiol Int 2024; 41:267-282. [PMID: 38267234 DOI: 10.1080/07420528.2024.2305218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 01/07/2024] [Indexed: 01/26/2024]
Abstract
In modern 24-hour society, various round-the-clock services have entailed shift work, resulting in non-24-hour schedules. However, the extent of behavioral and physiological alterations by non-24-hour schedules remains unclear, and particularly, effective interventions to restore the circadian functions of non-24-hour shift workers are rarely explored. In this study, we investigate the effects of a simulated non-24-hour military shift work schedule on daily rhythms and sleep, and establish an intervention measure to restore the circadian functions of non-24-hour shift workers. The three stages of experiments were conducted. The stage-one experiment was to establish a comprehensive evaluation index of the circadian rhythms and sleep for all 60 participants by analyzing wristwatch-recorded physiological parameters and sleep. The stage-two experiment evaluated the effects of an intervention strategy on physiological rhythms and sleep. The stage-three experiment was to examine the participants' physiological and behavioral disturbances under the simulated non-24-hour military shift work schedule and their improvements by the optimal lighting apparatus. We found that wristwatch-recorded physiological parameters display robust rhythmicity, and the phases of systolic blood pressures and heart rates can be used as reliable estimators for the human body time. The simulated non-24-hour military shift work schedule significantly disrupts the daily rhythms of oxygen saturation levels, blood pressures, heart rates, and reduces sleep quality. Administration of blue light in the morning and no blue-ray light in the evening improves the amplitude and synchronization of daily rhythms of the non-24-hour participants. These findings demonstrate the harmful consequences of the non-24-hour shift work schedule and provide a non-invasive strategy to improve the well-being and work efficiency of the non-24-hour shift population.
Collapse
Affiliation(s)
- Zhaomin Zhong
- Center for Circadian Clocks, Soochow University, Suzhou, Jiangsu, China
- School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Xiaohui Tan
- Center for Circadian Clocks, Soochow University, Suzhou, Jiangsu, China
- School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Xingna An
- Center for Circadian Clocks, Soochow University, Suzhou, Jiangsu, China
- School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Jie Li
- Center for Circadian Clocks, Soochow University, Suzhou, Jiangsu, China
- School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Jing Cai
- Center for Circadian Clocks, Soochow University, Suzhou, Jiangsu, China
- School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Yunchun Jiang
- Center for Circadian Clocks, Soochow University, Suzhou, Jiangsu, China
- School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - S K Tahajjul Taufique
- Center for Circadian Clocks, Soochow University, Suzhou, Jiangsu, China
- School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Bo Li
- Center for Circadian Clocks, Soochow University, Suzhou, Jiangsu, China
- School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Quan Shi
- Center for Circadian Clocks, Soochow University, Suzhou, Jiangsu, China
- School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Meng Zhao
- Center for Circadian Clocks, Soochow University, Suzhou, Jiangsu, China
- School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Yali Wang
- Department of Neurology, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Qun Luo
- Naval Medical Center, PLA Naval Medical University, Shanghai, China
| | - Han Wang
- Center for Circadian Clocks, Soochow University, Suzhou, Jiangsu, China
- School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
30
|
Kaiser S, Henrich L, Kiessling I, Loy B, Schallner N. Neuroprotection via Carbon Monoxide Depends on the Circadian Regulation of CD36-Mediated Microglial Erythrophagocytosis in Hemorrhagic Stroke. Int J Mol Sci 2024; 25:1680. [PMID: 38338958 PMCID: PMC10855856 DOI: 10.3390/ijms25031680] [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: 12/20/2023] [Revised: 01/19/2024] [Accepted: 01/27/2024] [Indexed: 02/12/2024] Open
Abstract
The molecular basis for circadian dependency in stroke due to subarachnoid hemorrhagic stroke (SAH) remains unclear. We reasoned that microglial erythrophagocytosis, crucial for SAH response, follows a circadian pattern involving carbon monoxide (CO) and CD36 surface expression. The microglial BV-2 cell line and primary microglia (PMG) under a clocked medium change were exposed to blood ± CO (250 ppm, 1 h) in vitro. Circadian dependency and the involvement of CD36 were analyzed in PMG isolated from control mice and CD36-/- mice and by RNA interference targeting Per-2. In vivo investigations, including phagocytosis, vasospasm, microglia activation and spatial memory, were conducted in an SAH model using control and CD36-/- mice at different zeitgeber times (ZT). In vitro, the surface expression of CD36 and its dependency on CO and phagocytosis occurred with changed circadian gene expression. CD36-/- PMG exhibited altered circadian gene expression, phagocytosis and impaired responsiveness to CO. In vivo, control mice with SAH demonstrated circadian dependency in microglia activation, erythrophagocytosis and CO-mediated protection at ZT2, in contrast to CD36-/- mice. Our study indicates that circadian rhythmicity modulates microglial activation and subsequent CD36-dependent phagocytosis. CO altered circadian-dependent neuroprotection and CD36 induction, determining the functional outcome in a hemorrhagic stroke model. This study emphasizes how circadian rhythmicity influences neuronal damage after neurovascular events.
Collapse
Affiliation(s)
- Sandra Kaiser
- Department of Anesthesiology & Critical Care Medicine, Medical Center, University of Freiburg, 79106 Freiburg, Germany (N.S.)
- Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Luise Henrich
- Department of Anesthesiology & Critical Care Medicine, Medical Center, University of Freiburg, 79106 Freiburg, Germany (N.S.)
- Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Iva Kiessling
- Department of Anesthesiology & Critical Care Medicine, Medical Center, University of Freiburg, 79106 Freiburg, Germany (N.S.)
- Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Benedikt Loy
- Department of Anesthesiology & Critical Care Medicine, Medical Center, University of Freiburg, 79106 Freiburg, Germany (N.S.)
- Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Nils Schallner
- Department of Anesthesiology & Critical Care Medicine, Medical Center, University of Freiburg, 79106 Freiburg, Germany (N.S.)
- Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
| |
Collapse
|
31
|
Kanan MK, Sheehan PW, Haines JN, Gomez PG, Dhuler A, Nadarajah CJ, Wargel ZM, Freeberg BM, Nelvagal HR, Izumo M, Takahashi JS, Cooper JD, Davis AA, Musiek ES. Neuronal deletion of the circadian clock gene Bmal1 induces cell-autonomous dopaminergic neurodegeneration. JCI Insight 2024; 9:e162771. [PMID: 38032732 PMCID: PMC10906231 DOI: 10.1172/jci.insight.162771] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 11/28/2023] [Indexed: 12/02/2023] Open
Abstract
Circadian rhythm dysfunction is a hallmark of Parkinson disease (PD), and diminished expression of the core clock gene Bmal1 has been described in patients with PD. BMAL1 is required for core circadian clock function but also serves nonrhythmic functions. Germline Bmal1 deletion can cause brain oxidative stress and synapse loss in mice, and it can exacerbate dopaminergic neurodegeneration in response to the toxin MPTP. Here we examined the effect of cell type-specific Bmal1 deletion on dopaminergic neuron viability in vivo. We observed that global, postnatal deletion of Bmal1 caused spontaneous loss of tyrosine hydroxylase+ (TH+) dopaminergic neurons in the substantia nigra pars compacta (SNpc). This was not replicated by light-induced disruption of behavioral circadian rhythms and was not induced by astrocyte- or microglia-specific Bmal1 deletion. However, either pan-neuronal or TH neuron-specific Bmal1 deletion caused cell-autonomous loss of TH+ neurons in the SNpc. Bmal1 deletion did not change the percentage of TH neuron loss after α-synuclein fibril injection, though Bmal1-KO mice had fewer TH neurons at baseline. Transcriptomics analysis revealed dysregulation of pathways involved in oxidative phosphorylation and Parkinson disease. These findings demonstrate a cell-autonomous role for BMAL1 in regulating dopaminergic neuronal survival and may have important implications for neuroprotection in PD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Hemanth R. Nelvagal
- Departments of Pediatrics, Genetics and Genomic Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | - Joseph S. Takahashi
- Department of Neuroscience and
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jonathan D. Cooper
- Departments of Pediatrics, Genetics and Genomic Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | - Erik S. Musiek
- Department of Neurology and
- Center On Biological Rhythms and Sleep (COBRAS), Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
32
|
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
|
33
|
Lateef RS, Pokharel B, Shafin TN. Dozing Off With Drosophila: The Effect of Disrupted Circadian Rhythms and Sleep Disturbance on Mortality, Mood, and Addiction. Neurosci Insights 2023; 18:26331055231218698. [PMID: 38146331 PMCID: PMC10749519 DOI: 10.1177/26331055231218698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 11/20/2023] [Indexed: 12/27/2023] Open
Abstract
Many environmental factors can disrupt sleep and circadian rhythms, yet the consequences of such disruptions are poorly understood. The main goals of this project were to study the effects of disrupted circadian rhythms and sleep disturbance on Drosophila melanogaster's: (1) lifespan, (2) depression-like behaviors, and (3) propensity to consume caffeine-containing media. Three experimental groups were used: controls, Circadian Dysfunction (CD), and Sleep Disturbance (SD). Circadian disruption (CD): used flies with Tim01 mutation, which eliminates circadian behavioral rhythms. Sleep disturbance (SD): used flies subjected to hourly light exposure and manual mechanical disruption, for 48 hours. To assess the effect on lifespan, the percent of flies surviving over time, within each group, was calculated. Impaired geotaxis, or loss of climbing motivation, was assessed as a measure of a depression-like state. Preference for caffeine-containing food was evaluated using a choice chamber where caffeine enriched, and regular media were presented to flies. Group differences were analyzed with survival curves. Chi-square tests were used for the categorical variables. Survival curve analysis showed that Flies with the timeless gene mutation (tim01) have a significantly shorter lifespan than controls. Geotaxis was not significantly impaired by sleep disturbance, but it was negatively affected by circadian dysfunction. Both the Circadian Dysfunction and Sleep Disturbance groups showed a preference for caffeine-containing food, after 72 hours of exposure to it, although the Circadian Dysfunction group was much more affected than the Sleep Disturbance group. Sleep and circadian disturbances can negatively influence physical and mental wellbeing and the accompanying molecular mechanisms, as well as disrupted brain physiology, must be studied. It is critical to identify and minimize social and environmental disruptors of such biological rhythms.
Collapse
Affiliation(s)
- Rania S Lateef
- Governor’s School at Innovation Park and George Mason University, Manassas, VA, USA
| | | | | |
Collapse
|
34
|
Henrich L, Kiessling I, Steimer M, Frase S, Kaiser S, Schallner N. Circadian dependency of microglial heme oxygenase-1 expression and inflammation determine neuronal injury in hemorrhagic stroke. J Inflamm (Lond) 2023; 20:43. [PMID: 38104143 PMCID: PMC10725034 DOI: 10.1186/s12950-023-00371-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 12/13/2023] [Indexed: 12/19/2023] Open
Abstract
BACKGROUND The heme oxygenase-1 (HO-1) enzyme pathway is of crucial importance in the removal of toxic blood components and regulation of neuroinflammation following hemorrhagic stroke. Although a circadian pattern dependency in the incidence and severity of hemorrhagic stroke exists, it is unknown whether the activity of the HO-1 system in the context of hemorrhagic injury also exhibits circadian dependency. We hypothesized that the circadian regulation of microglial HO-1 would determine the extent of neuroinflammation and neuronal injury in a murine model of subarachnoid hemorrhage (SAH). METHODS In vitro expression patterns of HO-1 and circadian rhythm genes were analyzed in the microglial BV-2 cell line and primary microglia (PMG) using Western blot and qPCR. PMG isolated from Hmox1fl/fl and LyzM-Cre-Hmox1fl/fl mice were used to evaluate the role of microglial HO-1. We further investigated the in vivo relevance in a murine subarachnoid hemorrhage (SAH) model using Hmox1fl/fl and LyzM-Cre-Hmox1fl/fl mice with myeloid cell HO-1 deficiency, inducing SAH at different zeitgeber (ZT) times and analyzing the expression of HO-1 and the circadian control gene Period-2 (Per-2), respectively. Furthermore, we measured the inflammatory cytokine Monocyte Chemoattractant Protein-1 (MCP-1) in the cerebrospinal fluid of SAH patients in correlation with clinical outcome. RESULTS HO-1 baseline expression and response to CO with blood exposure depended on ZT. In vitro expression of circadian control genes was de-synchronized in LyzM-Cre-Hmox1fl/fl PMG and did not respond to exogenous CO exposure. We found that circadian rhythm plays a crucial role in brain damage after SAH. At ZT2, we observed less phagocytic function, more vasospasm and increased microglial activation. CO reduced mortality at ZT12 in HO-1 deficient mice and reduced the difference between ZT2 and ZT12 in the inflammatory response. Induction of MCP-1 in the CSF from SAH patients was time-dependent and correlated with the expression of circadian control genes, SAH severity, functional impairment and delirium. CONCLUSIONS Our data point towards a crucial role for the HO-1 enzyme system and circadian control in neuronal injury after a hemorrhagic stroke.
Collapse
Affiliation(s)
- Luise Henrich
- Department of Anesthesiology & Critical Care, Medical Center, University of Freiburg, Hugstetter Str. 55, Freiburg, 79106, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Iva Kiessling
- Department of Anesthesiology & Critical Care, Medical Center, University of Freiburg, Hugstetter Str. 55, Freiburg, 79106, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Matti Steimer
- Department of Anesthesiology & Critical Care, Medical Center, University of Freiburg, Hugstetter Str. 55, Freiburg, 79106, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sibylle Frase
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Neurology and Neuroscience, Medical Center, University of Freiburg, Freiburg, Germany
| | - Sandra Kaiser
- Department of Anesthesiology & Critical Care, Medical Center, University of Freiburg, Hugstetter Str. 55, Freiburg, 79106, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nils Schallner
- Department of Anesthesiology & Critical Care, Medical Center, University of Freiburg, Hugstetter Str. 55, Freiburg, 79106, Germany.
- Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
35
|
Radahmadi M, Salehifard K, Reisi P. In vivo synaptic potency, short-term and long-term plasticity at the hippocampal Schaffer collateral-CA1 synapses: Role of different light-dark cycles in male rats. Brain Res 2023; 1817:148514. [PMID: 37499734 DOI: 10.1016/j.brainres.2023.148514] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
The changes in the light-dark(L/D) cycle could modify cellular mechanisms in some brain regions. The present study compared the effects of various L/D cycles on invivo synaptic potency, short-term and long-term plasticity in the hippocampal CA1 area, adrenal glands weight(AGWs), corticosterone (CORT) levels, and body weight differences(BWD) in male rats. Male rats were assigned into different L/D cycle groups: L4/D20, L8/D16, L12/D12(control), L16/D8, and L20/D4. The slope, amplitude, and the area under curve(AUC) related to the field excitatory postsynaptic potentials(fEPSPs) were assessed, using the input-output(I/O) functions, paired-pulse(PP) responses at different interpulse intervals, and after the induction of long-term potentiation(LTP) in the hippocampal CA1 area. Also, the CORT levels, AGWs, and BWDs were measured in all groups. The slope, amplitude, and AUC of fEPSP in the I/O functions, all three phases of PP, before and after the LTP induction, were significantly decreased in all experimental groups, especially in the L20/D4 and L4/D20 groups. As such, the CORT levels and AGWs were significantly increased in all experimental groups, especially in the L20/D4 group. Overall, the uncommon L/D cycles (minimum and particularly maximum durations of light) significantly reduced the cellular mechanism of learning and memory. Also, downtrends were observed in synaptic potency, as well as short-term and long-term plasticity. The changes in PP with high interpulse intervals, or activity of GABAB receptors, were more significant than the changes in other PP phases with different L/D durations. Additionally, the CORT levels, adrenal glands, and body weight gain occurred time-independently concerning different L/D lengths.
Collapse
Affiliation(s)
- Maryam Radahmadi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Kowsar Salehifard
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parham Reisi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
36
|
Sellinger EP, Brinks AS, Javeri RR, Theurer SL, Wang R, Juraska JM. Region- and age-specific effects of perinatal phthalate exposure on developmental cell death and adult anatomy of dorsal and ventral hippocampus and associated cognitive behaviors. Neurotoxicol Teratol 2023; 99:107288. [PMID: 37595675 PMCID: PMC10530334 DOI: 10.1016/j.ntt.2023.107288] [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: 02/16/2023] [Revised: 08/01/2023] [Accepted: 08/10/2023] [Indexed: 08/20/2023]
Abstract
Humans are exposed to phthalates, a class of endocrine-disrupting chemicals used in food packaging/processing, PVC plastics, and personal care products. Gestational exposure may lead to adverse neurodevelopmental outcomes. In a rat model, perinatal exposure to an environmentally relevant mixture and dose of phthalates leads to increased developmental apoptosis in the medial prefrontal cortex (mPFC) and a subsequent reduction in neurons and in cognitive flexibility measured in adults of both sexes (Sellinger et al., 2021b; Kougias et al., 2018b). However, whether these effects generalize to other cognitive regions, like the hippocampus, is less well understood as existing studies used single phthalates at large doses, unrepresentative of human exposure. In the current study, patterns of naturally occurring cell death were first established in the dorsal and ventral hippocampal subfields (CA3 and CA1). Both dorsal and ventral CA3 reached high levels of cell death on P2 while levels in dorsal and ventral CA1 peaked on P5 in both sexes. Exposure to a phthalate mixture (0.2 and 1 mg/kg/day) throughout gestation through postnatal day 10 resulted in subtle age- and region-specific decreases in developmental cell death, however there were no significant changes in adult neuron number or associated behaviors: the Morris water maze and social recognition. Therefore, perinatal exposure to a low dose mixture of phthalates does not result in the dramatic structural and behavioral changes seen with high doses of single phthalates. This study also adds to our understanding of the distinct neurodevelopmental effects of phthalates on different brain regions.
Collapse
Affiliation(s)
- Elli P Sellinger
- Neuroscience Program, University of Illinois at Urbana-Champaign, 603 E. Daniel Street, Champaign, IL 61820, United States of America.
| | - Amara S Brinks
- Neuroscience Program, University of Illinois at Urbana-Champaign, 603 E. Daniel Street, Champaign, IL 61820, United States of America.
| | - Rajvi R Javeri
- Department of Psychology, University of Illinois at Urbana-Champaign, 603 E. Daniel Street, Champaign, IL 61820, United States of America.
| | - Savannah L Theurer
- Department of Psychology, University of Illinois at Urbana-Champaign, 603 E. Daniel Street, Champaign, IL 61820, United States of America.
| | - Ruibin Wang
- Department of Psychology, University of Illinois at Urbana-Champaign, 603 E. Daniel Street, Champaign, IL 61820, United States of America.
| | - Janice M Juraska
- Neuroscience Program, University of Illinois at Urbana-Champaign, 603 E. Daniel Street, Champaign, IL 61820, United States of America; Department of Psychology, University of Illinois at Urbana-Champaign, 603 E. Daniel Street, Champaign, IL 61820, United States of America.
| |
Collapse
|
37
|
Duong HA, Baba K, DeBruyne JP, Davidson AJ, Ehlen C, Powell M, Tosini G. Environmental circadian disruption re-programs liver circadian gene expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.28.555175. [PMID: 37693605 PMCID: PMC10491124 DOI: 10.1101/2023.08.28.555175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Circadian gene expression is fundamental to the establishment and functions of the circadian clock, a cell-autonomous and evolutionary-conserved timing system. Yet, how it is affected by environmental-circadian disruption (ECD) such as shiftwork and jetlag, which impact millions of people worldwide, are ill-defined. Here, we provided the first comprehensive description of liver circadian gene expression under normal and after ECD conditions. We found that post-transcription and post-translation processes are dominant contributors to whole-cell or nuclear circadian proteome, respectively. Furthermore, rhythmicity of 64% transcriptome, 98% whole-cell proteome and 95% nuclear proteome is re-written by ECD. The re-writing, which is associated with changes of circadian cis-regulatory elements, RNA-processing and protein trafficking, diminishes circadian regulation of fat and carbohydrate metabolism and persists after one week of ECD-recovery.
Collapse
Affiliation(s)
- Hao A. Duong
- Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta GA 30310
- Neuroscience Institute, Morehouse School of Medicine, Atlanta GA 30310
| | - Kenkichi Baba
- Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta GA 30310
- Neuroscience Institute, Morehouse School of Medicine, Atlanta GA 30310
| | - Jason P. DeBruyne
- Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta GA 30310
- Neuroscience Institute, Morehouse School of Medicine, Atlanta GA 30310
| | - Alec J. Davidson
- Neuroscience Institute, Morehouse School of Medicine, Atlanta GA 30310
| | - Christopher Ehlen
- Neuroscience Institute, Morehouse School of Medicine, Atlanta GA 30310
| | - Michael Powell
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta GA 30310
| | - Gianluca Tosini
- Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta GA 30310
- Neuroscience Institute, Morehouse School of Medicine, Atlanta GA 30310
| |
Collapse
|
38
|
Pehlivan S. The circadian systems genes and their importance of human health. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 137:1-15. [PMID: 37709372 DOI: 10.1016/bs.apcsb.2023.02.010] [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: 09/16/2023]
Abstract
The circadian rhythm is the timing mechanism that creates approximately 24-hour rhythms in cellular and bodily functions in almost all living species. These internal clock systems enable living organisms to predict and respond to daily changes in their environment, optimizing temporal physiology and behavior. Circadian rhythms are regulated by both genetic and environmental risk factors. Circadian rhythms play an important role in maintaining homeostasis at the systemic and tissue levels. Disruption of this rhythm lays the groundwork for human health and disease. Disruption in these rhythms increases the susceptibility to many diseases, such as cancer, psychiatric disorders, and neurodegenerative diseases. In this chapter, the characteristics of circadian rhythm and its relationship with diseases will be discussed.
Collapse
Affiliation(s)
- S Pehlivan
- Department of Medical Biology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey.
| |
Collapse
|
39
|
Hand AJ, Stone JE, Shen L, Vetter C, Cain SW, Bei B, Phillips AJK. Measuring light regularity: sleep regularity is associated with regularity of light exposure in adolescents. Sleep 2023; 46:zsad001. [PMID: 36625482 PMCID: PMC10424172 DOI: 10.1093/sleep/zsad001] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/30/2022] [Indexed: 01/11/2023] Open
Abstract
STUDY OBJECTIVES Light is the main time cue for the human circadian system. Sleep and light are intrinsically linked; light exposure patterns can influence sleep patterns and sleep can influence light exposure patterns. However, metrics for quantifying light regularity are lacking, and the relationship between sleep and light regularity is underexplored. We developed new metrics for light regularity and demonstrated their utility in adolescents, across school term and vacation. METHODS Daily sleep/wake and light patterns were measured using wrist actigraphy in 75 adolescents (54% male, 17.17 ± 0.83 years) over 2 weeks of school term and a subsequent 2-week vacation. The Sleep Regularity Index (SRI) and social jetlag were computed for each 2-week block. Light regularity was assessed using (1) variation in mean daily light timing (MLiT); (2) variation in daily photoperiod; and (3) the Light Regularity Index (LRI). Associations between SRI and each light regularity metric were examined, and within-individual changes in metrics were examined between school and vacation. RESULTS Higher SRI was significantly associated with more regular LRI scores during both school and vacation. There were no significant associations of SRI with variation in MLiT or daily photoperiod. Compared to school term, all three light regularity metrics were less variable during the vacation. CONCLUSIONS Light regularity is a multidimensional construct, which until now has not been formally defined. Irregular sleep patterns are associated with lower LRI, indicating that irregular sleepers also have irregular light inputs to the circadian system, which likely contributes to circadian disruption.
Collapse
Affiliation(s)
- Anthony J Hand
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, VIC, Australia
| | - Julia E Stone
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, VIC, Australia
| | - Lin Shen
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, VIC, Australia
| | - Céline Vetter
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Sean W Cain
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, VIC, Australia
| | - Bei Bei
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, VIC, Australia
| | - Andrew J K Phillips
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, VIC, Australia
| |
Collapse
|
40
|
Xiong X, Kiperman T, Li W, Dhawan S, Lee J, Yechoor V, Ma K. The Clock-modulatory Activity of Nobiletin Suppresses Adipogenesis Via Wnt Signaling. Endocrinology 2023; 164:bqad096. [PMID: 37327385 PMCID: PMC10373950 DOI: 10.1210/endocr/bqad096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 06/09/2023] [Accepted: 06/13/2023] [Indexed: 06/18/2023]
Abstract
The circadian clock machinery exerts transcriptional control to modulate adipogenesis and its disruption leads to the development of obesity. Here, we report that Nobiletin, a circadian clock amplitude-enhancing molecule, displays antiadipogenic properties via activation of Wnt signaling pathway that is dependent on its clock modulation. Nobiletin augmented clock oscillatory amplitude with period lengthening in the adipogenic mesenchymal precursor cells and preadipocytes, accompanied by an induction of Bmal1 and clock components within the negative feedback arm. Consistent with its clock-modulatory activity, Nobiletin strongly inhibited the lineage commitment and terminal differentiation of adipogenic progenitors. Mechanistically, we show that Nobiletin induced the reactivation of Wnt signaling during adipogenesis via transcriptional up-regulation of key components within this pathway. Furthermore, Nobiletin administration in mice markedly reduced adipocyte hypertrophy, leading to a significant loss of fat mass and reduction of body weight. Last, Nobiletin inhibited the differentiation of primary preadipocytes, and this effect was dependent on a functional clock regulation. Collectively, our findings uncover a novel activity of Nobiletin in suppressing adipocyte development in a clock-dependent manner, implicating its potential application in countering obesity and associated metabolic consequences.
Collapse
Affiliation(s)
- Xuekai Xiong
- Department of Diabetes Complications and Metabolism, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Tali Kiperman
- Department of Diabetes Complications and Metabolism, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Weini Li
- Department of Diabetes Complications and Metabolism, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Sangeeta Dhawan
- Department of Translational Research and Cellular Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Jeongkyung Lee
- Diabetes and Beta Cell Biology Center, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Vijay Yechoor
- Diabetes and Beta Cell Biology Center, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Ke Ma
- Department of Diabetes Complications and Metabolism, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
41
|
Roberts BL, Karatsoreos IN. Circadian desynchronization disrupts physiological rhythms of prefrontal cortex pyramidal neurons in mice. Sci Rep 2023; 13:9181. [PMID: 37280307 PMCID: PMC10244337 DOI: 10.1038/s41598-023-35898-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 05/25/2023] [Indexed: 06/08/2023] Open
Abstract
Disruption of circadian rhythms, such as shift work and jet lag, are associated with negative physiological and behavioral outcomes, including changes in affective state, learning and memory, and cognitive function. The prefrontal cortex (PFC) is heavily involved in all of these processes. Many PFC-associated behaviors are time-of-day dependent, and disruption of daily rhythms negatively impacts these behavioral outputs. Yet how disruption of daily rhythms impacts the fundamental function of PFC neurons, and the mechanism(s) by which this occurs, remains unknown. Using a mouse model, we demonstrate that the activity and action potential dynamics of prelimbic PFC neurons are regulated by time-of-day in a sex specific manner. Further, we show that postsynaptic K+ channels play a central role in physiological rhythms, suggesting an intrinsic gating mechanism mediating physiological activity. Finally, we demonstrate that environmental circadian desynchronization alters the intrinsic functioning of these neurons independent of time-of-day. These key discoveries demonstrate that daily rhythms contribute to the mechanisms underlying the essential physiology of PFC circuits and provide potential mechanisms by which circadian disruption may impact the fundamental properties of neurons.
Collapse
Affiliation(s)
- Brandon L Roberts
- Neuroscience and Behavior Program, and Department of Psychological and Brain Sciences, University of Massachusetts Amherst, Tobin Hall, 135 Hicks Way, Amherst, MA, 01003S, USA
| | - Ilia N Karatsoreos
- Neuroscience and Behavior Program, and Department of Psychological and Brain Sciences, University of Massachusetts Amherst, Tobin Hall, 135 Hicks Way, Amherst, MA, 01003S, USA.
| |
Collapse
|
42
|
Peng X, Chen Y. The emerging role of circadian rhythms in the development and function of thermogenic fat. Front Endocrinol (Lausanne) 2023; 14:1175845. [PMID: 37293491 PMCID: PMC10244810 DOI: 10.3389/fendo.2023.1175845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/06/2023] [Indexed: 06/10/2023] Open
Abstract
Circadian rhythms regulate many biological processes in response to ambient influences. A disrupted circadian rhythm has been shown to be associated with obesity and obesity-related metabolic disorders. Thermogenic fat, including brown and beige fat, may play an important role in this process since it displays a high capacity to burn fat and release the stored energy as heat, contributing to the combat against obesity and its associated metabolic disorders. In this review, we summarize the relationship between the circadian clock and thermogenic fat and the prominent mechanisms which are involved in the regulation of the development and function of thermogenic fat by circadian rhythms, which may provide novel therapeutics for the prevention and treatment of metabolic diseases by targeting thermogenic fat in a circadian manner.
Collapse
Affiliation(s)
- Xuemin Peng
- Division of Endocrinology, Internal Medicine, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
- Laboratory of Endocrinology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Chen
- Division of Endocrinology, Internal Medicine, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
- Laboratory of Endocrinology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| |
Collapse
|
43
|
Chbeir S, Carrión V. Resilience by design: How nature, nurture, environment, and microbiome mitigate stress and allostatic load. World J Psychiatry 2023; 13:144-159. [PMID: 37303926 PMCID: PMC10251360 DOI: 10.5498/wjp.v13.i5.144] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/11/2023] [Accepted: 04/17/2023] [Indexed: 05/19/2023] Open
Abstract
Resilience to psychological stress is defined as adaption to challenging life experiences and not the absence of adverse life events. Determinants of resilience include personality traits, genetic/epigenetic modifications of genes involved in the stress response, cognitive and behavioral flexibility, secure attachment with a caregiver, social and community support systems, nutrition and exercise, and alignment of circadian rhythm to the natural light/dark cycle. Therefore, resilience is a dynamic and flexible process that continually evolves by the intersection of different domains in human’s life; biological, social, and psychological. The objective of this minireview is to summarize the existing knowledge about the multitude factors and molecular alterations that result from resilience to stress response. Given the multiple contributing factors in building resilience, we set out a goal to identify which factors were most supportive of a causal role by the current literature. We focused on resilience-related molecular alterations resulting from mind-body homeostasis in connection with psychosocial and environmental factors. We conclude that there is no one causal factor that differentiates a resilient person from a vulnerable one. Instead, building resilience requires an intricate network of positive experiences and a healthy lifestyle that contribute to a balanced mind-body connection. Therefore, a holistic approach must be adopted in future research on stress response to address the multiple elements that promote resilience and prevent illnesses and psychopathology related to stress allostatic load.
Collapse
Affiliation(s)
- Souhad Chbeir
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Stanford University, Stanford, CA 94305, United States
| | - Victor Carrión
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Stanford University, Stanford, CA 94305, United States
| |
Collapse
|
44
|
Delorme TC, Ozell-Landry W, Cermakian N, Srivastava LK. Behavioral and cellular responses to circadian disruption and prenatal immune activation in mice. Sci Rep 2023; 13:7791. [PMID: 37179433 PMCID: PMC10182998 DOI: 10.1038/s41598-023-34363-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023] Open
Abstract
Most individuals with neurodevelopmental disorders (NDDs), including schizophrenia and autism spectrum disorders, experience disruptions in sleep and circadian rhythms. Epidemiological studies indicate that exposure to prenatal infection increases the risk of developing NDDs. We studied how environmental circadian disruption contributes to NDDs using maternal immune activation (MIA) in mice, which models prenatal infection. Pregnant dams were injected with viral mimetic poly IC (or saline) at E9.5. Adult poly IC- and saline-exposed offspring were subjected to 4 weeks of each of the following: standard lighting (LD1), constant light (LL) and standard lighting again (LD2). Behavioral tests were conducted in the last 12 days of each condition. Poly IC exposure led to significant behavioral differences, including reduced sociability (males only) and deficits in prepulse inhibition. Interestingly, poly IC exposure led to reduced sociability specifically when males were tested after LL exposure. Mice were exposed again to either LD or LL for 4 weeks and microglia were characterized. Notably, poly IC exposure led to increased microglial morphology index and density in dentate gyrus, an effect attenuated by LL exposure. Our findings highlight interactions between circadian disruption and prenatal infection, which has implications in informing the development of circadian-based therapies for individuals with NDDs.
Collapse
Affiliation(s)
- Tara C Delorme
- Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montréal, QC, H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montréal, QC, H3A 2B4, Canada
| | - William Ozell-Landry
- Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montréal, QC, H4H 1R3, Canada
| | - Nicolas Cermakian
- Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montréal, QC, H4H 1R3, Canada.
- Department of Psychiatry, McGill University, Montréal, QC, H3A 1A1, Canada.
| | - Lalit K Srivastava
- Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montréal, QC, H4H 1R3, Canada.
- Department of Psychiatry, McGill University, Montréal, QC, H3A 1A1, Canada.
| |
Collapse
|
45
|
Wright CJ, Milosavljevic S, Pocivavsek A. The stress of losing sleep: Sex-specific neurobiological outcomes. Neurobiol Stress 2023; 24:100543. [PMID: 37252645 PMCID: PMC10209346 DOI: 10.1016/j.ynstr.2023.100543] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/20/2023] [Accepted: 05/06/2023] [Indexed: 05/31/2023] Open
Abstract
Sleep is a vital and evolutionarily conserved process, critical to daily functioning and homeostatic balance. Losing sleep is inherently stressful and leads to numerous detrimental physiological outcomes. Despite sleep disturbances affecting everyone, women and female rodents are often excluded or underrepresented in clinical and pre-clinical studies. Advancing our understanding of the role of biological sex in the responses to sleep loss stands to greatly improve our ability to understand and treat health consequences of insufficient sleep. As such, this review discusses sex differences in response to sleep deprivation, with a focus on the sympathetic nervous system stress response and activation of the hypothalamic-pituitary-adrenal (HPA) axis. We review sex differences in several stress-related consequences of sleep loss, including inflammation, learning and memory deficits, and mood related changes. Focusing on women's health, we discuss the effects of sleep deprivation during the peripartum period. In closing, we present neurobiological mechanisms, including the contribution of sex hormones, orexins, circadian timing systems, and astrocytic neuromodulation, that may underlie potential sex differences in sleep deprivation responses.
Collapse
Affiliation(s)
- Courtney J. Wright
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Snezana Milosavljevic
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Ana Pocivavsek
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| |
Collapse
|
46
|
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: 9] [Impact Index Per Article: 4.5] [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
|
47
|
Gao X, Wei Y, Sun H, Hao S, Ma M, Sun H, Zang D, Qi C, Ge J. Role of Bmal1 in Type 2 Diabetes Mellitus-Related Glycolipid Metabolic Disorder and Neuropsychiatric Injury: Involved in the Regulation of Synaptic Plasticity and Circadian Rhythms. Mol Neurobiol 2023:10.1007/s12035-023-03360-5. [PMID: 37126129 DOI: 10.1007/s12035-023-03360-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/19/2023] [Indexed: 05/02/2023]
Abstract
Increasing data suggest a crucial role of circadian rhythm in regulating metabolic and neurological diseases, and Bmal1 is regarded as a key regulator of circadian transcription. The aim of this study is to investigate the role of Bmal1 in the disruption of circadian rhythm and neuropsychiatric injuries in type 2 diabetes mellitus (T2DM). A T2DM model was induced by the combination of high-fat-diet (HFD) and streptozotocin (STZ) in vivo or HT-22 cells challenged with palmitic-acid (PA) in vitro. The glucolipid metabolism indicators, behavioral performance, and expression of synaptic plasticity proteins and circadian rhythm-related proteins were detected. These changes were also observed after interference of Bmal1 expression via overexpressed plasmid or small interfering RNAs in vitro. The results showed that HFD/STZ could induce T2DM-like glycolipid metabolic turmoil and abnormal neuropsychiatric behaviors in mice, as indicated by the increased concentrations of fasting blood-glucose (FBG), HbA1c and lipids, the impaired glucose tolerance, and the decreased preference index of novel object or novel arm in the novel object recognition test (NOR) and Y-maze test (Y-maze). Consistently, the protein expression of synaptic plasticity proteins and circadian rhythm-related proteins and the positive fluorescence intensity of MT1B and Bmal1 were decreased in the hippocampus of HFD/STZ-induced mice or PA-challenged HT-22 cells. Furthermore, overexpression of Bmal1 could improve the PA-induced lipid metabolic dysfunction and increase the decreased expressions of synaptic plasticity proteins and circadian rhythm-related proteins, and vice versa. These results suggested a crucial role of Bmal1 in T2DM-related glycolipid metabolic disorder and neuropsychiatric injury, which mechanism might be involved in the regulation of synaptic plasticity and circadian rhythms.
Collapse
Affiliation(s)
- Xinran Gao
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
- The Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, China
| | - Yadong Wei
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
- The Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, China
| | - Huaizhi Sun
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
- The Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, China
| | - Shengwei Hao
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
- The Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, China
| | - Mengdie Ma
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
- The Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, China
| | - Huimin Sun
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
- The Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, China
| | - Dandan Zang
- The Center for Scientific Research of Anhui Medical University, Hefei, China
| | - Congcong Qi
- Department of Laboratory Animal Science, Fudan University, Shanghai, China
| | - Jinfang Ge
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China.
- The Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China.
- Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, China.
| |
Collapse
|
48
|
Wang W, Hao Z, Wu Z, Cui J, Liu H. Long-term artificial/natural daytime light affects mood, melatonin, corticosterone, and gut microbiota in rats. Appl Microbiol Biotechnol 2023; 107:2689-2705. [PMID: 36912904 DOI: 10.1007/s00253-023-12446-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/02/2023] [Accepted: 02/13/2023] [Indexed: 03/14/2023]
Abstract
The desynchronization of circadian rhythms affected by light may induce physiological and psychological disequilibrium. We aimed to elucidate changes of growth, depression-anxiety like behaviors, melatonin and corticosterone (CORT) secretion, and gut microbiota in rats influenced by long-term light inputs. Thirty male Sprague-Dawley rats were exposed to a 16/8 h light/dark regime for 8 weeks. The light period was set to 13 h of daylight with artificial light (AL group, n = 10), or with natural light (NL group, n = 10), or with mixed artificial-natural light (ANL group, n = 10), and 3 h of artificial night light after sunset. The obtained findings indicated that the highest weight gain and food efficiency were observed in the AL group and the lowest in NL group. In the behavioral tests, the NL and ANL groups showed lower anxiety level than AL group, and ANL groups showed lower depression level than AL group. The NL and ANL groups had delayed acrophases and maintained higher concentrations of melatonin compared to AL group. The circadian rhythm of CORT was only found in ANL group. At the phylum level, the mixed light contributed to a lower abundance of Bacteroidetes. The genus level results recommend a synergistic effect of artificial light and natural light on Lactobacillus abundance and an antagonistic effect on the Lachnospiraceae_NK4A136_group abundance. The study indicated that the mixture of artificial and natural light as well as the alignment of the proportions had beneficial influences on depression-anxiety-like levels, melatonin and corticosterone secretion, and the composition of the gut microbiota. KEY POINTS: • The mixed light can reduce the depression-anxiety level • The mixed light can maintain the secretion rhythm of melatonin and CORT • The mixed light can increase Lactobacillus and decrease Lachnospiraceae_NK4A136_group.
Collapse
Affiliation(s)
- Wei Wang
- Institute of Environmental Biology and Life Support Technology, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
- Institute of Medical Psychology, Faculty of Medicine, Ludwig-Maximilian-University of Munich, 80336, Munich, Germany
| | - Zikai Hao
- Key Laboratory of Molecular Medicine and Biotherapy, Ministry of Industry and Information Technology, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
| | - Zizhou Wu
- Institute of Environmental Biology and Life Support Technology, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Jingwei Cui
- Institute of Environmental Biology and Life Support Technology, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Hong Liu
- Institute of Environmental Biology and Life Support Technology, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China.
- International Joint Research Center of Aerospace Biotechnology & Medical Engineering, Beihang University, Beijing, 100083, China.
| |
Collapse
|
49
|
de Zavalia N, Ferraro S, Amir S. Sexually dimorphic role of circadian clock genes in alcohol drinking behavior. Psychopharmacology (Berl) 2023; 240:431-440. [PMID: 36184679 DOI: 10.1007/s00213-022-06247-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 09/17/2022] [Indexed: 11/25/2022]
Abstract
Sex differences in alcohol use and abuse are pervasive and carry important implications for the prevention and treatment of alcohol use disorder (AUD), yet insight into underlying sexually dimorphic mechanisms is limited. Growing experimental and clinical evidence points to an important influence of circadian rhythms and circadian clock genes in the control of alcohol drinking behavior and AUD. Sex differences in the expression of circadian rhythms and in the molecular circadian clock that drive these rhythms have been reported in humans and animals. While studying the role of striatal circadian clock gene expression in the control of affective and goal-directed behaviors, we uncovered a novel sexually dimorphic function of the clock genes Bmal1 and Per2 in the control of voluntary alcohol consumption in mice, which may contribute to sex differences in alcohol drinking behavior. In this mini review, we briefly discuss relevant literature on AUD, circadian rhythms and clock genes, and on sex differences in these domains, and describe our own findings on clock genes as sexually dimorphic regulators of alcohol drinking behavior in mice.
Collapse
Affiliation(s)
- Nuria de Zavalia
- Center for Studies in Behavioural Neurobiology, Department of Psychology, Concordia University, Montreal, QC, Canada
| | - Sarah Ferraro
- Center for Studies in Behavioural Neurobiology, Department of Psychology, Concordia University, Montreal, QC, Canada
| | - Shimon Amir
- Center for Studies in Behavioural Neurobiology, Department of Psychology, Concordia University, Montreal, QC, Canada.
| |
Collapse
|
50
|
Gonzalez D, Justin H, Reiss S, Faulkner J, Mahoney H, Yunus A, Gamsby J, Gulick D. Circadian rhythm shifts and alcohol access in adolescence synergistically increase alcohol preference and intake in adulthood in male C57BL/6 mice. Behav Brain Res 2023; 438:114216. [PMID: 36400236 DOI: 10.1016/j.bbr.2022.114216] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Adolescents have a natural tendency to be night owls, maintaining delayed circadian rhythms, and this rhythm is in direct conflict with the early wake times required during the school year. This leads to 'social jetlag', chronic circadian stress or desynchrony (CD) in which the rhythm of the intrinsic body clock is out of sync with behavior. CD increases alcohol intake in adolescents and adults, yet it is unknown whether adolescent CD also increases long-term addiction risk. The goal of this study was to determine whether adolescent alcohol intake in CD would increase adult alcohol preference and intake in male C57BL/6 J mice. METHODS We measured free access alcohol intake, water intake, and wheel-running activity during a normal 12 h (h) baseline photoperiod and then during shifting lighting schedules (Experiment 1) or a shortened circadian day (Experiment 2). RESULTS In Experiment 1, altered lighting produced a persistent increase in adolescent alcohol intake and in binge-like drinking (drinking at least 5 licks per minute, with no more than a 1 min break in drinking) in adulthood, but only a transient increase in total alcohol intake for the first week after alcohol was reintroduced in adulthood. In Experiment 2, the circadian shift produced a significant increase in alcohol intake in both adolescence and adulthood. Molecular analysis demonstrated changes in plasma corticosterone and neuronal markers of stress and addiction at the conclusion of these experiments in the CD and alcohol-exposed groups. CONCLUSIONS Thus, we conclude that circadian stress during adolescence is sufficient to produce a long-lasting susceptibility to alcohol use.
Collapse
Affiliation(s)
- David Gonzalez
- USF Neuroscience Institute, Byrd Institute, University of South Florida Health, Tampa, FL, USA; Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Hannah Justin
- USF Neuroscience Institute, Byrd Institute, University of South Florida Health, Tampa, FL, USA; Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Samantha Reiss
- USF Neuroscience Institute, Byrd Institute, University of South Florida Health, Tampa, FL, USA; Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - John Faulkner
- USF Neuroscience Institute, Byrd Institute, University of South Florida Health, Tampa, FL, USA
| | - Heather Mahoney
- USF Neuroscience Institute, Byrd Institute, University of South Florida Health, Tampa, FL, USA; Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Amara Yunus
- USF Neuroscience Institute, Byrd Institute, University of South Florida Health, Tampa, FL, USA
| | - Joshua Gamsby
- USF Neuroscience Institute, Byrd Institute, University of South Florida Health, Tampa, FL, USA; Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Danielle Gulick
- USF Neuroscience Institute, Byrd Institute, University of South Florida Health, Tampa, FL, USA; Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
| |
Collapse
|