101
|
Hasenmajer V, Sbardella E, Sciarra F, Simeoli C, Pivonello C, Ceccato F, Pofi R, Minnetti M, Rizzo F, Ferrari D, Bonaventura I, Barbagallo F, Giannetta E, Alunni Fegatelli D, Conia S, Navigli R, Arnaldi G, Scaroni C, Pivonello R, Gianfrilli D, Venneri MA, Isidori AM. Circadian clock disruption impairs immune oscillation in chronic endogenous hypercortisolism: a multi-level analysis from a multicentre clinical trial. EBioMedicine 2024; 110:105462. [PMID: 39612654 PMCID: PMC11647478 DOI: 10.1016/j.ebiom.2024.105462] [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: 04/16/2024] [Revised: 10/18/2024] [Accepted: 11/05/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND Glucocorticoids (GC) are potent entrainers of the circadian clock. However, their effects on biological rhythms in chronic human exposure have yet to be studied. Endogenous hypercortisolism (Cushing's Syndrome, CS) is a rare condition in which circadian disruption is sustained by a tumorous source of GC excess, offering the unique opportunity to investigate GC's chronic effects in vivo. METHODS In a 12-month prospective case-control multicentre trial, the daily fluctuations in the number of circulating peripheral blood mononuclear cells (PBMCs) and the time-specific expression of clock-related genes were analysed in a cohort of 68 subjects, 34 affected by CS and 34 matched controls. Cosinor mixed effects model, rhythmicity algorithms and machine learning techniques were applied to the multi-level dataset. FINDINGS Multiple, 5-point daily sampling revealed profound changes in the levels, amplitude, and rhythmicity of several PBMC populations during active CS, only partially restored after remission. Clock gene analyses in isolated PBMCs showed a significant flattening of circadian oscillation of CLOCK, PER1, PER2, PER3, and TIMELESS expression. In active CS, all methods confirmed a loss of rhythmicity of those genes which were circadian in the PBMCs of controls. Most, but not all, genes regained physiological oscillation after remission. Machine learning revealed that while combined time-course sets of clock genes were highly effective in separating patients from controls, immune profiling was efficient even as single time points. INTERPRETATION In conclusion, the oscillation of circulating immune cells is profoundly altered in patients with CS, representing a convergence point of circadian rhythm disruption and metabolic and steroid hormone imbalances. Machine learning techniques proved the superiority of immune profiling over parameters such as cortisol, anthropometric and metabolic variables, and circadian gene expression analysis to identify CS activity. FUNDING The research leading to these results has received funding from the European Union in the context of the National Recovery and Resilience Plan, Investment PE8 - Project Age-It: "Ageing Well in an Ageing Society". This resource was co-financed by the Next Generation EU [DM 1557 11.10.2022], the PRecisiOn Medicine to Target Frailty of Endocrine-metabolic Origin (PROMETEO) project (NET-2018-12365454) by the Italian Ministry of Health, and through internal funding to Sapienza University of Rome.
Collapse
Affiliation(s)
- Valeria Hasenmajer
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy
| | - Emilia Sbardella
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy
| | - Francesca Sciarra
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy
| | - Chiara Simeoli
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Diabetologia, Andrologia e Nutrizione, Università Federico II di Napoli, Naples, Italy
| | - Claudia Pivonello
- Dipartimento di Sanità Pubblica, Università Federico II di Napoli, Naples, Italy
| | - Filippo Ceccato
- Endocrinology, Dep of Medicine, DIMED, University-Hospital of Padova, Padua, Italy; Endocrine Disease Unit, University-Hospital of Padova, Padova, Italy
| | - Riccardo Pofi
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, UK
| | - Marianna Minnetti
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy
| | - Flavio Rizzo
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy
| | - Davide Ferrari
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy
| | - Ilaria Bonaventura
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy
| | - Federica Barbagallo
- Department of Medicine and Surgery, Kore University of Enna, 94100, Enna, Italy
| | - Elisa Giannetta
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy
| | - Danilo Alunni Fegatelli
- Department of Public Health and Infectious Disease, "Sapienza" University of Rome, Rome, Italy
| | - Simone Conia
- Sapienza NLP, Department of Computer, Control and Management Engineering, "Sapienza" University of Rome, Rome, Italy
| | - Roberto Navigli
- Sapienza NLP, Department of Computer, Control and Management Engineering, "Sapienza" University of Rome, Rome, Italy
| | - Giorgio Arnaldi
- Departement of Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro" (PROMISE), Palermo University, 90127, Palermo, Italy
| | - Carla Scaroni
- Endocrinology, Dep of Medicine, DIMED, University-Hospital of Padova, Padua, Italy; Endocrine Disease Unit, University-Hospital of Padova, Padova, Italy
| | - Rosario Pivonello
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Diabetologia, Andrologia e Nutrizione, Università Federico II di Napoli, Naples, Italy
| | - Daniele Gianfrilli
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy
| | - Mary Anna Venneri
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy
| | - Andrea M Isidori
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy; Centre for Rare Diseases (Endo-ERN Accredited), Policlinico Umberto I, Rome, Italy.
| |
Collapse
|
102
|
Wang P, Li F, Sun Y, Li Y, Xie X, Du X, Liu L, Wu Y, Song D, Xiong H, Chen J, Li X. Novel insights into the circadian modulation of lipid metabolism in chicken livers revealed by RNA sequencing and weighted gene co-expression network analysis. Poult Sci 2024; 103:104321. [PMID: 39361997 PMCID: PMC11474196 DOI: 10.1016/j.psj.2024.104321] [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: 07/19/2024] [Revised: 09/06/2024] [Accepted: 09/07/2024] [Indexed: 10/05/2024] Open
Abstract
The circadian clock is crucial for maintaining lipid metabolism homeostasis in mammals. Despite the economic importance of fat content in poultry, research on the regulatory effects and molecular mechanisms of the circadian clock on avian hepatic lipid metabolism has been limited. In this study, we observed significant diurnal variations (P<0.05) in triglyceride (TG), free fatty acids (FFA), fatty acid synthase (FAS), and total cholesterol (TC) levels in the chicken embryonic liver under 12-h light/12-h dark incubation conditions, with TG, FFA, and TC concentrations showing significant cosine rhythmic oscillations (P<0.05). However, such rhythmic variations were not observed under complete darkness incubation conditions. Using transcriptome sequencing technology, we identified 157 genes significantly upregulated at night and 313 genes significantly upregulated during the 12-h light/12-h dark cycle. These circadian differential genes are involved in processes and pathways such as lipid catabolic process regulation, meiotic cell cycle, circadian rhythm regulation, positive regulation of the MAPK cascade, and glycerolipid metabolism. Weighted gene co-expression network analysis (WGCNA) revealed 3 modules-green, blue, and red-that significantly correlate with FFA, FAS, and TG, respectively. Genes within these modules were enriched in processes and pathways including the cell cycle, light stimulus response, circadian rhythm regulation, phosphorylation, positive regulation of the MAPK cascade, and lipid biosynthesis. Notably, we identified ten hub genes, including protein kinase C delta (PRKCD), polo like kinase 4 (PLK4), clock circadian regulator (CLOCK), steroid 5 alpha-reductase 3 (SRD5A3), BUB1 mitotic checkpoint serine/threonine kinase (BUB1B), shugoshin 1 (SGO1), NDC80 kinetochore complex component (NDC80), NIMA related kinase 2 (NEK2), minichromosome maintenance complex component 4 (MCM4), polo like kinase 1 (PLK1), potentially link circadian regulation with lipid metabolic homeostasis. These findings demonstrate the regulatory role of the circadian clock in chicken liver lipid metabolism homeostasis and provide a theoretical basis and molecular targets for optimizing the circadian clock to reduce excessive fat deposition in chickens, which is significant for the healthy development of the poultry industry.
Collapse
Affiliation(s)
- Panlin Wang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Fang Li
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Yanyan Sun
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Yunlei Li
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Xiuyu Xie
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Xue Du
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Lu Liu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Yongshu Wu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Dan Song
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Hui Xiong
- Beijing Seeme Medical Technology Co Ltd, Beijing, 100093, China
| | - Jilan Chen
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| | - Xiangchen Li
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China.
| |
Collapse
|
103
|
Harmsen J, Kotte M, Habets I, Bosschee F, Frenken K, Jorgensen JA, de Kam S, Moonen‐Kornips E, Cissen J, Doligkeit D, van de Weijer T, Erazo‐Tapia E, Buitinga M, Hoeks J, Schrauwen P. Exercise training modifies skeletal muscle clock gene expression but not 24-hour rhythmicity in substrate metabolism of men with insulin resistance. J Physiol 2024; 602:6417-6433. [PMID: 38051503 PMCID: PMC11607886 DOI: 10.1113/jp285523] [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: 08/25/2023] [Accepted: 11/14/2023] [Indexed: 12/07/2023] Open
Abstract
Twenty-four hour rhythmicity in whole-body substrate metabolism, skeletal muscle clock gene expression and mitochondrial respiration is compromised upon insulin resistance. With exercise training known to ameliorate insulin resistance, our objective was to test if exercise training can reinforce diurnal variation in whole-body and skeletal muscle metabolism in men with insulin resistance. In a single-arm longitudinal design, 10 overweight and obese men with insulin resistance performed 12 weeks of high-intensity interval training recurrently in the afternoon (between 14.00 and 18.00 h) and were tested pre- and post-exercise training, while staying in a metabolic research unit for 2 days under free-living conditions with regular meals. On the second days, indirect calorimetry was performed at 08.00, 13.00, 18.00, 23.00 and 04.00 h, muscle biopsies were taken from the vastus lateralis at 08.30, 13.30 and 23.30 h, and blood was drawn at least bi-hourly over 24 h. Participants did not lose body weight over 12 weeks, but improved body composition and exercise capacity. Exercise training resulted in reduced 24-h plasma glucose levels, but did not modify free fatty acid and triacylglycerol levels. Diurnal variation of muscle clock gene expression was modified by exercise training with period genes showing an interaction (time × exercise) effect and reduced mRNA levels at 13.00 h. Exercise training increased mitochondrial respiration without inducing diurnal variation. Twenty-four-hour substrate metabolism and energy expenditure remained unchanged. Future studies should investigate alternative exercise strategies or types of interventions (e.g. diet or drugs aiming at improving insulin sensitivity) for their capacity to reinforce diurnal variation in substrate metabolism and mitochondrial respiration. KEY POINTS: Insulin resistance is associated with blunted 24-h flexibility in whole-body substrate metabolism and skeletal muscle mitochondrial respiration, and disruptions in the skeletal muscle molecular circadian clock. We hypothesized that exercise training modifies 24-h rhythmicity in whole-body substrate metabolism and diurnal variation in skeletal muscle molecular clock and mitochondrial respiration in men with insulin resistance. We found that metabolic inflexibility over 24 h persisted after exercise training, whereas mitochondrial respiration increased independent of time of day. Gene expression of Per1-3 and Rorα in skeletal muscle changed particularly close to the time of day at which exercise training was performed. These results provide the rationale to further investigate the differential metabolic impact of differently timed exercise to treat metabolic defects of insulin resistance that manifest at a particular time of day.
Collapse
Affiliation(s)
- Jan‐Frieder Harmsen
- Department of Nutrition and Movement SciencesNUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Marit Kotte
- Department of Nutrition and Movement SciencesNUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Ivo Habets
- Department of Nutrition and Movement SciencesNUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Frederieke Bosschee
- Department of Nutrition and Movement SciencesNUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Koen Frenken
- Department of Nutrition and Movement SciencesNUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Johanna A. Jorgensen
- Department of Nutrition and Movement SciencesNUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Soraya de Kam
- Department of Nutrition and Movement SciencesNUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Esther Moonen‐Kornips
- Department of Nutrition and Movement SciencesNUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Jochem Cissen
- Department of Nutrition and Movement SciencesNUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Daniel Doligkeit
- Department of Nutrition and Movement SciencesNUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Tineke van de Weijer
- Department of Nutrition and Movement SciencesNUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Edmundo Erazo‐Tapia
- Department of Nutrition and Movement SciencesNUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Mijke Buitinga
- Department of Nutrition and Movement SciencesNUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Joris Hoeks
- Department of Nutrition and Movement SciencesNUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Patrick Schrauwen
- Department of Nutrition and Movement SciencesNUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| |
Collapse
|
104
|
Hebl JT, Velasco J, McHill AW. Work Around the Clock: How Work Hours Induce Social Jetlag and Sleep Deficiency. Sleep Med Clin 2024; 19:569-579. [PMID: 39455178 DOI: 10.1016/j.jsmc.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2024]
Abstract
A growing body of evidence has placed an increasing emphasis on how sleep affects health. Not only does insufficient sleep make one subjectively feel worse, but is associated with chronic diseases that are considered epidemics in industrialized nations. This is partly caused by the growing need for prolonged work and social schedules, exemplified by shift work, late-night weekends, and early morning work/school start times (social jetlag). Here, we consider fundamental relationships between the circadian clock and biologic processes and discuss how common practices, such as shift work and social jetlag, contribute to sleep disruption, circadian misalignment, and adverse health outcomes.
Collapse
Affiliation(s)
- Joseph T Hebl
- Oregon Health and Sciences University, School of Medicine, 3455 SW US Veterans Hospital Road, Mailcode: SN-ORD, Portland, OR 97239, USA
| | - Josie Velasco
- Sleep, Chronobiology, and Health Laboratory, School of Nursing, Oregon Health & Science University, 3455 SW US Veterans Hospital Road, Mailcode: SN-ORD, Portland, OR 97239, USA; Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, 3455 SW US Veterans Hospital Road, Mailcode: SN-ORD, Portland, OR 97239, USA
| | - Andrew W McHill
- Sleep, Chronobiology, and Health Laboratory, School of Nursing, Oregon Health & Science University, 3455 SW US Veterans Hospital Road, Mailcode: SN-ORD, Portland, OR 97239, USA; Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, 3455 SW US Veterans Hospital Road, Mailcode: SN-ORD, Portland, OR 97239, USA.
| |
Collapse
|
105
|
Agamme ALDA, Tufik S, Torterolo P, D'Almeida V. Effects of Paradoxical Sleep Deprivation on MCH and Hypocretin Systems. Sleep Sci 2024; 17:e392-e400. [PMID: 39698172 PMCID: PMC11651861 DOI: 10.1055/s-0044-1782171] [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: 12/17/2022] [Accepted: 12/20/2023] [Indexed: 12/20/2024] Open
Abstract
Melanin-concentrating hormone (MCH) and hypocretins (Hcrt) 1 and 2 are neuropeptides synthesized in the lateral hypothalamic area by neurons that are critical in the regulation of sleep and wakefulness. Their receptors are located in the same cerebral regions, including the frontal cortex and hippocampus. The present study aimed to assess whether 96 hours of paradoxical sleep deprivation alters the functioning of the MCH and hypocretin systems. To do this, in control rats with normal sleep (CTL) and in rats that were deprived of paradoxical sleep (SD), we quantified the following parameters: 1) levels of MCH and hypocretin-1 in the cerebrospinal fluid (CSF); 2) expression of the prepro-MCH ( Pmch ) and prepro-hypocretin ( Hcrt ) genes in the hypothalamus; 3) expression of the Mchr1 and Hcrtr1 genes in the frontal cortex and hippocampus; and 4) expression of the Hcrtr2 gene in the hippocampus. These measures were performed at 6 Zeitgeber time (ZT) points of the day (ZTs: 0, 4, 8, 12, 16, and 20). In the SD group, we found higher levels of MCH in the CSF at the beginning of the dark phase. In the frontal cortex, sleep deprivation decreased the expression of Hcrtr1 at ZT0 . Moreover, we identified significant differences between the light and dark phases in the expression of Mchr1 and Hcrtr1 , but only in the CTL animals . We conclude that there is a day/night modulation in the expression of components of the MCH and hypocretin systems, and this profile is affected by paradoxical sleep deprivation.
Collapse
Affiliation(s)
- Ana Luiza Dias Abdo Agamme
- Departamento de Psicobiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Sergio Tufik
- Departamento de Psicobiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Pablo Torterolo
- Department of Physiology, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Vânia D'Almeida
- Departamento de Psicobiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
106
|
Jasińska M, Jasek-Gajda E, Ziaja M, Litwin JA, Lis GJ, Pyza E. Light-Modulated Circadian Synaptic Plasticity in the Somatosensory Cortex: Link to Locomotor Activity. Int J Mol Sci 2024; 25:12870. [PMID: 39684579 DOI: 10.3390/ijms252312870] [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] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
The circadian clock controls various physiological processes, including synaptic function and neuronal activity, affecting the functioning of the entire organism. Light is an important external factor regulating the day-night cycle. This study examined the effects of the circadian clock and light on synaptic plasticity, and explored how locomotor activity contributes to these processes. We analyzed synaptic protein expression and excitatory synapse density in the somatosensory cortex of mice from four groups exposed to different lighting conditions (LD 12:12, DD, LD 16:8, and LL). Locomotor activity was assessed through individual wheel-running monitoring. To explore daily and circadian changes in synaptic proteins, we performed double-immunofluorescence labeling and laser scanning confocal microscopy imaging, targeting three pairs of presynaptic and postsynaptic proteins (Synaptophysin 1/PSD95, Piccolo/Homer 1, Neurexins/PICK1). Excitatory synapse density was evaluated by co-labeling presynaptic and postsynaptic markers. Our results demonstrated that all the analyzed synaptic proteins exhibited circadian regulation modulated by light. Under constant light conditions, only Piccolo and Homer 1 showed rhythmicity. Locomotor activity was also associated with the circadian clock's effects on synaptic proteins, showing a stronger connection to changes in postsynaptic protein levels. Excitatory synapse density peaked during the day/subjective day and exhibited an inverse relationship with locomotor activity. Continued light exposure disrupted cyclic changes in synapse density but kept it consistently elevated. These findings underscore the crucial roles of light and locomotor activity in regulating synaptic plasticity.
Collapse
Affiliation(s)
- Małgorzata Jasińska
- Department of Histology, Jagiellonian University Medical College, 31-034 Krakow, Poland
| | - Ewa Jasek-Gajda
- Department of Histology, Jagiellonian University Medical College, 31-034 Krakow, Poland
| | - Marek Ziaja
- Department of Histology, Jagiellonian University Medical College, 31-034 Krakow, Poland
| | - Jan A Litwin
- Department of Histology, Jagiellonian University Medical College, 31-034 Krakow, Poland
| | - Grzegorz J Lis
- Department of Histology, Jagiellonian University Medical College, 31-034 Krakow, Poland
| | - Elżbieta Pyza
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, 30-387 Krakow, Poland
| |
Collapse
|
107
|
Ryu JE, Shim KW, Roh HW, Park M, Lee JH, Kim EY. Circadian regulation of endoplasmic reticulum calcium response in cultured mouse astrocytes. eLife 2024; 13:RP96357. [PMID: 39601391 PMCID: PMC11602189 DOI: 10.7554/elife.96357] [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: 11/29/2024] Open
Abstract
The circadian clock, an internal time-keeping system orchestrates 24 hr rhythms in physiology and behavior by regulating rhythmic transcription in cells. Astrocytes, the most abundant glial cells, play crucial roles in CNS functions, but the impact of the circadian clock on astrocyte functions remains largely unexplored. In this study, we identified 412 circadian rhythmic transcripts in cultured mouse cortical astrocytes through RNA sequencing. Gene Ontology analysis indicated that genes involved in Ca2+ homeostasis are under circadian control. Notably, Herpud1 (Herp) exhibited robust circadian rhythmicity at both mRNA and protein levels, a rhythm disrupted in astrocytes lacking the circadian transcription factor, BMAL1. HERP regulated endoplasmic reticulum (ER) Ca2+ release by modulating the degradation of inositol 1,4,5-trisphosphate receptors (ITPRs). ATP-stimulated ER Ca2+ release varied with the circadian phase, being more pronounced at subjective night phase, likely due to the rhythmic expression of ITPR2. Correspondingly, ATP-stimulated cytosolic Ca2+ increases were heightened at the subjective night phase. This rhythmic ER Ca2+ response led to circadian phase-dependent variations in the phosphorylation of Connexin 43 (Ser368) and gap junctional communication. Given the role of gap junction channel (GJC) in propagating Ca2+ signals, we suggest that this circadian regulation of ER Ca2+ responses could affect astrocytic modulation of synaptic activity according to the time of day. Overall, our study enhances the understanding of how the circadian clock influences astrocyte function in the CNS, shedding light on their potential role in daily variations of brain activity and health.
Collapse
Affiliation(s)
- Ji Eun Ryu
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of MedicineSuwonRepublic of Korea
- Department of Brain Science, Ajou University School of MedicineSuwonRepublic of Korea
| | - Kyu-Won Shim
- Interdisciplinary Program in Bioinformatics, Seoul National UniversitySeoulRepublic of Korea
| | - Hyun Woong Roh
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of MedicineSuwonRepublic of Korea
- Department of Psychiatry, Ajou University School of MedicineSuwonRepublic of Korea
| | - Minsung Park
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of MedicineSuwonRepublic of Korea
- Department of Brain Science, Ajou University School of MedicineSuwonRepublic of Korea
| | - Jae-Hyung Lee
- Department of Oral Microbiology, College of Dentistry, Kyung Hee UniversitySeoulRepublic of Korea
| | - Eun Young Kim
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of MedicineSuwonRepublic of Korea
- Department of Brain Science, Ajou University School of MedicineSuwonRepublic of Korea
| |
Collapse
|
108
|
Lian JW, Li SY, Clarke RB, Howell SJ, Meng QJ. Can we utilise the circadian clock to target cancer stem cells? Cancer Lett 2024; 611:217360. [PMID: 39608441 DOI: 10.1016/j.canlet.2024.217360] [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: 07/21/2024] [Revised: 11/03/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
The 24-hourly circadian clock has been implicated in the regulation of multiple cancer hallmarks and characteristics. Cancer stem cells (CSCs) are a small but significant population of cells within many cancers, characterised by their self-renewal and clonogenic capacities. Increasing evidence points to CSCs having prominent roles in metastasis and drug resistance. However, it remains largely unknown how circadian clocks are involved with CSCs and what implications these interactions have for cancer progression and therapeutics. In this review, we examine the growing evidence on the role of circadian clocks in CSCs and discuss the potential therapeutic implications. This opens up new opportunities to target CSCs through various chronotherapeutic approaches, potentially improving clinical cancer outcomes. We propose different scenarios in which targeting circadian clocks in CSCs or their surrounding microenvironment could be developed into effective therapeutic strategies, including: (1) direct pharmacological targeting of core clock molecules, (2) optimising the timing of systemic anticancer therapies, and (3) targeting the neighbouring cells or systemic factors that influence tumour cells in a circadian-dependent manner.
Collapse
Affiliation(s)
- Jia-Wen Lian
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Shi-Yang Li
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Robert B Clarke
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Sacha J Howell
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Qing-Jun Meng
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.
| |
Collapse
|
109
|
Smith JG. Emerging interactions between circadian rhythms and extracellular vesicles. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 393:73-93. [PMID: 40390464 DOI: 10.1016/bs.ircmb.2024.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2025]
Abstract
Circadian rhythms are present across species, tuning internal processes to daily changes in the environment. Driven by genetically encoded circadian clocks present throughout the body, and modulated by external inputs, the circadian system is a key player in metabolic control. However, the molecular mediators underlying coordination between cells and tissues are not well known. Extracellular vesicles (EVs) have emerged over recent years as important players in cell-cell and organ-organ communication, however the influence of circadian rhythms on EVs is not yet understood. Research into this area is still scarce, yet already offers glimpses into the potential impact of circadian rhythms on EV biology. In this review, recent discoveries that reveal, directly or indirectly, a potential role for circadian rhythms in EV abundance, properties, cargo and signalling functions are first discussed. Next, the feedback of EV signalling on circadian clocks is considered. Last, unanswered questions regarding the interaction between circadian rhythms and EVs are examined alongside potential approaches to address them. Overall, the circadian impact on EV signalling is an exciting yet understudied aspect that warrants further investigation.
Collapse
Affiliation(s)
- Jacob G Smith
- Department of Cell Biology, Physiology and Immunology, School of Biology, University of Barcelona, Barcelona, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain.
| |
Collapse
|
110
|
Jiang Y, Shi J, Tai J, Yan L. Circadian Regulation in Diurnal Mammals: Neural Mechanisms and Implications in Translational Research. BIOLOGY 2024; 13:958. [PMID: 39765625 PMCID: PMC11727363 DOI: 10.3390/biology13120958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/16/2024] [Accepted: 11/19/2024] [Indexed: 01/15/2025]
Abstract
Diurnal and nocturnal mammals have evolved unique behavioral and physiological adaptations to optimize survival for their day- or night-active lifestyle. The mechanisms underlying the opposite activity patterns are not fully understood but likely involve the interplay between the circadian time-keeping system and various arousal- or sleep-promoting factors, e.g., light or melatonin. Although the circadian systems between the two chronotypes share considerable similarities, the phase relationships between the principal and subordinate oscillators are chronotype-specific. While light promotes arousal and wakefulness in diurnal species like us, it induces sleep in nocturnal ones. Similarly, melatonin, the hormone of darkness, is commonly used as a hypnotic in humans but is secreted in the active phase of nocturnal animals. Thus, the difference between the two chronotypes is more complex than a simple reversal, as the physiological and neurological processes in diurnal mammals during the day are not equivalent to that of nocturnal ones at night. Such chronotype differences could present a significant translational gap when applying research findings obtained from nocturnal rodents to diurnal humans. The potential advantages of diurnal models are being discussed in a few sleep-related conditions including familial natural short sleep (FNSS), obstructive sleep apnea (OSA), and Smith-Magenis syndrome (SMS). Considering the difference in chronotype, a diurnal model will be more adequate for revealing the physiology and physiopathology pertaining to human health and disease, especially in conditions in which circadian rhythm disruption, altered photic response, or melatonin secretion is involved. We hope the recent advances in gene editing in diurnal rodents will promote greater utility of the diurnal models in basic and translational research.
Collapse
Affiliation(s)
- Yirun Jiang
- Department of Otolaryngology, Head and Neck Surgery, Capital Institute of Pediatrics, Beijing 100020, China; (Y.J.); (J.T.)
| | - Jiaming Shi
- Department of Psychology, Michigan State University, East Lansing, MI 48824, USA;
| | - Jun Tai
- Department of Otolaryngology, Head and Neck Surgery, Capital Institute of Pediatrics, Beijing 100020, China; (Y.J.); (J.T.)
| | - Lily Yan
- Department of Psychology, Michigan State University, East Lansing, MI 48824, USA;
- Neuroscience Program, Interdisciplinary Science & Technology Building (ISTB), Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
111
|
Barko K, Shelton MA, DePoy LM, Gayden-Kozel J, Kim SM, Puig S, Xue X, Parekh PK, Tseng GC, Williams BR, Oliver-Smith J, Zhu X, Freyberg Z, Logan RW. Sex-specific Regulation of Fentanyl Reward by the Circadian Transcription Factor NPAS2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.12.623242. [PMID: 39605449 PMCID: PMC11601467 DOI: 10.1101/2024.11.12.623242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Synthetic opioids like fentanyl are highly potent and prevalent in the illicit drug market, leading to tolerance, dependence, and opioid use disorder (OUD). Chronic opioid use disrupts sleep and circadian rhythms, which persist even during treatment and abstinence, increasing the risk of relapse. The body's molecular clock, regulated by transcriptional and translational feedback loops, controls various physiological processes, including the expression of endogenous opioids and their receptors. The circadian transcription factor NPAS2, highly expressed in the nucleus accumbens, may have a crucial function in opioid-related behaviors. Our study found sex-specific roles for NPAS2-mediated reward behaviors in male and female mice, including in fentanyl seeking and craving. We also identified specific cell types and transcriptional targets in the nucleus accumbens of both mice and humans by which NPAS2 may mediate the impact of fentanyl on brain physiology and in opioid reward-related behaviors. Ultimately, our findings begin to uncover the mechanisms underlying circadian rhythm dysfunction and opioid addiction.
Collapse
|
112
|
Li X, He Y, Wang D, Momeni MR. Chronobiological disruptions: unravelling the interplay of shift work, circadian rhythms, and vascular health in the context of stroke risk. Clin Exp Med 2024; 25:6. [PMID: 39541048 PMCID: PMC11564290 DOI: 10.1007/s10238-024-01514-w] [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: 06/20/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Shift work, particularly night shifts, disrupts circadian rhythms and increases stroke risk. This manuscript explores the mechanisms connecting shift work with stroke, focusing on circadian rhythms, hypertension, and diabetes. The circadian system, controlled by different mechanisms including central and peripheral clock genes, suprachiasmatic nuclei (SCN), and pineal gland (through melatonin production), regulates body functions and responds to environmental signals. Disruptions in this system affect endothelial cells, leading to blood pressure issues. Type 2 diabetes mellitus (T2DM) is significantly associated with night shifts, with circadian disturbances affecting glucose metabolism, insulin sensitivity, and hormone regulation. The manuscript examines the relationship between melatonin, insulin, and glucose balance, highlighting pathways that link T2DM to stroke risk. Additionally, dyslipidemia, particularly reduced HDL-c levels, results from shift work and contributes to stroke development. High lipid levels cause oxidative stress, inflammation, and endothelial dysfunction, increasing cerebrovascular risks. The manuscript details the effects of dyslipidemia on brain functions, including disruptions in blood flow, blood-brain barrier integrity, and neural cell death. This comprehensive analysis emphasizes the complex interplay of circadian disruption, hypertension, diabetes, and dyslipidemia in increasing stroke risk among shift workers. Understanding these mechanisms is essential for developing targeted interventions to reduce stroke susceptibility and improve cerebrovascular health in this vulnerable population.
Collapse
Affiliation(s)
- Xiaohong Li
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yanjin He
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Dawu Wang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | | |
Collapse
|
113
|
Lawrence JH, Patel A, King MW, Nadarajah CJ, Daneman R, Musiek ES. Microglia drive diurnal variation in susceptibility to inflammatory blood-brain barrier breakdown. JCI Insight 2024; 9:e180081. [PMID: 39513366 PMCID: PMC11601573 DOI: 10.1172/jci.insight.180081] [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: 02/07/2024] [Accepted: 09/12/2024] [Indexed: 11/15/2024] Open
Abstract
The blood-brain barrier (BBB) is critical for maintaining brain homeostasis but is susceptible to inflammatory dysfunction. While transporter-dependent efflux of some lipophilic substrates across the BBB shows circadian variation due to rhythmic transporter expression, basal transporter-independent permeability and leakage is nonrhythmic. Whether daily timing influences BBB permeability in response to inflammation is unknown. Here, we induced systemic inflammation through repeated LPS injections either in the morning (ZT1) or evening (ZT13) under standard lighting conditions; we then examined BBB permeability to a polar molecule that is not a transporter substrate, sodium fluorescein. We observed clear diurnal variation in inflammatory BBB permeability, with a striking increase in paracellular leak across the BBB specifically following evening LPS injection. Evening LPS led to persisting glia activation as well as inflammation in the brain that was not observed in the periphery. The exaggerated evening neuroinflammation and BBB disruption were suppressed by microglial depletion or through keeping mice in constant darkness. Our data show that diurnal rhythms in microglial inflammatory responses to LPS drive daily variability in BBB breakdown and reveal time of day as a key regulator of inflammatory BBB disruption.
Collapse
Affiliation(s)
- Jennifer H. Lawrence
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Asha Patel
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Melvin W. King
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Collin J. Nadarajah
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Richard Daneman
- Department of Pharmacology, UCSD, San Diego, California, USA
| | - Erik S. Musiek
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
- Center On Biological Rhythms And Sleep (COBRAS), Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
114
|
Chen YL, Wang R, Pang R, Sun ZP, He XL, Tang WH, Ou JY, Yi HM, Cheng X, Chen JH, Yu Y, Ren CH, Wang QJ, Zhang ZJ. Transcriptome-Based Revelation of the Effects of Sleep Deprivation on Hepatic Metabolic Rhythms in Tibetan Sheep ( Ovis aries). Animals (Basel) 2024; 14:3165. [PMID: 39595218 PMCID: PMC11591132 DOI: 10.3390/ani14223165] [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: 09/30/2024] [Revised: 10/26/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
Sleep deprivation (SD) disrupts circadian rhythms; however, its effects on SD and the mechanisms involved require further investigation. Previous studies on SD were mainly conducted on rodents, such as mice, with few studies on its effects on the liver of large diurnal animals, such as sheep. In this study, we used a Tibetan sheep model for the first time to investigate the effects of SD on the liver by exposing Tibetan sheep (Ovis aries) to 7 days of SD (6 h/day) and performed transcriptome sequencing analysis on liver samples taken at 4 h intervals over 24 h. The results revealed that SD significantly altered the circadian expression of genes and their expression patterns in the liver of Tibetan sheep. Enrichment analysis of the circadian rhythm-altered genes revealed changes in the pathways related to lipid metabolism in the liver. Further evidence from serum markers and gene expression analyses using qualitative real-time polymerase chain reaction and Oil Red O and apoptosis staining indicated that SD leads to abnormal lipid metabolism in the liver, potentially causing liver damage. Therefore, our results suggest that SD disrupts the circadian rhythms of metabolism-related genes in the Tibetan sheep liver, thereby affecting metabolic homeostasis.
Collapse
Affiliation(s)
- Ya-Le Chen
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.-L.C.); (R.W.); (R.P.); (X.-L.H.); (W.-H.T.); (J.-Y.O.); (H.-M.Y.); (X.C.); (C.-H.R.)
| | - Ru Wang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.-L.C.); (R.W.); (R.P.); (X.-L.H.); (W.-H.T.); (J.-Y.O.); (H.-M.Y.); (X.C.); (C.-H.R.)
| | - Rui Pang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.-L.C.); (R.W.); (R.P.); (X.-L.H.); (W.-H.T.); (J.-Y.O.); (H.-M.Y.); (X.C.); (C.-H.R.)
| | - Zhi-Peng Sun
- Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing 400715, China;
| | - Xiao-Long He
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.-L.C.); (R.W.); (R.P.); (X.-L.H.); (W.-H.T.); (J.-Y.O.); (H.-M.Y.); (X.C.); (C.-H.R.)
| | - Wen-Hui Tang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.-L.C.); (R.W.); (R.P.); (X.-L.H.); (W.-H.T.); (J.-Y.O.); (H.-M.Y.); (X.C.); (C.-H.R.)
| | - Jing-Yu Ou
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.-L.C.); (R.W.); (R.P.); (X.-L.H.); (W.-H.T.); (J.-Y.O.); (H.-M.Y.); (X.C.); (C.-H.R.)
| | - Huan-Ming Yi
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.-L.C.); (R.W.); (R.P.); (X.-L.H.); (W.-H.T.); (J.-Y.O.); (H.-M.Y.); (X.C.); (C.-H.R.)
| | - Xiao Cheng
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.-L.C.); (R.W.); (R.P.); (X.-L.H.); (W.-H.T.); (J.-Y.O.); (H.-M.Y.); (X.C.); (C.-H.R.)
| | - Jia-Hong Chen
- Center of Agriculture Technology Cooperation and Promotion of Dingyuan County, Chuzhou 233200, China;
| | - Yang Yu
- Qinghai Provincial Key Laboratory of Adaptive Management on Alpine Grassland, Qinghai Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining 810016, China;
| | - Chun-Huan Ren
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.-L.C.); (R.W.); (R.P.); (X.-L.H.); (W.-H.T.); (J.-Y.O.); (H.-M.Y.); (X.C.); (C.-H.R.)
- Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing 400715, China;
| | - Qiang-Jun Wang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.-L.C.); (R.W.); (R.P.); (X.-L.H.); (W.-H.T.); (J.-Y.O.); (H.-M.Y.); (X.C.); (C.-H.R.)
| | - Zi-Jun Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.-L.C.); (R.W.); (R.P.); (X.-L.H.); (W.-H.T.); (J.-Y.O.); (H.-M.Y.); (X.C.); (C.-H.R.)
- Center of Agriculture Technology Cooperation and Promotion of Dingyuan County, Chuzhou 233200, China;
| |
Collapse
|
115
|
Schwarz JE, Mrčela A, Lahens NF, Li Y, Hsu C, Grant GR, Skarke C, Zhang SL, Sehgal A. Evidence for a role of human blood-borne factors in mediating age-associated changes in molecular circadian rhythms. eLife 2024; 12:RP88322. [PMID: 39485282 PMCID: PMC11530234 DOI: 10.7554/elife.88322] [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: 11/03/2024] Open
Abstract
Aging is associated with a number of physiologic changes including perturbed circadian rhythms; however, mechanisms by which rhythms are altered remain unknown. To test the idea that circulating factors mediate age-dependent changes in peripheral rhythms, we compared the ability of human serum from young and old individuals to synchronize circadian rhythms in culture. We collected blood from apparently healthy young (age 25-30) and old (age 70-76) individuals at 14:00 and used the serum to synchronize cultured fibroblasts. We found that young and old sera are equally competent at initiating robust ~24 hr oscillations of a luciferase reporter driven by clock gene promoter. However, cyclic gene expression is affected, such that young and old sera promote cycling of different sets of genes. Genes that lose rhythmicity with old serum entrainment are associated with oxidative phosphorylation and Alzheimer's Disease as identified by STRING and IPA analyses. Conversely, the expression of cycling genes associated with cholesterol biosynthesis increased in the cells entrained with old serum. Genes involved in the cell cycle and transcription/translation remain rhythmic in both conditions. We did not observe a global difference in the distribution of phase between groups, but found that peak expression of several clock-controlled genes (PER3, NR1D1, NR1D2, CRY1, CRY2, and TEF) lagged in the cells synchronized ex vivo with old serum. Taken together, these findings demonstrate that age-dependent blood-borne factors affect circadian rhythms in peripheral cells and have the potential to impact health and disease via maintaining or disrupting rhythms respectively.
Collapse
Affiliation(s)
- Jessica E Schwarz
- Howard Hughes Medical Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Antonijo Mrčela
- Institute for Translational Medicine and Therapeutics (ITMAT), Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Nicholas F Lahens
- Institute for Translational Medicine and Therapeutics (ITMAT), Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Yongjun Li
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Cynthia Hsu
- Howard Hughes Medical Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Gregory R Grant
- Institute for Translational Medicine and Therapeutics (ITMAT), Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Department of Genetics, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Carsten Skarke
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Institute for Translational Medicine and Therapeutics (ITMAT), Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Shirley L Zhang
- Howard Hughes Medical Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Amita Sehgal
- Howard Hughes Medical Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
116
|
Deyurka NA, Navigatore-Fonzo LS, Coria-Lucero CD, Ferramola ML, Delgado SM, Lacoste MG, Anzulovich AC. Aging abolishes circadian rhythms and disrupts temporal organization of antioxidant-prooxidant status, endogenous clock activity and neurotrophin gene expression in the rat temporal cortex. Neuroscience 2024; 559:125-138. [PMID: 39244007 DOI: 10.1016/j.neuroscience.2024.09.008] [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: 04/09/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
Disruption of circadian rhythms contributes to deficits in cognitive functions during aging. Up to date, the biochemical, molecular and chronobiological bases of such deterioration have not been completely elucidated. Here, we aim: 1) to investigate the endogenous nature of 24 h-rhythms of antioxidant defenses, oxidative stress, clocḱ's, and neurotrophic factors expression, in the rat temporal cortex (TC), and 2) to study the consequences of aging on the circadian organization of those factors. We observed a circadian organization of antioxidant enzymes activity, lipoperoxidation and the clock, BMAL1 and RORa, proteins, in the TC of young rats. Such temporal organization suggests the existence of a two-way communication among clock transcription factors and antioxidant defenses. This might generate the rhythmic and circadian expression of Bdnf and Rc3 genes involved in the TC-depending cognitive function. Noteworthy, such circadian organization disappears in the TC of aged rats. Aging also reduces glutathione peroxidase activity and expression, and it increases lipid peroxidation, throughout a 24 h-period. An increased oxidative stress makes the cellular redox environment change into an oxidative status which alters the endogenous clock activity and disrupts the circadian organization of, at least part, of the molecular basis of the synaptic plasticity in the TC.
Collapse
Affiliation(s)
- Nicolás Andrés Deyurka
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research-San Luis (IMIBIO-SL), National Council of Science and Technology (CONICET), National University of San Luis (UNSL), Av Ejército de los Andes N° 950, D5700HHW, San Luis, Argentina; Faculty of Chemistry, Biochemistry and Pharmacy, National University of San Luis (UNSL), Av Ejército de los Andes N° 950, D5700HHW, San Luis, Argentina
| | - Lorena Silvina Navigatore-Fonzo
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research-San Luis (IMIBIO-SL), National Council of Science and Technology (CONICET), National University of San Luis (UNSL), Av Ejército de los Andes N° 950, D5700HHW, San Luis, Argentina; Faculty of Chemistry, Biochemistry and Pharmacy, National University of San Luis (UNSL), Av Ejército de los Andes N° 950, D5700HHW, San Luis, Argentina
| | - Cinthia Daiana Coria-Lucero
- Faculty of Health Sciences, National University of San Luis (UNSL), Av Ejército de los Andes N° 950, D5700HHW, San Luis, Argentina
| | - Mariana Lucila Ferramola
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research-San Luis (IMIBIO-SL), National Council of Science and Technology (CONICET), National University of San Luis (UNSL), Av Ejército de los Andes N° 950, D5700HHW, San Luis, Argentina; Faculty of Chemistry, Biochemistry and Pharmacy, National University of San Luis (UNSL), Av Ejército de los Andes N° 950, D5700HHW, San Luis, Argentina
| | - Silvia Marcela Delgado
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research-San Luis (IMIBIO-SL), National Council of Science and Technology (CONICET), National University of San Luis (UNSL), Av Ejército de los Andes N° 950, D5700HHW, San Luis, Argentina; Faculty of Chemistry, Biochemistry and Pharmacy, National University of San Luis (UNSL), Av Ejército de los Andes N° 950, D5700HHW, San Luis, Argentina
| | - María Gabriela Lacoste
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research-San Luis (IMIBIO-SL), National Council of Science and Technology (CONICET), National University of San Luis (UNSL), Av Ejército de los Andes N° 950, D5700HHW, San Luis, Argentina; Faculty of Chemistry, Biochemistry and Pharmacy, National University of San Luis (UNSL), Av Ejército de los Andes N° 950, D5700HHW, San Luis, Argentina.
| | - Ana Cecilia Anzulovich
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research-San Luis (IMIBIO-SL), National Council of Science and Technology (CONICET), National University of San Luis (UNSL), Av Ejército de los Andes N° 950, D5700HHW, San Luis, Argentina; Faculty of Chemistry, Biochemistry and Pharmacy, National University of San Luis (UNSL), Av Ejército de los Andes N° 950, D5700HHW, San Luis, Argentina.
| |
Collapse
|
117
|
Benjamin JI, Pati P, Luong T, Liu X, De Miguel C, Pollock JS, Pollock DM. Chronic mistimed feeding results in renal fibrosis and disrupted circadian blood pressure rhythms. Am J Physiol Renal Physiol 2024; 327:F683-F696. [PMID: 39205662 PMCID: PMC11563648 DOI: 10.1152/ajprenal.00047.2024] [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: 02/13/2024] [Revised: 08/12/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024] Open
Abstract
Circadian disruption is a disturbance in biological timing, which can occur within or between different organizational levels, ranging from molecular rhythms within specific cells to the misalignment of behavioral and environmental cycles. Previous work from our group showed that less than 1 wk of food restriction to the light (inactive) period is sufficient to invert diurnal blood pressure rhythms in mice. However, kidney excretory rhythms and functions remained aligned with the light-dark cycle. Shift workers have an increased risk of cardiovascular disease that may different between sexes and often have irregular mealtimes, making the possibility of mistimed feeding as a potential contributor to the development of kidney disease. Thus, we hypothesized that chronic mistimed food intake would result in adverse cardiorenal effects, with sex differences in severity. Here, we show that chronic circadian disruption via mistimed feeding results in renal fibrosis and aortic stiffness in a sex-dependent manner. Our results indicate the importance of meal timing for the maintenance of blood pressure rhythms and kidney function, particularly in males. Our results also demonstrate that females are better able to acclimate to circadian-related behavioral change. NEW & NOTEWORTHY Circadian disruption through mistimed feeding resulted in nondipping blood pressure, renal fibrosis, and arterial stiffness that were less severe in females versus males. Mice fed exclusively during the daytime maintain their circadian rhythms of locomotor activity regardless of their loss of blood pressure rhythms. Although these mice ate less food, they maintained body weight, suggesting inefficiencies in overall metabolism. These findings demonstrate the importance of maintaining optimal food intake patterns to prevent cardiorenal pathophysiology.
Collapse
Affiliation(s)
- Jazmine I Benjamin
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Paramita Pati
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Tha Luong
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Xiaofen Liu
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Carmen De Miguel
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Jennifer S Pollock
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - David M Pollock
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
118
|
Tauber E. Uncovering the circadian transcriptome of Nasonia vitripennis: insights into a non-canonical insect model. Proc Biol Sci 2024; 291:20241848. [PMID: 39591997 DOI: 10.1098/rspb.2024.1848] [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: 08/01/2024] [Revised: 10/27/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
The study of the circadian clock has greatly benefited from using Drosophila as a model system. Yet accumulating evidence suggests that the fly might not be the canonical insect model. Here, I have analysed the circadian transcriptome of the jewel wasp Nasonia vitripennis by using RNA-seq in both constant darkness and constant light (in contrast to flies, the wasps are rhythmic under continuous light). I identify approximately 6% of the transcriptome as cycling under constant conditions, revealing a bimodal distribution of phases and low cycling amplitude. I examine the biological processes under circadian control in Nasonia, identifying clock control of functions such as metabolism, light response and a variety of neural processes, drawing comparisons between Nasonia and Drosophila. Although there was little similarity between cycling genes in Drosophila and Nasonia, the functions fulfilled by cycling transcripts were similar in both species. Interestingly, of the known Drosophila core clock genes, only Pdp1e, shaggy and Clock showed significant cycling in Nasonia, highlighting the potential diversity in molecular clock mechanisms across insect species.
Collapse
Affiliation(s)
- Eran Tauber
- Department of Evolutionary & Environmental Biology, Institute of Evolution, University of Haifa, Haifa 3498838, Israel
| |
Collapse
|
119
|
Lou L, Tu ZJ, Lahondère C, Vinauger C. Rhythms in insect olfactory systems: underlying mechanisms and outstanding questions. J Exp Biol 2024; 227:jeb244182. [PMID: 39508241 PMCID: PMC11574354 DOI: 10.1242/jeb.244182] [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: 11/08/2024]
Abstract
Olfaction is a critical sensory modality for invertebrates, and it mediates a wide range of behaviors and physiological processes. Like most living organisms, insects live in rhythmic environments: the succession of nights and days is accompanied by cyclic variations in light intensity and temperature, as well as in the availability of resources and the activity of predators. Responding to olfactory cues in the proper temporal context is thus highly adaptive and allows for the efficient allocation of energy resources. Given the agricultural or epidemiological importance of some insect species, understanding olfactory rhythms is critical for the development of effective control strategies. Although the vinegar fly Drosophila melanogaster has been a classical model for the study of olfaction and circadian rhythms, recent studies focusing on non-model species have expanded our understanding of insect olfactory rhythms. Additionally, recent evidence revealing receptor co-expression by sensory neurons has brought about an ongoing paradigm shift in our understanding of insect olfaction, making it timely to review the state of our knowledge on olfactory rhythms and identify critical future directions for the field. In this Review, we discuss the multiple biological scales at which insect olfactory rhythms are being analyzed, and identify outstanding questions.
Collapse
Affiliation(s)
- Lan Lou
- Department of Biochemistry, Virginia Tech, Blacksburg, VA 24061, USA
- Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA 24061, USA
- Center for Emerging Zoonotic and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA 24061, USA
| | - Zhijian Jake Tu
- Department of Biochemistry, Virginia Tech, Blacksburg, VA 24061, USA
- Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA 24061, USA
- Center for Emerging Zoonotic and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA 24061, USA
| | - Chloé Lahondère
- Department of Biochemistry, Virginia Tech, Blacksburg, VA 24061, USA
- Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA 24061, USA
- Center for Emerging Zoonotic and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA 24061, USA
- Global Change Center, Virginia Tech, Blacksburg, VA 24061, USA
| | - Clément Vinauger
- Department of Biochemistry, Virginia Tech, Blacksburg, VA 24061, USA
- Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA 24061, USA
- Center for Emerging Zoonotic and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
120
|
Zhou Z, Zhang R, Zhang Y, Xu Y, Wang R, Chen S, Lv Y, Chen Y, Ren Y, Luo P, Cheng Q, Xu H, Weng S, Zuo A, Ba Y, Liu S, Han X, Liu Z. Circadian disruption in cancer hallmarks: Novel insight into the molecular mechanisms of tumorigenesis and cancer treatment. Cancer Lett 2024; 604:217273. [PMID: 39306230 DOI: 10.1016/j.canlet.2024.217273] [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: 06/27/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
Circadian rhythms are 24-h rhythms governing temporal organization of behavior and physiology generated by molecular clocks composed of autoregulatory transcription-translation feedback loops (TTFLs). Disruption of circadian rhythms leads to a spectrum of pathologies, including cancer by triggering or being involved in different hallmarks. Clock control of phenotypic plasticity involved in tumorigenesis operates in aberrant dedifferentiating to progenitor-like cell states, generation of cancer stem cells (CSCs) and epithelial-to-mesenchymal transition (EMT) events. Circadian rhythms might act as candidates for regulatory mechanisms of cellular senescent and functional determinants of senescence-associated secretory phenotype (SASP). Reciprocal control between clock and epigenetics sheds light on post-transcriptional regulation of circadian rhythms and opens avenues for novel anti-cancer strategies. Additionally, disrupting circadian rhythms influences microbiota communities that could be associated with altered homeostasis contributing to cancer development. Herein, we summarize recent advances in support of the nexus between disruptions of circadian rhythms and cancer hallmarks of new dimensions, thus providing novel perspectives on potentially effective treatment approaches for cancer management.
Collapse
Affiliation(s)
- Zhaokai Zhou
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ruiqi Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yudi Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ruizhi Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Shuang Chen
- Center of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yingying Lv
- Department of Pediatrics, The First Affliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Department of Pediatrics, The Third Affliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yifeng Chen
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yuqing Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Peng Luo
- The Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Anning Zuo
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yuhao Ba
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Shutong Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Interventional Institute of Zhengzhou University, Zhengzhou, Henan, 450052, China; Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China.
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Interventional Institute of Zhengzhou University, Zhengzhou, Henan, 450052, China; Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China; Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
121
|
Knudsen-Clark AM, Mwangi D, Cazarin J, Morris K, Baker C, Hablitz LM, McCall MN, Kim M, Altman BJ. Circadian rhythms of macrophages are altered by the acidic tumor microenvironment. EMBO Rep 2024; 25:5080-5112. [PMID: 39415049 PMCID: PMC11549407 DOI: 10.1038/s44319-024-00288-2] [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: 08/01/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/18/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are prime therapeutic targets due to their pro-tumorigenic functions, but varying efficacy of macrophage-targeting therapies highlights our incomplete understanding of how macrophages are regulated within the tumor microenvironment (TME). The circadian clock is a key regulator of macrophage function, but how circadian rhythms of macrophages are influenced by the TME remains unknown. Here, we show that conditions associated with the TME such as polarizing stimuli, acidic pH, and lactate can alter circadian rhythms in macrophages. While cyclic AMP (cAMP) has been reported to play a role in macrophage response to acidic pH, our results indicate pH-driven changes in circadian rhythms are not mediated solely by cAMP signaling. Remarkably, circadian disorder of TAMs was revealed by clock correlation distance analysis. Our data suggest that heterogeneity in circadian rhythms within the TAM population level may underlie this circadian disorder. Finally, we report that circadian regulation of macrophages suppresses tumor growth in a murine model of pancreatic cancer. Our work demonstrates a novel mechanism by which the TME influences macrophage biology through modulation of circadian rhythms.
Collapse
Affiliation(s)
- Amelia M Knudsen-Clark
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Daniel Mwangi
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
| | - Juliana Cazarin
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
| | - Kristina Morris
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
| | - Cameron Baker
- Genomics Research Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Lauren M Hablitz
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Matthew N McCall
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Minsoo Kim
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Brian J Altman
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA.
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
122
|
Sweatt AJ, Griffiths CD, Groves SM, Paudel BB, Wang L, Kashatus DF, Janes KA. Proteome-wide copy-number estimation from transcriptomics. Mol Syst Biol 2024; 20:1230-1256. [PMID: 39333715 PMCID: PMC11535397 DOI: 10.1038/s44320-024-00064-3] [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: 08/02/2023] [Revised: 08/22/2024] [Accepted: 09/02/2024] [Indexed: 09/29/2024] Open
Abstract
Protein copy numbers constrain systems-level properties of regulatory networks, but proportional proteomic data remain scarce compared to RNA-seq. We related mRNA to protein statistically using best-available data from quantitative proteomics and transcriptomics for 4366 genes in 369 cell lines. The approach starts with a protein's median copy number and hierarchically appends mRNA-protein and mRNA-mRNA dependencies to define an optimal gene-specific model linking mRNAs to protein. For dozens of cell lines and primary samples, these protein inferences from mRNA outmatch stringent null models, a count-based protein-abundance repository, empirical mRNA-to-protein ratios, and a proteogenomic DREAM challenge winner. The optimal mRNA-to-protein relationships capture biological processes along with hundreds of known protein-protein complexes, suggesting mechanistic relationships. We use the method to identify a viral-receptor abundance threshold for coxsackievirus B3 susceptibility from 1489 systems-biology infection models parameterized by protein inference. When applied to 796 RNA-seq profiles of breast cancer, inferred copy-number estimates collectively re-classify 26-29% of luminal tumors. By adopting a gene-centered perspective of mRNA-protein covariation across different biological contexts, we achieve accuracies comparable to the technical reproducibility of contemporary proteomics.
Collapse
Affiliation(s)
- Andrew J Sweatt
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22908, USA
| | - Cameron D Griffiths
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22908, USA
| | - Sarah M Groves
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22908, USA
| | - B Bishal Paudel
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22908, USA
| | - Lixin Wang
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22908, USA
| | - David F Kashatus
- Department of Microbiology, Immunology & Cancer Biology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Kevin A Janes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22908, USA.
- Department of Biochemistry & Molecular Genetics, University of Virginia, Charlottesville, VA, 22908, USA.
| |
Collapse
|
123
|
Huyen VT, Echizen K, Yamagishi R, Kumagai M, Nonaka Y, Kodama T, Ando T, Yano M, Takada N, Takasugi M, Kamachi F, Ohtani N. Regular exercise suppresses steatosis-associated liver cancer development by degrading E2F1 and c-Myc via circadian gene upregulation. Genes Cells 2024; 29:1012-1025. [PMID: 39357875 DOI: 10.1111/gtc.13161] [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: 07/13/2024] [Revised: 08/20/2024] [Accepted: 08/30/2024] [Indexed: 10/04/2024]
Abstract
Regular exercise is believed to suppress cancer progression. However, the precise molecular mechanisms by which exercise prevents cancer development remain unclear. In this study, using a steatosis-associated liver cancer mouse model, we found that regular exercise at a speed of 18 m/min for 20 min daily suppressed liver cancer development. To explore the underlying mechanisms, we examined the gene expression profiles in the livers of the exercise and non-exercise groups. The expressions of circadian genes, such as Per1 and Cry2, were upregulated in the exercise group. As circadian rhythm disruption is known to cause various diseases, including cancer, improving circadian rhythm through exercise could contribute to cancer prevention. We further found that the expression of a series of E2F1 and c-Myc target genes that directly affect the proliferation of cancer cells was downregulated in the exercise group. However, the expression of E2F1 and c-Myc was transcriptionally unchanged but degraded at the post-translational level by exercise. Cry2, which is regulated by the Skp1-Cul1-FBXL3 (SCFFBXL3) ubiquitin ligase complex by binding to FBXL3, can form a complex with E2F1 and c-Myc, which we think is the mechanism to degrade them. Our study revealed a previously unknown mechanism by which exercise prevents cancer development.
Collapse
Affiliation(s)
- Vu Thuong Huyen
- Department of Pathophysiology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
- Department of Pathophysiology, Graduate School of Medicine, Osaka City University, Osaka, Japan
- Department of Pediatrics, Hanoi Medical University, Hanoi, Vietnam
| | - Kanae Echizen
- Department of Pathophysiology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Ryota Yamagishi
- Department of Pathophysiology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Miho Kumagai
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Chiba, Japan
| | - Yoshiki Nonaka
- Department of Pathophysiology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
- Department of Pathophysiology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Takahiro Kodama
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Tatsuya Ando
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Chiba, Japan
| | - Megumu Yano
- Department of Pathophysiology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Naoki Takada
- Department of Pathophysiology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
- Department of Orthopedic Surgery, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan
| | - Masaki Takasugi
- Department of Pathophysiology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Fumitaka Kamachi
- Department of Pathophysiology, Graduate School of Medicine, Osaka City University, Osaka, Japan
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Chiba, Japan
| | - Naoko Ohtani
- Department of Pathophysiology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
- Department of Pathophysiology, Graduate School of Medicine, Osaka City University, Osaka, Japan
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Chiba, Japan
| |
Collapse
|
124
|
Ding H, Meng L, Zhang Y, Bryant AJ, Xing C, Esser KA, Chen L, Huo Z. A Bayesian Framework for Genome-wide Circadian Rhythmicity Biomarker Detection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.28.620703. [PMID: 39554018 PMCID: PMC11565714 DOI: 10.1101/2024.10.28.620703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Circadian rhythms are endogenous ∼24-hour cycles that significantly influence physiological and behavioral processes. These rhythms are governed by a transcriptional-translational feedback loop of core circadian genes and are essential for maintaining overall health. The study of circadian rhythms has expanded into various omics datasets, necessitating accurate analytical methodology for circadian biomarker detection. Here, we introduce a novel Bayesian framework for the genome-wide detection of circadian rhythms that is capable of incorporating prior biological knowledge and adjusting for multiple testing issue via a false discovery rate approach. Our framework leverages a Bayesian hierarchical model and employs a reverse jump Markov chain Monte Carlo (rjMCMC) technique for model selection. Through extensive simulations, our method, BayesCircRhy, demonstrated superior false discovery rate control over competing methods, robustness against heavier-tailed error distributions, and better performance compared to existing approaches. The method's efficacy was further validated in two RNA-Sequencing data, including a human resitrcted feeding data and a mouse aging data, where it successfully identified known and novel circadian genes. R package "BayesianCircadian" for the method is publicly available on GitHub https://github.com/jxncdhc/BayesianCircadian .
Collapse
|
125
|
Zee A, Deng DZ, DiTacchio L, Vollmers C. The Circadian Isoform Landscape of Mouse Livers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.16.618716. [PMID: 39464136 PMCID: PMC11507890 DOI: 10.1101/2024.10.16.618716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
The mammalian circadian clock is an autoregulatory feedback process that is responsible for homeostasis in mouse livers. These circadian processes are well understood at the gene-level, however, not well understood at the isoform-level. To investigate circadian oscillations at the isoform-level, we used the nanopore-based R2C2 method to create over 78 million highly-accurate, full-length cDNA reads for 12 RNA samples extracted from mouse livers collected at 2 hour intervals. To generate a circadian mouse liver isoform-level transcriptome, we processed these reads using the Mandalorion tool which identified and quantified 58,612 isoforms, 1806 of which showed circadian oscillations. We performed detailed analysis on the circadian oscillation of these isoforms, their coding sequences, and transcription start sites and compiled easy-to-access resources for other researchers. This study and its results add a new layer of detail to the quantitative analysis of transcriptomes.
Collapse
Affiliation(s)
- Alexander Zee
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, California 95064, USA
| | - Dori Z.Q. Deng
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Cruz, Santa Cruz, California 95064, USA
| | - Luciano DiTacchio
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66101, USA
- Current affiliation: Synexin LLC, Carlsbad, CA 92010
| | - Christopher Vollmers
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, California 95064, USA
| |
Collapse
|
126
|
Cederroth CR, Dyhrfjeld-Johnsen J, Canlon B. Pharmacological Approaches to Hearing Loss. Pharmacol Rev 2024; 76:1063-1088. [PMID: 39164117 PMCID: PMC11549935 DOI: 10.1124/pharmrev.124.001195] [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: 02/14/2024] [Revised: 07/08/2024] [Accepted: 07/16/2024] [Indexed: 08/22/2024] Open
Abstract
Hearing disorders pose significant challenges to individuals experiencing them and their overall quality of life, emphasizing the critical need for advanced pharmacological approaches to address these conditions. Current treatment options often focus on amplification devices, cochlear implants, or other rehabilitative therapies, leaving a substantial gap regarding effective pharmacological interventions. Advancements in our understanding of the molecular and cellular mechanisms involved in hearing disorders induced by noise, aging, and ototoxicity have opened new avenues for drug development, some of which have led to numerous clinical trials, with promising results. The development of optimal drug delivery solutions in animals and humans can also enhance the targeted delivery of medications to the ear. Moreover, large genome studies contributing to a genetic understanding of hearing loss in humans combined with advanced molecular technologies in animal studies have shown a great potential to increase our understanding of the etiologies of hearing loss. The auditory system exhibits circadian rhythms and temporal variations in its physiology, its vulnerability to auditory insults, and its responsiveness to drug treatments. The cochlear clock rhythms are under the control of the glucocorticoid system, and preclinical evidence suggests that the risk/benefit profile of hearing disorder treatments using chronopharmacological approaches would be beneficial. If translatable to the bedside, such approaches may improve the outcome of clinical trials. Ongoing research into the molecular and genetic basis of auditory disorders, coupled with advancements in drug formulation and delivery as well as optimized timing of drug administration, holds great promise of more effective treatments. SIGNIFICANCE STATEMENT: Hearing disorders pose significant challenges to individuals and their overall quality of life, emphasizing the critical need for advanced pharmacological approaches to address these conditions. Ongoing research into the molecular and genetic basis of auditory disorders, coupled with advancements in drug delivery procedures and optimized timing of drug administration, holds the promise of more effective treatments.
Collapse
Affiliation(s)
- Christopher R Cederroth
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden (C.R.C., B.C.); Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany (C.R.C.); and Acousia Therapeutics GmbH, Tübingen, Germany (J.D.-J.)
| | - Jonas Dyhrfjeld-Johnsen
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden (C.R.C., B.C.); Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany (C.R.C.); and Acousia Therapeutics GmbH, Tübingen, Germany (J.D.-J.)
| | - Barbara Canlon
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden (C.R.C., B.C.); Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany (C.R.C.); and Acousia Therapeutics GmbH, Tübingen, Germany (J.D.-J.)
| |
Collapse
|
127
|
Uriu K, Hernandez-Sanchez JP, Kojima S. Impacts of the feedback loop between sense-antisense RNAs in regulating circadian rhythms. NPJ Syst Biol Appl 2024; 10:119. [PMID: 39414861 PMCID: PMC11484753 DOI: 10.1038/s41540-024-00451-4] [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: 06/04/2024] [Accepted: 10/03/2024] [Indexed: 10/18/2024] Open
Abstract
Antisense transcripts are a unique group of non-coding RNAs and play regulatory roles in a variety of biological processes, including circadian rhythms. Per2AS is an antisense transcript to the sense core clock gene Period2 (Per2) in mouse and its expression is rhythmic and antiphasic to Per2. To understand the impact of Per2AS-Per2 interaction, we developed a new mathematical model that mechanistically described the mutually repressive relationship between Per2 and Per2AS. This mutual repression can regulate both amplitude and period of circadian oscillation by affecting a negative feedback regulation of Per2. Simulations from this model also fit with experimental observations that could not be fully explained by our previous model. Our revised model can not only serve as a foundation to build more detailed models to better understand the impact of Per2AS-Per2 interaction in the future, but also be used to analyze other sense-antisense RNA pairs that mutually repress each other.
Collapse
Affiliation(s)
- Koichiro Uriu
- School of Life Science and Technology, Institute of Science Tokyo, Meguro, Tokyo, Japan.
- Graduate School of Natural Science and Technology, Kanazawa University, Kanazawa, Ishikawa, Japan.
| | - Juan P Hernandez-Sanchez
- Department of Biological Sciences, Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, USA
| | - Shihoko Kojima
- Department of Biological Sciences, Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, USA.
- Center for the Mathematics of Biosystems, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
128
|
Latha Laxmi IP, Tamizhselvi R. Epigenetic events influencing the biological clock: Panacea for neurodegeneration. Heliyon 2024; 10:e38836. [PMID: 39430507 PMCID: PMC11489350 DOI: 10.1016/j.heliyon.2024.e38836] [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: 01/24/2024] [Revised: 09/28/2024] [Accepted: 10/01/2024] [Indexed: 10/22/2024] Open
Abstract
The human biological clock is the 24-h internal molecular network of circadian genes in synchronization with other cells in response to external stimuli. The rhythmicity of the clock genes is maintained by positive and negative transcriptional feedback loops coordinating the 24-h oscillation in different tissues. The superchiasmatic nucleus, the central pacemaker of the biological clock diminishes with aging causing alterations in the clock rhythmicity leading to the onset of neurodegenerative diseases mainly Alzheimer's disease, Parkinson's disease, and Huntington's disease. Studies have shown that brain and muscle Arnt -like 1 (Bmal1) and Circadian Locomotor Output Cycles Kaput (Clock) gene expression is altered in the onset of neurodegeneration. One of the major symptoms of neurodegeneration is changes in the sleep/wake cycle. Moreover, variations in circadian clock oscillations can happen due to lifestyle changes, addiction to alcohol, cocaine, drugs, smoking, food habits and most importantly eating and sleep/awake cycle patterns which can significantly impact the expression of circadian genes. Recent studies have focused on the molecular function of clock genes affected due to environmental cues. Epigenetic modifications are influenced by the external environmental factors. This review aims to focus on the principal mechanism of epigenetics influencing circadian rhythm disruption leading to neurodegeneration and as well as targeting the epigenetic modulators could be a novel therapeutic approach to combat neurodegenerative disorders.
Collapse
Affiliation(s)
| | - Ramasamy Tamizhselvi
- School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| |
Collapse
|
129
|
Su Z, Hu Q, Li X, Wang Z, Xie Y. The Influence of Circadian Rhythms on DNA Damage Repair in Skin Photoaging. Int J Mol Sci 2024; 25:10926. [PMID: 39456709 PMCID: PMC11507642 DOI: 10.3390/ijms252010926] [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: 08/15/2024] [Revised: 09/29/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Circadian rhythms, the internal timekeeping systems governing physiological processes, significantly influence skin health, particularly in response to ultraviolet radiation (UVR). Disruptions in circadian rhythms can exacerbate UVR-induced skin damage and increase the risk of skin aging and cancer. This review explores how circadian rhythms affect various aspects of skin physiology and pathology, with a special focus on DNA repair. Circadian regulation ensures optimal DNA repair following UVR-induced damage, reducing mutation accumulation, and enhancing genomic stability. The circadian control over cell proliferation and apoptosis further contributes to skin regeneration and response to UVR. Oxidative stress management is another critical area where circadian rhythms exert influence. Key circadian genes like brain and muscle ARNT-like 1 (BMAL1) and circadian locomotor output cycles kaput (CLOCK) modulate the activity of antioxidant enzymes and signaling pathways to protect cells from oxidative stress. Circadian rhythms also affect inflammatory and immune responses by modulating the inflammatory response and the activity of Langerhans cells and other immune cells in the skin. In summary, circadian rhythms form a complex defense network that manages UVR-induced damage through the precise regulation of DNA damage repair, cell proliferation, apoptosis, inflammatory response, oxidative stress, and hormonal signaling. Understanding these mechanisms provides insights into developing targeted skin protection and improving skin cancer prevention.
Collapse
Affiliation(s)
- Zhi Su
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha 410081, China
| | - Qianhua Hu
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha 410081, China
| | - Xiang Li
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha 410081, China
| | - Zirun Wang
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha 410081, China
| | - Ying Xie
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha 410081, China
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha 410081, China
| |
Collapse
|
130
|
Masuda S, Kurabayashi N, Nunokawa R, Otobe Y, Kozuka-Hata H, Oyama M, Shibata Y, Inoue JI, Koebis M, Aiba A, Yoshitane H, Fukada Y. TRAF7 determines circadian period through ubiquitination and degradation of DBP. Commun Biol 2024; 7:1280. [PMID: 39379486 PMCID: PMC11461874 DOI: 10.1038/s42003-024-07002-x] [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: 11/09/2023] [Accepted: 10/01/2024] [Indexed: 10/10/2024] Open
Abstract
D-site binding protein, DBP, is a clock-controlled transcription factor and drives daily rhythms of physiological processes through the regulation of an array of genes harboring a DNA binding motif, D-box. DBP protein levels show a circadian oscillation with an extremely robust peak/trough ratio, but it is elusive how the temporal pattern is regulated by post-translational regulation. In this study, we show that DBP protein levels are down-regulated by the ubiquitin-proteasome pathway. Analysis using 19 dominant-negative forms of E2 enzymes have revealed that UBE2G1 and UBE2T mediate the degradation of DBP. A proteomic analysis of DBP-interacting proteins and database screening have identified Tumor necrosis factor Receptor-Associated Factor 7 (TRAF7), a RING-type E3 ligase, that forms a complex with UBE2G1 and/or UBE2T. Ubiquitination analysis have revealed that TRAF7 enhances K48-linked polyubiquitination of DBP in cultured cells. Overexpression of TRAF7 down-regulates DBP protein level, while knockdown of TRAF7 up-regulates DBP in cultured cells. Knockout of TRAF7 in NIH3T3 cells have revealed that TRAF7 mediates the time-of-the-day-dependent regulation of DBP levels. Furthermore, TRAF7 has a period-shortening effect on the cellular clock. Together, TRAF7 plays an important role in circadian clock oscillation through destabilization of DBP.
Collapse
Affiliation(s)
- Shusaku Masuda
- Department of Biological Sciences, School of Science, The University of Tokyo, Tokyo, Japan
| | - Nobuhiro Kurabayashi
- Circadiain Clock Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Rina Nunokawa
- Department of Biological Sciences, School of Science, The University of Tokyo, Tokyo, Japan
| | - Yuta Otobe
- Department of Biological Sciences, School of Science, The University of Tokyo, Tokyo, Japan
- Circadiain Clock Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Hiroko Kozuka-Hata
- Medical Proteomics Laboratory, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masaaki Oyama
- Medical Proteomics Laboratory, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yuri Shibata
- Division of Cellular and Molecular Biology, Department of Cancer Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Jun-Ichiro Inoue
- Medical Proteomics Laboratory, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Division of Cellular and Molecular Biology, Department of Cancer Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Michinori Koebis
- Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Atsu Aiba
- Department of Biological Sciences, School of Science, The University of Tokyo, Tokyo, Japan
- Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hikari Yoshitane
- Department of Biological Sciences, School of Science, The University of Tokyo, Tokyo, Japan.
- Circadiain Clock Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.
| | - Yoshitaka Fukada
- Department of Biological Sciences, School of Science, The University of Tokyo, Tokyo, Japan.
- Circadiain Clock Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.
- Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
131
|
Xu B, Ma D, Abruzzi K, Braun R. Detecting Rhythmic Gene Expression in Single-cell Transcriptomics. J Biol Rhythms 2024:7487304241273182. [PMID: 39377613 DOI: 10.1177/07487304241273182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
An autonomous, environmentally synchronizable circadian rhythm is a ubiquitous feature of life on Earth. In multicellular organisms, this rhythm is generated by a transcription-translation feedback loop present in nearly every cell that drives daily expression of thousands of genes in a tissue-dependent manner. Identifying the genes that are under circadian control can elucidate the mechanisms by which physiological processes are coordinated in multicellular organisms. Today, transcriptomic profiling at the single-cell level provides an unprecedented opportunity to understand the function of cell-level clocks. However, while many cycling detection algorithms have been developed to identify genes under circadian control in bulk transcriptomic data, it is not known how best to adapt these algorithms to single-cell RNA seq data. Here, we benchmark commonly used circadian detection methods on their reliability and efficiency when applied to single-cell RNA seq data. Our results provide guidance on adapting existing cycling detection methods to the single-cell domain and elucidate opportunities for more robust and efficient rhythm detection in single-cell data. We also propose a subsampling procedure combined with harmonic regression as an efficient strategy to detect circadian genes in the single-cell setting.
Collapse
Affiliation(s)
- Bingxian Xu
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois, USA
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston, Illinois, USA
| | - Dingbang Ma
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- Shanghai Key Laboratory of Aging Studies, Shanghai, China
| | - Katharine Abruzzi
- HHMI, Brandeis University, Waltham, Massachusetts, USA
- Department of Biology, Brandeis University, Waltham, Massachusetts, USA
| | - Rosemary Braun
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois, USA
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston, Illinois, USA
- Department of Engineering Sciences and Applied Mathematics, Northwestern University, Evanston, Illinois, USA
- Department of Physics and Astronomy, Northwestern University, Evanston, Illinois, USA
- Northwestern Institute on Complex Systems, Northwestern University, Evanston, Illinois, USA
| |
Collapse
|
132
|
Levy JL, Mirek ET, Rodriguez EM, Tolentino MJ, Zalma BA, Roepke TA, Wek RC, Cao R, Anthony TG. GCN2 drives diurnal patterns in the hepatic integrated stress response and maintains circadian rhythms in whole body metabolism during amino acid insufficiency. Am J Physiol Endocrinol Metab 2024; 327:E563-E576. [PMID: 39196798 PMCID: PMC11482268 DOI: 10.1152/ajpendo.00129.2024] [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: 03/29/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 08/30/2024]
Abstract
Disruptions in circadian rhythms are associated with an increased risk of developing metabolic diseases. General control nonderepressible 2 (GCN2), a primary sensor of amino acid insufficiency and activator of the integrated stress response (ISR), has emerged as a conserved regulator of the circadian clock in multiple organisms. The objective of this study was to examine diurnal patterns in hepatic ISR activation in the liver and whole body rhythms in metabolism. We hypothesized that GCN2 activation cues hepatic ISR signaling over a natural 24-h feeding-fasting cycle. To address our objective, wild-type (WT) and whole body Gcn2 knockout (GCN2 KO) mice were housed in metabolic cages and provided free access to either a control or leucine-devoid diet (LeuD) for 8 days in total darkness. On the last day, blood and livers were collected at CT3 (CT = circadian time) and CT15. In livers of WT mice, GCN2 phosphorylation followed a diurnal pattern that was guided by intracellular branched-chain amino acid concentrations (r2 = 0.93). Feeding LeuD to WT mice increased hepatic ISR activation at CT15 only. Diurnal oscillations in hepatic ISR signaling, the hepatic transcriptome including lipid metabolic genes, and triglyceride concentrations were substantially reduced or absent in GCN2 KO mice. Furthermore, mice lacking GCN2 were unable to maintain circadian rhythms in whole body energy expenditure, respiratory exchange ratio, and physical activity when fed LeuD. In conclusion, GCN2 activation functions to maintain diurnal ISR activation in the liver and has a vital role in the mechanisms by which nutrient stress affects whole body metabolism.NEW & NOTEWORTHY This work reveals that the eIF2 kinase GCN2 functions to support diurnal patterns in the hepatic integrated stress response during natural feeding and is necessary to maintain circadian rhythms in energy expenditure, respiratory exchange ratio, and physical activity during amino acid stress.
Collapse
Affiliation(s)
- Jordan L Levy
- Department of Nutritional Sciences, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, New Jersey, United States
| | - Emily T Mirek
- Department of Nutritional Sciences, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, New Jersey, United States
| | - Esther M Rodriguez
- Department of Nutritional Sciences, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, New Jersey, United States
| | - Maria J Tolentino
- Department of Nutritional Sciences, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, New Jersey, United States
| | - Brian A Zalma
- Department of Nutritional Sciences, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, New Jersey, United States
| | - Troy A Roepke
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, New Jersey, United States
| | - Ronald C Wek
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Ruifeng Cao
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, United States
- Department of Neurology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, United States
| | - Tracy G Anthony
- Department of Nutritional Sciences, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, New Jersey, United States
- New Jersey Institute for Food, Nutrition and Health, Rutgers University, New Brunswick, New Jersey, United States
| |
Collapse
|
133
|
Cox OH, Gianonni-Guzmán MA, Cartailler JP, Cottam MA, McMahon DG. Transcriptomic Plasticity of the Circadian Clock in Response to Photoperiod: A Study in Male Melatonin-Competent Mice. J Biol Rhythms 2024; 39:423-439. [PMID: 39096022 PMCID: PMC11425976 DOI: 10.1177/07487304241265439] [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: 08/04/2024]
Abstract
Seasonal daylength, or circadian photoperiod, is a pervasive environmental signal that profoundly influences physiology and behavior. In mammals, the central circadian clock resides in the suprachiasmatic nuclei (SCN) of the hypothalamus where it receives retinal input and synchronizes, or entrains, organismal physiology and behavior to the prevailing light cycle. The process of entrainment induces sustained plasticity in the SCN, but the molecular mechanisms underlying SCN plasticity are incompletely understood. Entrainment to different photoperiods persistently alters the timing, waveform, period, and light resetting properties of the SCN clock and its driven rhythms. To elucidate novel candidate genes for molecular mechanisms of photoperiod plasticity, we performed RNA sequencing on whole SCN dissected from mice raised in long (light:dark [LD] 16:8) and short (LD 8:16) photoperiods. Fewer rhythmic genes were detected in mice subjected to long photoperiod, and in general, the timing of gene expression rhythms was advanced 4-6 h. However, a few genes showed significant delays, including Gem. There were significant changes in the expression of the clock-associated gene Timeless and in SCN genes related to light responses, neuropeptides, gamma aminobutyric acid (GABA), ion channels, and serotonin. Particularly striking were differences in the expression of the neuropeptide signaling genes Prokr2 and Cck, as well as convergent regulation of the expression of 3 SCN light response genes, Dusp4, Rasd1, and Gem. Transcriptional modulation of Dusp4 and Rasd1 and phase regulation of Gem are compelling candidate molecular mechanisms for plasticity in the SCN light response through their modulation of the critical NMDAR-MAPK/ERK-CREB/CRE light signaling pathway in SCN neurons. Modulation of Prokr2 and Cck may critically support SCN neural network reconfiguration during photoperiodic entrainment. Our findings identify the SCN light response and neuropeptide signaling gene sets as rich substrates for elucidating novel mechanisms of photoperiod plasticity. Data are also available at http://circadianphotoperiodseq.com/, where users can view the expression and rhythmic properties of genes across these photoperiod conditions.
Collapse
Affiliation(s)
- Olivia H. Cox
- Neuroscience Graduate Program, Vanderbilt University, Nashville, Tennessee
| | | | - Jean-Philippe Cartailler
- The Vanderbilt Creative Data Solutions Shared Resource, Vanderbilt University, Nashville, Tennessee
| | - Matthew A. Cottam
- The Vanderbilt Creative Data Solutions Shared Resource, Vanderbilt University, Nashville, Tennessee
| | - Douglas G. McMahon
- Neuroscience Graduate Program, Vanderbilt University, Nashville, Tennessee
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
134
|
Alkan I, Durkut B, Ucak M, Bozkurt M, Canat HL, Celik-Ozenci C. Uncovering the Penile Clock: Expression of Molecular Clock Proteins in Human Penile Cavernous Tissue. World J Mens Health 2024; 42:797-809. [PMID: 38311375 PMCID: PMC11439802 DOI: 10.5534/wjmh.230136] [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: 05/18/2023] [Revised: 10/10/2023] [Accepted: 10/23/2023] [Indexed: 02/10/2024] Open
Abstract
PURPOSE To evaluate the expression of core molecular clock genes/proteins in penile cavernous tissue from healthy male subjects and to determine whether their expression has circadian variation. MATERIALS AND METHODS Corpus cavernosum biopsy samples were obtained from 10 healthy males with penile deviation or fracture who underwent surgical intervention during the day and night. The daytime group (n=5) underwent corpus cavernosum tissue sampling during zeitgeber time (ZT) 8-12, while the nighttime group (n=5) underwent sampling during ZT 20-24. The expression and localization of BMAL1, CLOCK, PER1, PER2, PER3, CRY1, and CRY2 proteins were analyzed using immunohistochemistry and quantified using H-score analysis. RT-qPCR analysis was performed to assess the expression of core molecular clock genes in the corpus cavernosum tissue of 5 additional daytime patients. RESULTS The expression of core molecular clock proteins was detected in vascular endothelial cells (VECs) and smooth muscle cells (SMCs) in corpus cavernosum during daytime and nighttime. BMAL1 exhibited the most significant nuclear expression during daytime in both cell types, whereas its expression decreased significantly at night. In VECs, a significant decrease in the nuclear expression of CRY1 was observed at night. In SMCs, a significant decrease in the cytoplasmic expression of PER3 was observed at night. The expression patterns of the core molecular clock genes were ascertained through a RT-qPCR analysis. CONCLUSIONS Our research provides compelling evidence that core molecular clock genes are distinctly expressed in penile tissue in humans. Furthermore, we observed the expression of molecular clock proteins within the VECs and SMCs of the corpus cavernosum, with BMAL1 being the most prominently expressed. The discovery of core molecular clock genes in penile tissue, as well as proteins within the SMCs and VECs of the corpus cavernosum, introduces the potential significance of the molecular clock mechanism in the physiology of penile erection.
Collapse
Affiliation(s)
- Ilter Alkan
- Department of Urology, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Begum Durkut
- Programme of Reproductive Medicine, Graduate School of Health Sciences, Koç University, Istanbul, Turkey
| | - Melike Ucak
- Programme of Reproductive Medicine, Graduate School of Health Sciences, Koç University, Istanbul, Turkey
| | - Muammer Bozkurt
- Department of Urology, Cam and Sakura City Hospital, Istanbul, Turkey
| | - Halil Lutfi Canat
- Department of Urology, Cam and Sakura City Hospital, Istanbul, Turkey
| | - Ciler Celik-Ozenci
- Department of Histology and Embryology, School of Medicine, Koç University, Istanbul, Turkey
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey.
| |
Collapse
|
135
|
Kisamore C, Kisamore C, Walker W. Circadian Rhythm Disruption in Cancer Survivors: From Oncogenesis to Quality of Life. Cancer Med 2024; 13:e70353. [PMID: 39463009 PMCID: PMC11513439 DOI: 10.1002/cam4.70353] [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: 06/26/2024] [Revised: 10/04/2024] [Accepted: 10/09/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Circadian rhythms are approximately 24-hour cycles in physiological and behavioral processes. They are entrained to the external solar day via blue wavelength light. Disruptions in these intrinsic rhythms can lead to circadian dysfunction, which has several negative implications on human health, including cancer development and progression. AIMS Here we review the molecular mechanisms of circadian disruption and their impact on tumor development and progression, discuss the interplay between circadian dysfunction and cancer in basic scientific studies and clinical data, and propose the potential clinical implications of these data that may be used to improve patient outcomes and reduce cost of treatment. MATERIALS & METHODS Using scientific literature databases, relevant studies were analyzed to draw overarching conclusions of the relationship between circadian rhythm dysruption and cancer. CONCLUSIONS Circadian disruption can be mediated by a number of environmental factors such as exposure to light at night, shift work, jetlag, and social jetlag which drive oncogenesis. Tumor growth and progression, as well as treatment, can lead to long-term alterations in circadian rhythms that negatively affect quality of life in cancer survivors.
Collapse
Affiliation(s)
- Claire O. Kisamore
- Department of Neuroscience, Rockefeller Neuroscience InstituteWest Virginia UniversityMorgantownWest VirginiaUSA
| | - Caleb A. Kisamore
- Department of Neuroscience, Rockefeller Neuroscience InstituteWest Virginia UniversityMorgantownWest VirginiaUSA
| | - William H. Walker
- Department of Neuroscience, Rockefeller Neuroscience InstituteWest Virginia UniversityMorgantownWest VirginiaUSA
- West Virginia University Cancer InstituteMorgantownWest VirginiaUSA
| |
Collapse
|
136
|
Ma C, Li H, Shen B, Zheng H, Chen Y, Chen L, Yang G. Differential Effects of Light and Dark Phase Modifications on Jet Lag Adaptability in Mice. J Pineal Res 2024; 76:e13010. [PMID: 40008645 DOI: 10.1111/jpi.13010] [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: 08/14/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 02/27/2025]
Abstract
In chronobiology, shifting light/dark cycles is a common method to disrupt circadian rhythms. While the direction and magnitude of a phase shift (e.g., +6 denoting a 6-h advanced shift) dictate the temporal change before and after the shift, little attention has been paid to the duration and relative proportion of daytime and nighttime during the shift, leading to a critical, unexamined variable in circadian research. In this study, we introduce the concepts of "L-shift" (longer light phase on the shift day) and "D-shift" (longer dark phase), and investigate how these variations impact the adaptability of mice to jet lag. By examining multiple phase shifts (12L vs. 12D, +6L vs. +6D, -6L vs. -6D), we demonstrate that L-shifts not only facilitate faster adaptation but also significantly reduce the severity of sepsis in a jet lag-sensitive lipopolysaccharide-induced sepsis model. Further investigations with additional phase shifts at 1-h intervals (+8 to +11) reinforced the enhanced fitness of mice under L-shifts. Mechanistically, L-shifts were found to increase sleep duration, thereby improving circadian entrainment, with sleep deprivation nullifying the adaptability differences between lighting protocols. These findings underscore a previously unrecognized factor in circadian biology and suggest that optimizing lighting protocols could profoundly improve adaptability to circadian disruptions. This research opens new avenues for enhancing therapeutic strategies and refining experimental designs in the field of chronobiology.
Collapse
Affiliation(s)
- Changxiao Ma
- Health Science Center, East China Normal University, Shanghai, China
- School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Haonan Li
- Health Science Center, East China Normal University, Shanghai, China
- School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Bingyi Shen
- School of Bioengineering, Dalian University of Technology, Dalian, China
- School of Clinical Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Huiwen Zheng
- School of Bioengineering, Dalian University of Technology, Dalian, China
- School of Clinical Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Yunfei Chen
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lihong Chen
- Health Science Center, East China Normal University, Shanghai, China
| | - Guangrui Yang
- School of Clinical Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| |
Collapse
|
137
|
Chen K, Ashtiani KC, Monfared RV, Baldi P, Alachkar A. Circadian cilia transcriptome in mouse brain across physiological and pathological states. Mol Brain 2024; 17:67. [PMID: 39304885 PMCID: PMC11414107 DOI: 10.1186/s13041-024-01143-0] [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: 08/02/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024] Open
Abstract
Primary cilia are dynamic sensory organelles that continuously undergo structural modifications in response to environmental and cellular signals, many of which exhibit rhythmic patterns. Building on our previous findings of rhythmic cilia-related gene expression in diurnal primates (baboon), this study extends the investigation to the nocturnal mouse brain to identify circadian patterns of cilia gene expression across brain regions. We used computational techniques and transcriptomic data from four publicly available databases, to examine the circadian expression of cilia-associated genes within six brain areas: brainstem, cerebellum, hippocampus, hypothalamus, striatum, and suprachiasmatic nucleus. Our analysis reveals that a substantial proportion of cilia transcripts exhibit circadian rhythmicity across the examined regions, with notable overrepresentation in the striatum, hippocampus, and cerebellum. We also demonstrate region-specific variations in the abundance and timing of circadian cilia genes' peaks, indicating an adaptation to the distinct physiological roles of each brain region. Additionally, we show that the rhythmic patterns of cilia transcripts are shifted under various physiological and pathological conditions, including modulation of the dopamine system, high-fat diet, and epileptic conditions, indicating the adaptable nature of cilia transcripts' oscillation. While limited to a few mouse brain regions, our study provides initial insights into the distinct circadian patterns of cilia transcripts and highlights the need for future research to expand the mapping across wider brain areas to fully understand the role of cilia's spatiotemporal dynamics in brain functions.
Collapse
Affiliation(s)
- Kiki Chen
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, 356A Med Surge II, Irvine, CA, 92697-4625, USA
| | - Kousha Changizi Ashtiani
- Departments of Computer Science, School of Information and Computer Sciences, University of California, Irvine, CA, 92697-4625, USA
| | - Roudabeh Vakil Monfared
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, 356A Med Surge II, Irvine, CA, 92697-4625, USA
| | - Pierre Baldi
- Departments of Computer Science, School of Information and Computer Sciences, University of California, Irvine, CA, 92697-4625, USA.
- Institute for Genomics and Bioinformatics, School of Information and Computer Sciences, University of California, Irvine, CA, 92697, USA.
| | - Amal Alachkar
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, 356A Med Surge II, Irvine, CA, 92697-4625, USA.
- Institute for Genomics and Bioinformatics, School of Information and Computer Sciences, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
138
|
Yang B, Wei W, Fang J, Xue Y, Wei J. Diabetic Neuropathic Pain and Circadian Rhythm: A Future Direction Worthy of Study. J Pain Res 2024; 17:3005-3020. [PMID: 39308994 PMCID: PMC11414757 DOI: 10.2147/jpr.s467249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 07/11/2024] [Indexed: 09/25/2024] Open
Abstract
More than half of people with diabetes experience neuropathic pain. Previous research has shown that diabetes patients' neuropathic pain exhibits a circadian cycle, which is characterized by increased pain sensitivity at night. Additional clinical research has revealed that the standard opioid drugs are ineffective at relieving pain and do not change the circadian rhythm. This article describes diabetic neuropathic pain and circadian rhythms separately, with a comprehensive focus on circadian rhythms. It is hoped that this characteristic of diabetic neuropathic pain can be utilized in the future to obtain more effective treatments for it.
Collapse
Affiliation(s)
- Baozhong Yang
- Department of Anaesthesiology, Taiyuan Central Hospital, Taiyuan, People’s Republic of China
| | - Wei Wei
- Department of Anaesthesiology, Taiyuan Central Hospital, Taiyuan, People’s Republic of China
- School of Anesthesia, Shanxi Medical University, Shanxi, People’s Republic of China
| | - Jun Fang
- Department of Anaesthesiology, Taiyuan Central Hospital, Taiyuan, People’s Republic of China
- School of Anesthesia, Shanxi Medical University, Shanxi, People’s Republic of China
| | - Yating Xue
- Department of Anaesthesiology, Taiyuan Central Hospital, Taiyuan, People’s Republic of China
- School of Anesthesia, Shanxi Medical University, Shanxi, People’s Republic of China
| | - Jiacheng Wei
- Department of Anaesthesiology, Taiyuan Central Hospital, Taiyuan, People’s Republic of China
- School of Anesthesia, Shanxi Medical University, Shanxi, People’s Republic of China
| |
Collapse
|
139
|
Francis EA, Rangamani P. Computational modeling establishes mechanotransduction as a potent modulator of the mammalian circadian clock. J Cell Sci 2024; 137:jcs261782. [PMID: 39140137 PMCID: PMC11423814 DOI: 10.1242/jcs.261782] [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: 01/03/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024] Open
Abstract
Mechanotransduction, which is the integration of mechanical signals from the external environment of a cell to changes in intracellular signaling, governs many cellular functions. Recent studies have shown that the mechanical state of the cell is also coupled to the cellular circadian clock. To investigate possible interactions between circadian rhythms and cellular mechanotransduction, we have developed a computational model that integrates the two pathways. We postulated that translocation of the transcriptional regulators MRTF (herein referring to both MRTF-A and MRTF-B), YAP and TAZ (also known as YAP1 and WWTR1, respectively; collectively denoted YAP/TAZ) into the nucleus leads to altered expression of circadian proteins. Simulations from our model predict that lower levels of cytoskeletal activity are associated with longer circadian oscillation periods and higher oscillation amplitudes, which is consistent with recent experimental observations. Furthermore, accumulation of YAP/TAZ and MRTF in the nucleus causes circadian oscillations to decay in our model. These effects hold both at the single-cell level and within a population-level framework. Finally, we investigated the effects of mutations in YAP or lamin A, the latter of which result in a class of diseases known as laminopathies. In silico, oscillations in circadian proteins are substantially weaker in populations of cells with mutations in YAP or lamin A, suggesting that defects in mechanotransduction can disrupt the circadian clock in certain disease states; however, reducing substrate stiffness in the model restores normal oscillatory behavior, suggesting a possible compensatory mechanism. Thus, our study identifies that mechanotransduction could be a potent modulatory cue for cellular clocks and that this crosstalk can be leveraged to rescue the circadian clock in disease states.
Collapse
Affiliation(s)
- Emmet A. Francis
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
140
|
Held J, Sivaraman K, Wrenger S, Si W, Welte T, Immenschuh S, Janciauskiene S. Ex vivo study on the human blood neutrophil circadian features and effects of alpha1-antitrypsin and lipopolysaccharide. Vascul Pharmacol 2024; 156:107396. [PMID: 38897556 DOI: 10.1016/j.vph.2024.107396] [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: 03/05/2024] [Revised: 06/10/2024] [Accepted: 06/16/2024] [Indexed: 06/21/2024]
Abstract
AIMS Neutrophils perform various functions in a circadian-dependent manner; therefore, we investigated here whether the effect of alpha1-antitrypsin (AAT), used as augmentation therapy, is dependent on the neutrophil circadian clock. AAT is a vital regulator of neutrophil functions, and its qualitative and/or quantitative defects have significant implications for the development of respiratory diseases. METHODS Whole blood from 12 healthy women age years, mean (SD) 29.92 (5.48) was collected twice daily, 8 h apart, and incubated for 30 min at 37 °C alone or with additions of 2 mg/ml AAT (Respreeza) and/or 5 μg/ml lipopolysaccharide (LPS) from Escherichia coli. Neutrophils were then isolated to examine gene expression, migration and phagocytosis. RESULTS The expression of CD14, CD16, CXCR2 and SELL (encoding CD62L) genes was significantly higher while CDKN1A lower in the afternoon than in the morning neutrophils from untreated blood. Neutrophils isolated in the afternoon had higher migratory and phagocytic activity. Morning neutrophils isolated from AAT-pretreated blood showed higher expression of CXCR2 and SELL than those from untreated morning blood. Pretreatment of blood with AAT enhanced migratory properties of morning but not afternoon neutrophils. Of all genes analysed, only CXCL8 expression was strongly upregulated in morning and afternoon neutrophils isolated from LPS-pretreated blood, whereas CXCR2 expression was downregulated in afternoon neutrophils. The addition of AAT did not reverse the effects of LPS. SIGNIFICANCE The circadian clock of myeloid cells may affect the effectiveness of various therapies, including AAT therapy used to treat patients with AAT deficiency, and needs further investigation.
Collapse
Affiliation(s)
- Julia Held
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany; Hannover Medical School, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - Kokilavani Sivaraman
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany; Hannover Medical School, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - Sabine Wrenger
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany; Hannover Medical School, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - Wenzhang Si
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany; Hannover Medical School, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - Tobias Welte
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany; Hannover Medical School, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - Stephan Immenschuh
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Sabina Janciauskiene
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany; Hannover Medical School, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany.
| |
Collapse
|
141
|
Francia M, Bot M, Boltz T, De la Hoz JF, Boks M, Kahn RS, Ophoff RA. Fibroblasts as an in vitro model of circadian genetic and genomic studies. Mamm Genome 2024; 35:432-444. [PMID: 38960898 PMCID: PMC11329553 DOI: 10.1007/s00335-024-10050-7] [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: 05/09/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
Bipolar disorder (BD) is a heritable disorder characterized by shifts in mood that manifest in manic or depressive episodes. Clinical studies have identified abnormalities of the circadian system in BD patients as a hallmark of underlying pathophysiology. Fibroblasts are a well-established in vitro model for measuring circadian patterns. We set out to examine the underlying genetic architecture of circadian rhythm in fibroblasts, with the goal to assess its contribution to the polygenic nature of BD disease risk. We collected, from primary cell lines of 6 healthy individuals, temporal genomic features over a 48 h period from transcriptomic data (RNA-seq) and open chromatin data (ATAC-seq). The RNA-seq data showed that only a limited number of genes, primarily the known core clock genes such as ARNTL, CRY1, PER3, NR1D2 and TEF display circadian patterns of expression consistently across cell cultures. The ATAC-seq data identified that distinct transcription factor families, like those with the basic helix-loop-helix motif, were associated with regions that were increasing in accessibility over time. Whereas known glucocorticoid receptor target motifs were identified in those regions that were decreasing in accessibility. Further evaluation of these regions using stratified linkage disequilibrium score regression analysis failed to identify a significant presence of them in the known genetic architecture of BD, and other psychiatric disorders or neurobehavioral traits in which the circadian rhythm is affected. In this study, we characterize the biological pathways that are activated in this in vitro circadian model, evaluating the relevance of these processes in the context of the genetic architecture of BD and other disorders, highlighting its limitations and future applications for circadian genomic studies.
Collapse
Affiliation(s)
- Marcelo Francia
- Interdepartmental Program for Neuroscience, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| | - Merel Bot
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, USA
| | - Toni Boltz
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Juan F De la Hoz
- Bioinformatics Interdepartamental Program, University of California Los Angeles, Los Angeles, CA, USA
| | - Marco Boks
- Department Psychiatry, Brain Center University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - René S Kahn
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Roel A Ophoff
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
142
|
Ritter AJ, Wallace A, Ronaghi N, Sanford J. junctionCounts: comprehensive alternative splicing analysis and prediction of isoform-level impacts to the coding sequence. NAR Genom Bioinform 2024; 6:lqae093. [PMID: 39131822 PMCID: PMC11310779 DOI: 10.1093/nargab/lqae093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/15/2024] [Accepted: 07/22/2024] [Indexed: 08/13/2024] Open
Abstract
Alternative splicing (AS) is emerging as an important regulatory process for complex biological processes. Transcriptomic studies therefore commonly involve the identification and quantification of alternative processing events, but the need for predicting the functional consequences of changes to the relative inclusion of alternative events remains largely unaddressed. Many tools exist for the former task, albeit each constrained to its own event type definitions. Few tools exist for the latter task; each with significant limitations. To address these issues we developed junctionCounts, which captures both simple and complex pairwise AS events and quantifies them with straightforward exon-exon and exon-intron junction reads in RNA-seq data, performing competitively among similar tools in terms of sensitivity, false discovery rate and quantification accuracy. Its partner utility, cdsInsertion, identifies transcript coding sequence (CDS) information via in silico translation from annotated start codons, including the presence of premature termination codons. Finally, findSwitchEvents connects AS events with CDS information to predict the impact of individual events to the isoform-level CDS. We used junctionCounts to characterize splicing dynamics and NMD regulation during neuronal differentiation across four primates, demonstrating junctionCounts' capacity to robustly characterize AS in a variety of organisms and to predict its effect on mRNA isoform fate.
Collapse
Affiliation(s)
- Alexander J Ritter
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Andrew Wallace
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Neda Ronaghi
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Jeremy R Sanford
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| |
Collapse
|
143
|
Xu B, Braun R. Variational inference of single cell time series. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.29.610389. [PMID: 39257806 PMCID: PMC11384007 DOI: 10.1101/2024.08.29.610389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Time course single-cell RNA sequencing (scRNA-seq) enables researchers to probe genome-wide expression dynamics at the the single cell scale. However, when gene expression is affected jointly by time and cellular identity, analyzing such data - including conducting cell type annotation and modeling cell type-dependent dynamics - becomes challenging. To address this problem, we propose SNOW (SiNgle cell flOW map), a deep learning algorithm to deconvolve single cell time series data into time-dependent and time-independent contributions. SNOW has a number of advantages. First, it enables cell type annotation based on the time-independent dimensions. Second, it yields a probabilistic model that can be used to discriminate between biological temporal variation and batch effects contaminating individual timepoints, and provides an approach to mitigate batch effects. Finally, it is capable of projecting cells forward and backward in time, yielding time series at the individual cell level. This enables gene expression dynamics to be studied without the need for clustering or pseudobulking, which can be error prone and result in information loss. We describe our probabilistic framework in detail and demonstrate SNOW using data from three distinct time course scRNA-seq studies. Our results show that SNOW is able to construct biologically meaningful latent spaces, remove batch effects, and generate realistic time-series at the single-cell level. By way of example, we illustrate how the latter may be used to enhance the detection of cell type-specific circadian gene expression rhythms, and may be readily extended to other time-series analyses.
Collapse
Affiliation(s)
- Bingxian Xu
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
- NSF-Simons National Institute for Theory and Mathematics in Biology, Chicago, IL 60611, USA
| | - Rosemary Braun
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
- NSF-Simons National Institute for Theory and Mathematics in Biology, Chicago, IL 60611, USA
- Department of Engineering Sciences and Applied Mathematics, Northwestern University, Evanston, IL 60208, USA
- Department of Physics and Astronomy, Northwestern University, Evanston, IL 60208, USA
- Northwestern Institute on Complex Systems, Northwestern University, Evanston, IL 60208, USA
- Santa Fe Institute, Santa Fe, NM 87501, USA
| |
Collapse
|
144
|
Ector C, Schmal C, Didier J, De Landtsheer S, Finger AM, Müller-Marquardt F, Schulte JH, Sauter T, Keilholz U, Herzel H, Kramer A, Granada AE. Time-of-day effects of cancer drugs revealed by high-throughput deep phenotyping. Nat Commun 2024; 15:7205. [PMID: 39169017 PMCID: PMC11339390 DOI: 10.1038/s41467-024-51611-3] [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: 11/07/2023] [Accepted: 08/13/2024] [Indexed: 08/23/2024] Open
Abstract
The circadian clock, a fundamental biological regulator, governs essential cellular processes in health and disease. Circadian-based therapeutic strategies are increasingly gaining recognition as promising avenues. Aligning drug administration with the circadian rhythm can enhance treatment efficacy and minimize side effects. Yet, uncovering the optimal treatment timings remains challenging, limiting their widespread adoption. In this work, we introduce a high-throughput approach integrating live-imaging and data analysis techniques to deep-phenotype cancer cell models, evaluating their circadian rhythms, growth, and drug responses. We devise a streamlined process for profiling drug sensitivities across different times of the day, identifying optimal treatment windows and responsive cell types and drug combinations. Finally, we implement multiple computational tools to uncover cellular and genetic factors shaping time-of-day drug sensitivity. Our versatile approach is adaptable to various biological models, facilitating its broad application and relevance. Ultimately, this research leverages circadian rhythms to optimize anti-cancer drug treatments, promising improved outcomes and transformative treatment strategies.
Collapse
Affiliation(s)
- Carolin Ector
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Faculty of Life Sciences, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christoph Schmal
- Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jeff Didier
- Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Sébastien De Landtsheer
- Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Anna-Marie Finger
- Institute for Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
| | - Francesca Müller-Marquardt
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Institute of Research for Development, University of Montpellier, Montpellier, France
| | - Johannes H Schulte
- Department of Pediatric Oncology, Hematology and Stem Cell Transplantation, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Clinic for Pediatrics and Adolescent Medicine, Universitätsklinikum Tübingen, Tübingen, Germany
| | - Thomas Sauter
- Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Ulrich Keilholz
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Berlin, Germany
| | - Hanspeter Herzel
- Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Achim Kramer
- Institute for Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Adrián E Granada
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, Berlin, Germany.
- German Cancer Consortium (DKTK), Berlin, Germany.
| |
Collapse
|
145
|
Malik DM, Rhoades SD, Zhang SL, Sengupta A, Barber A, Haynes P, Arnadottir ES, Pack A, Kibbey RG, Kain P, Sehgal A, Weljie AM. Glucose Challenge Uncovers Temporal Fungibility of Metabolic Homeostasis over a day:night cycle. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.30.564837. [PMID: 37961230 PMCID: PMC10634956 DOI: 10.1101/2023.10.30.564837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Rhythmicity is a cornerstone of behavioral and biological processes, especially metabolism, yet the mechanisms behind metabolite cycling remain elusive. This study uncovers a robust oscillation in key metabolite pathways downstream of glucose in humans. A purpose-built 13C6-glucose isotope tracing platform was used to sample Drosophila every 4h and probe these pathways, revealing a striking peak in biosynthesis shortly after lights-on in wild-type flies. A hyperactive mutant (fumin) demonstrates increased Krebs cycle labelling and dawn-specific glycolysis labelling. Surprisingly, neither underlying feeding rhythms nor the presence of food availability explain the rhythmicity of glucose processing across genotypes, suggesting a robust internal mechanism for metabolic control of glucose processing. These results align with clinical data highlighting detrimental effects of mistimed energy intake. Our approach offers a unique insight into the dynamic range of daily metabolic processing and provides a mechanistic foundation for exploring circadian metabolic homeostasis in disease contexts.
Collapse
Affiliation(s)
- Dania M. Malik
- Pharmacology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics
- Institute for Translational Medicine and Therapeutics
- These authors contributed equally
| | - Seth D. Rhoades
- Pharmacology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics
- Institute for Translational Medicine and Therapeutics
- Fulgens Consulting, LLC, Cambridge, Massachusetts 02142, USA
- These authors contributed equally
| | - Shirley L. Zhang
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Howard Hughes Medical Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Arjun Sengupta
- Department of Systems Pharmacology and Translational Therapeutics
- Institute for Translational Medicine and Therapeutics
| | - Annika Barber
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Howard Hughes Medical Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers, The State University of New Jersey, New Brunswick, New Jersey 08854, USA
| | - Paula Haynes
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Howard Hughes Medical Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Erna Sif Arnadottir
- Division of Sleep Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Allan Pack
- Division of Sleep Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Richard G. Kibbey
- Department of Internal Medicine, Department of Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | - Pinky Kain
- Department of Systems Pharmacology and Translational Therapeutics
- Institute for Translational Medicine and Therapeutics
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Amita Sehgal
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Howard Hughes Medical Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Aalim M. Weljie
- Department of Systems Pharmacology and Translational Therapeutics
- Institute for Translational Medicine and Therapeutics
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
146
|
Jamadar A, Ward CJ, Remadevi V, Varghese MM, Pabla NS, Gumz ML, Rao R. Circadian clock disruption and growth of kidney cysts in autosomal dominant polycystic kidney disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.05.606676. [PMID: 39211074 PMCID: PMC11361200 DOI: 10.1101/2024.08.05.606676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Background Autosomal dominant polycystic kidney disease (ADPKD) is caused by mutations in the PKD1 and PKD2 genes, and often progresses to kidney failure. ADPKD progression is not uniform among patients, suggesting that factors secondary to the PKD1/2 gene mutation could regulate the rate of disease progression. Here we tested the effect of circadian clock disruption on ADPKD progression. Circadian rhythms are regulated by cell-autonomous circadian clocks composed of clock proteins. BMAL1 is a core constituent of the circadian clock. Methods To disrupt the circadian clock, we deleted Bmal1 gene in the renal collecting ducts of the Pkd1 RC/RC (RC/RC) mouse model of ADPKD (RC/RC; Bmal1 f/f ; Pkhd1 cre , called DKO mice), and in Pkd1 knockout mouse inner medullary collecting duct cells ( Pkd1Bmal1 KO mIMCD3 cells). Only male mice were used. Results Human nephrectomy ADPKD kidneys and Pkd1 KO mIMCD3 cells showed reduced Bmal1 gene expression compared to normal controls. When compared to RC/RC kidneys, DKO kidneys showed significantly altered clock gene expression, increased cyst growth, cell proliferation, apoptosis and fibrosis. DKO kidneys also showed increased lipogenesis and cholesterol synthesis-related gene expression, and increased tissue triglyceride levels compared to RC/RC kidneys. Similarly, in vitro, Pkd1Bmal1 KO cells showed altered clock genes, increased lipogenesis and cholesterol synthesis-related genes, and reduced fatty-acid oxidation-related gene expression compared to Pkd1KO cells. The Pkd1Bmal1 KO cells showed increased cell proliferation compared to Pkd1KO cells, which was rescued by pharmacological inhibition of lipogenesis. Conclusion Renal collecting duct specific Bmal1 gene deletion disrupts the circadian clock and triggers accelerated ADPKD progression by altering lipid metabolism-related gene expression. Key points Lack of BMAL1, a circadian clock protein in renal collecting ducts disrupted the clock and increased cyst growth and fibrosis in an ADPKD mouse model.BMAL1 gene deletion increased cell proliferation by increasing lipogenesis in kidney cells.Thus, circadian clock disruption could be a risk factor for accelerated disease progression in patients with ADPKD.
Collapse
|
147
|
Xu B, Ma D, Abruzzi K, Braun R. Detecting Rhythmic Gene Expression in Single Cell Transcriptomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.07.570691. [PMID: 38105950 PMCID: PMC10723455 DOI: 10.1101/2023.12.07.570691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
An autonomous, environmentally-synchronizable circadian rhythm is a ubiquitous feature of life on Earth. In multicellular organisms, this rhythm is generated by a transcription-translation feedback loop present in nearly every cell that drives daily expression of thousands of genes in a tissue-dependent manner. Identifying the genes that are under circadian control can elucidate the mechanisms by which physiological processes are coordinated in multicellular organisms. Today, transcriptomic profiling at the single-cell level provides an unprecedented opportunity to understand the function of cell-level clocks. However, while many cycling detection algorithms have been developed to identify genes under circadian control in bulk transcriptomic data, it is not known how best to adapt these algorithms to single-cell RNAseq data. Here, we benchmark commonly used circadian detection methods on their reliability and efficiency when applied to single cell RNAseq data. Our results provide guidance on adapting existing cycling detection methods to the single-cell domain, and elucidate opportunities for more robust and efficient rhythm detection in single-cell data. We also propose a subsampling procedure combined with harmonic regression as an efficient strategy to detect circadian genes in the single-cell setting.
Collapse
Affiliation(s)
- Bingxian Xu
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston, IL 60208, USA
| | - Dingbang Ma
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- Shanghai Key Laboratory of Aging Studies, Shanghai 201210, China
| | - Katherine Abruzzi
- HHMI, Brandeis University, Waltham, MA 02453, USA
- Department of Biology, Brandeis University, Waltham, MA 02453
| | - Rosemary Braun
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston, IL 60208, USA
- Department of Engineering Sciences and Applied Mathematics, Northwestern University, Evanston, IL 60208, USA
- Department of Physics and Astronomy, Northwestern University, Evanston, IL 60208, USA
- Northwestern Institute on Complex Systems, Northwestern University, Evanston, IL 60208, USA
| |
Collapse
|
148
|
Jin Y, Wan K, Liu C, Cheng W, Wang R. Mechanisms of exercise intervention in type 2 diabetes: a bibliometric and visualization analysis based on CiteSpace. Front Endocrinol (Lausanne) 2024; 15:1401342. [PMID: 39149117 PMCID: PMC11324446 DOI: 10.3389/fendo.2024.1401342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 07/16/2024] [Indexed: 08/17/2024] Open
Abstract
Objective Type 2 diabetes (T2D) is a common chronic metabolic disease, and its prevalence is increasing globally. Exercise is crucial for T2D management, yet many aspects of its mechanisms remain unclear. This study employs CiteSpace to reveal research hotspots and frontier issues in exercise intervention for T2D. Method A literature review spanning from January 1, 2013 to December 31, 2022, was conducted using the Web of Science Core Collection (WoSCC), with keywords including "exercise," "type 2 diabetes," and "mechanisms." We analyzed network diagrams generated by CiteSpace, which depicted relationships among countries, authors, and keywords. Results This study includes 1,210 English papers from 555 journals, affiliated with 348 institutions across 80 countries/regions. Notably, the United States, China, and the United Kingdom account for nearly half of all publications. The University of Copenhagen leads in publication volume, followed by Harvard Medical School and the University of Colorado. Key authors include Kirwan, John P (Case Western Reserve University), Malin, Steven K (Rutgers University), and Pedersen, Bente Klarlund (University of Copenhagen). Based on co-occurrence analysis of keywords, it is evident that terms such as "disease," "glucagon-like peptide 1," and "cardiovascular risk factor" exhibit high intermediary centrality. Conclusion The analysis highlights ongoing investigations into molecular mechanisms, such as β-cell function enhancement, exerkines, and epigenetic mechanisms. Emerging areas include exercise response heterogeneity, circadian rhythm regulation, transcription factors, neurotrophic factors, and mitochondrial function. Future studies should prioritize understanding interactions between different exercise mechanisms and optimizing exercise prescriptions for T2D. Exercise prescriptions are crucial for effective interventions. Collaboration between countries and institutions is essential to understand the influences of different genetic backgrounds and environmental factors. Currently, a combination of aerobic and resistance training is considered the optimal form of exercise. However, considering time efficiency, high-intensity interval training (HIIT) has gained widespread attention and research due to its ability to achieve similar exercise effects in a shorter duration. Additionally, circadian rhythm regulation may affect the exercise outcomes of diabetic individuals at different times of the day, particularly concerning the specific types, doses, and intensities used for precision intervention in T2D.
Collapse
Affiliation(s)
- Yue Jin
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Kang Wan
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
- Physical Education College, Henan Sport University, Zhengzhou, China
| | - Cheng Liu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Wei Cheng
- Department of Endocrinology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ru Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
149
|
Llewellyn J, Hubbard SJ, Swift J. Translation is an emerging constraint on protein homeostasis in ageing. Trends Cell Biol 2024; 34:646-656. [PMID: 38423854 DOI: 10.1016/j.tcb.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 01/23/2024] [Accepted: 02/01/2024] [Indexed: 03/02/2024]
Abstract
Proteins are molecular machines that provide structure and perform vital transport, signalling and enzymatic roles. Proteins expressed by cells require tight regulation of their concentration, folding, localisation, and modifications; however, this state of protein homeostasis is continuously perturbed by tissue-level stresses. While cells in healthy tissues are able to buffer against these perturbations, for example, by expression of chaperone proteins, protein homeostasis is lost in ageing, and can lead to protein aggregation characteristic of protein folding diseases. Here, we review reports of a progressive disconnect between transcriptomic and proteomic regulation during cellular ageing. We discuss how age-associated changes to cellular responses to specific stressors in the tissue microenvironment are exacerbated by loss of ribosomal proteins, ribosomal pausing, and mistranslation.
Collapse
Affiliation(s)
- Jack Llewellyn
- Wellcome Centre for Cell-Matrix Research, Oxford Road, Manchester, M13 9PT, UK; Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PT, UK
| | - Simon J Hubbard
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PT, UK.
| | - Joe Swift
- Wellcome Centre for Cell-Matrix Research, Oxford Road, Manchester, M13 9PT, UK; Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PT, UK.
| |
Collapse
|
150
|
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
|