1
|
Chatham JC, Patel RP. Protein glycosylation in cardiovascular health and disease. Nat Rev Cardiol 2024; 21:525-544. [PMID: 38499867 DOI: 10.1038/s41569-024-00998-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/13/2024] [Indexed: 03/20/2024]
Abstract
Protein glycosylation, which involves the attachment of carbohydrates to proteins, is one of the most abundant protein co-translational and post-translational modifications. Advances in technology have substantially increased our knowledge of the biosynthetic pathways involved in protein glycosylation, as well as how changes in glycosylation can affect cell function. In addition, our understanding of the role of protein glycosylation in disease processes is growing, particularly in the context of immune system function, infectious diseases, neurodegeneration and cancer. Several decades ago, cell surface glycoproteins were found to have an important role in regulating ion transport across the cardiac sarcolemma. However, with very few exceptions, our understanding of how changes in protein glycosylation influence cardiovascular (patho)physiology remains remarkably limited. Therefore, in this Review, we aim to provide an overview of N-linked and O-linked protein glycosylation, including intracellular O-linked N-acetylglucosamine protein modification. We discuss our current understanding of how all forms of protein glycosylation contribute to normal cardiovascular function and their roles in cardiovascular disease. Finally, we highlight potential gaps in our knowledge about the effects of protein glycosylation on the heart and vascular system, highlighting areas for future research.
Collapse
Affiliation(s)
- John C Chatham
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Rakesh P Patel
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
2
|
Arrieta A, Chapski DJ, Reese A, Kimball TH, Song K, Rosa-Garrido M, Vondriska TM. Circadian control of histone turnover during cardiac development and growth. J Biol Chem 2024; 300:107434. [PMID: 38830405 PMCID: PMC11261805 DOI: 10.1016/j.jbc.2024.107434] [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: 11/30/2023] [Revised: 04/30/2024] [Accepted: 05/21/2024] [Indexed: 06/05/2024] Open
Abstract
During postnatal cardiac hypertrophy, cardiomyocytes undergo mitotic exit, relying on DNA replication-independent mechanisms of histone turnover to maintain chromatin organization and gene transcription. In other tissues, circadian oscillations in nucleosome occupancy influence clock-controlled gene expression, suggesting a role for the circadian clock in temporal control of histone turnover and coordinated cardiomyocyte gene expression. We sought to elucidate roles for the master circadian transcription factor, Bmal1, in histone turnover, chromatin organization, and myocyte-specific gene expression and cell growth in the neonatal period. Bmal1 knockdown in neonatal rat ventricular myocytes decreased myocyte size, total cellular protein synthesis, and transcription of the fetal hypertrophic gene Nppb after treatment with serum or the α-adrenergic agonist phenylephrine. Depletion of Bmal1 decreased the expression of clock-controlled genes Per2 and Tcap, as well as Sik1, a Bmal1 target upregulated in adult versus embryonic hearts. Bmal1 knockdown impaired Per2 and Sik1 promoter accessibility as measured by micrococcal nuclease-quantitative PCR and impaired histone turnover as measured by metabolic labeling of acid-soluble chromatin fractions. Sik1 knockdown in turn decreased myocyte size, while simultaneously inhibiting natriuretic peptide B transcription and activating Per2 transcription. Linking these changes to chromatin remodeling, depletion of the replication-independent histone variant H3.3a inhibited myocyte hypertrophy and prevented phenylephrine-induced changes in clock-controlled gene transcription. Bmal1 is required for neonatal myocyte growth, replication-independent histone turnover, and chromatin organization at the Sik1 promoter. Sik1 represents a novel clock-controlled gene that coordinates myocyte growth with hypertrophic and clock-controlled gene transcription. Replication-independent histone turnover is required for transcriptional remodeling of clock-controlled genes in cardiac myocytes in response to growth stimuli.
Collapse
Affiliation(s)
- Adrian Arrieta
- Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California, USA
| | - Douglas J Chapski
- Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California, USA
| | - Anna Reese
- Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California, USA
| | - Todd H Kimball
- Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California, USA
| | - Kunhua Song
- Internal Medicine, Heart Institute, Center for Regenerative Medicine, University of South Florida, Tampa, Florida, USA
| | - Manuel Rosa-Garrido
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Thomas M Vondriska
- Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California, USA; Division of Cardiology, Department of Medicine, UCLA, Los Angeles, California, USA; Department of Physiology, UCLA, Los Angeles, California, USA; Molecular Biology Institute, UCLA, Los Angeles, California, USA.
| |
Collapse
|
3
|
Gouchoe DA, Ganapathi AM, Cui EY, Henn MC, Yim WY, Geng B, Whitson BA, Zhu H. Is Time Scheduling Important? An Analysis of Donor Heart Cross-clamp Times During Heart Transplantation. Transplant Direct 2024; 10:e1588. [PMID: 38529355 PMCID: PMC10962895 DOI: 10.1097/txd.0000000000001588] [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: 06/07/2023] [Revised: 12/19/2023] [Accepted: 12/29/2023] [Indexed: 03/27/2024] Open
Abstract
Background Outcomes in heart transplantation are affected by a variety of variables and patient factors. However, the impact of circadian rhythms, gene expression, and transcription remain underexplored. We thus evaluated the potential role of donor heart cross-clamp times on short-term and long-term outcomes after heart transplantation. Methods A total of 31 713 heart transplants were identified from the United Network for Organ Sharing Database. Patients were first stratified on the basis of time of donor procurement: 12 am to 12 pm or 12 pm to 12 am. To evaluate a possible effect of circadian rhythms, donor time was further divided into 5 groups based on preclinical data: 4 am to 8 am; 8 am to 11 am; 11 am to 5 pm; 5 pm to 10 pm; 10 pm to 4 am. Groups were assessed with comparative statistics. Long-term survival was evaluated using Kaplan-Meier methods and a multivariate Cox proportional hazard model. Results Patients who received hearts recovered between 12 am and 12 pm had significantly higher survival than those who received hearts recovered between 12 pm and 12 am. This survival difference was observed in both unadjusted (P = 0.002) and adjusted analyses (hazard ratio [HR]: 0.93; 95% confidence interval [CI], 0.89-0.97; P < 0.001). On unadjusted analysis, the survival difference among the 5 groups was insignificant (P = 0.07). Following adjustment, the periods of 11 am to 5 pm (HR: 1.09, 95% CI, 1.02-1.17; P = 0.012), 5 pm to 10 pm (HR: 1.11; 95% CI, 1.04-1.19; P = 0.002), and 10 pm to 4 am (HR: 1.07; 95% CI, 1.01-1.15; P = 0.034), were all independently associated with increased long-term mortality. Notably, the time of 8 am to 11 am was not associated with a change in survival (HR: 1.04; 95% CI, 0.96-1.14; P = 0.3). Conclusions Given the independent association of donor timing and survival after adjustment in a large national cohort, further investigation into the role of donor circadian rhythm and donor procurement time is warranted in preclinical and clinical studies. Understanding the underlying mechanisms of this observation could potentially lead to the development of effective treatments and donor procurement processes that prepare the organs for transplantation in a better condition.
Collapse
Affiliation(s)
- Doug A. Gouchoe
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Asvin M. Ganapathi
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Ervin Y. Cui
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Matthew C. Henn
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Wai Yen Yim
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bingchuan Geng
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bryan A. Whitson
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, College of Medicine, Columbus, OH
| | - Hua Zhu
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, College of Medicine, Columbus, OH
| |
Collapse
|
4
|
Malhan D, Relógio A. A matter of timing? The influence of circadian rhythms on cardiac physiology and disease. Eur Heart J 2024; 45:561-563. [PMID: 38104261 DOI: 10.1093/eurheartj/ehad816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2023] Open
Affiliation(s)
- Deeksha Malhan
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Am Kaiserkai 1, Hamburg 20457, Germany
| | - Angela Relógio
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Am Kaiserkai 1, Hamburg 20457, Germany
- Institute for Theoretical Biology (ITB), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Philippstraße 13, Berlin 10117, Germany
| |
Collapse
|
5
|
Personnic E, Gerard G, Poilbout C, Jetten AM, Gómez AM, Benitah JP, Perrier R. Circadian regulation of Ca V 1.2 expression by RORα in the mouse heart. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.15.575657. [PMID: 38293155 PMCID: PMC10827087 DOI: 10.1101/2024.01.15.575657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Background In addition to show autonomous beating rhythmicity, the physiological functions of the heart present daily periodic oscillations. Notably the ventricular repolarization itself varies throughout the circadian cycle which was mainly related to the periodic expression of K + channels. However, the involvement of the L-type Ca 2+ channel (Ca V 1.2 encoded by Cacna1c gene) in these circadian variations remains elusive. Methods We used a transgenic mouse model (PCa-luc) that expresses the luciferase reporter under the control of the cardiac Cacna1c promoter and analyzed promoter activity by bioluminescent imaging, qPCR, immunoblot, Chromatin immunoprecipitation assay (ChIP) and Ca V 1.2 activity. Results Under normal 12:12h light-dark cycle, we observed in vivo a biphasic diurnal variation of promoter activities peaking at 9 and 19.5 Zeitgeber time (ZT). This was associated with a periodicity of Cacna1c mRNA levels preceding 24-h oscillations of Ca V 1.2 protein levels in ventricle (with a 1.5 h phase shift) but not in atrial heart tissues. The periodicity of promoter activities and Ca V 1.2 proteins, which correlated with biphasic oscillations of L-type Ca 2+ current conductance, persisted in isolated ventricular cardiomyocytes from PCa-Luc mice over the course of the 24-h cycle, suggesting an endogenous cardiac circadian regulation. Comparison of 24-h temporal patterns of clock gene expressions in ventricles and atrial tissues of the same mice revealed conserved circadian oscillations of the core clock genes except for the retinoid-related orphan receptor α gene (RORα), which remained constant throughout the course of a day in atrial tissues. In vitro we found that RORα is recruited to two specific regions on the Cacna1c promoter and that incubation with specific RORα inhibitor disrupted 24-h oscillations of ventricular promoter activities and Ca V 1.2 protein levels. Similar results were observed for pore forming subunits of the K + transient outward currents, K V 4.2 and K V 4.3. Conclusions These findings raise the possibility that the RORα-dependent rhythmic regulation of cardiac Ca V 1.2 and K V 4.2/4.3 throughout the daily cycle may play an important role in physiopathology of heart function.
Collapse
|
6
|
Rabinovich-Nikitin I, Kirshenbaum LA. Circadian regulated control of myocardial ischemia-reperfusion injury. Trends Cardiovasc Med 2024; 34:1-7. [PMID: 36150629 DOI: 10.1016/j.tcm.2022.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/15/2022] [Accepted: 09/16/2022] [Indexed: 01/14/2023]
Abstract
Circadian mechanisms have been associated with the pathogenesis of a variety of cardiovascular diseases, including myocardial ischemia-reperfusion injury (I-R). Myocardial ischemia resulting from impaired oxygen delivery to cardiac muscle sets into motion a cascade of cellular events that paradoxically triggers greater cardiac dysfunction upon reinstitution of coronary blood supply, a phenomenon known as I-R. I-R injury has been attributed to a number of cellular defects including increased reactive oxygen species (ROS), increased intracellular calcium and impaired mitochondrial bioenergetics that ultimately lead to cardiac cell death, ventricular remodeling and heart failure. Emerging evidence has identified a strong correlation between cellular defects that underlie I-R and the disrupted circadian. In fact, recent studies have shown that circadian dysfunction exacerbates cardiac injury following MI from impaired cellular quality control mechanisms such as autophagy, which are vital in the clearance of damaged cellular proteins and organelles such as mitochondria from the cell. The accumulation of cellular debris is posited as the central underlying cause of excessive cardiac cell death and ventricular dysfunction following MI. The complexities that govern the interplay between circadian biology and I-R injury following MI is at its infancy and understanding how circadian misalignment, such as in shift workers impacts I-R injury is of great scientific and clinical importance toward development of new therapeutic strategies using chronotherapy and circadian regulation to mitigate cardiac injury and improve cardiac outcomes after injury. In this review, we highlight recent advances in circadian biology and adaptive cellular quality control mechanisms that influence cardiac injury in response to MI injury with a specific focus on how circadian biology can be utilized to further cardiovascular medicine and patient care.
Collapse
Affiliation(s)
- Inna Rabinovich-Nikitin
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Canada
| | - Lorrie A Kirshenbaum
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Canada; Department of Pharmacology and Therapeutics Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba R2H 2H6, Canada.
| |
Collapse
|
7
|
Santos EW, Khatoon S, Di Mise A, Zheng YM, Wang YX. Mitochondrial Dynamics in Pulmonary Hypertension. Biomedicines 2023; 12:53. [PMID: 38255160 PMCID: PMC10813473 DOI: 10.3390/biomedicines12010053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/12/2023] [Accepted: 12/19/2023] [Indexed: 01/24/2024] Open
Abstract
Mitochondria are essential organelles for energy production, calcium homeostasis, redox signaling, and other cellular responses involved in pulmonary vascular biology and disease processes. Mitochondrial homeostasis depends on a balance in mitochondrial fusion and fission (dynamics). Mitochondrial dynamics are regulated by a viable circadian clock. Hypoxia and nicotine exposure can cause dysfunctions in mitochondrial dynamics, increases in mitochondrial reactive oxygen species generation and calcium concentration, and decreases in ATP production. These mitochondrial changes contribute significantly to pulmonary vascular oxidative stress, inflammatory responses, contractile dysfunction, pathologic remodeling, and eventually pulmonary hypertension. In this review article, therefore, we primarily summarize recent advances in basic, translational, and clinical studies of circadian roles in mitochondrial metabolism in the pulmonary vasculature. This knowledge may not only be crucial to fully understanding the development of pulmonary hypertension, but also greatly help to create new therapeutic strategies for treating this devastating disease and other related pulmonary disorders.
Collapse
Affiliation(s)
- Ed Wilson Santos
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (E.W.S.); (S.K.); (A.D.M.)
| | - Subika Khatoon
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (E.W.S.); (S.K.); (A.D.M.)
| | - Annarita Di Mise
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (E.W.S.); (S.K.); (A.D.M.)
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Via Orabona, 4, 70125 Bari, Italy
| | - Yun-Min Zheng
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (E.W.S.); (S.K.); (A.D.M.)
| | - Yong-Xiao Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (E.W.S.); (S.K.); (A.D.M.)
| |
Collapse
|
8
|
Goodman MO, Dashti HS, Lane JM, Windred DP, Burns A, Jones SE, Sofer T, Purcell SM, Zhu X, Ollila HM, Kyle SD, Spiegelhalder K, Peker Y, Huang T, Cain SW, Phillips AJK, Saxena R, Rutter MK, Redline S, Wang H. Causal Association Between Subtypes of Excessive Daytime Sleepiness and Risk of Cardiovascular Diseases. J Am Heart Assoc 2023; 12:e030568. [PMID: 38084713 PMCID: PMC10863774 DOI: 10.1161/jaha.122.030568] [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: 06/09/2023] [Accepted: 10/03/2023] [Indexed: 12/20/2023]
Abstract
BACKGROUND Excessive daytime sleepiness (EDS), experienced in 10% to 20% of the population, has been associated with cardiovascular disease and death. However, the condition is heterogeneous and is prevalent in individuals having short and long sleep duration. We sought to clarify the relationship between sleep duration subtypes of EDS with cardiovascular outcomes, accounting for these subtypes. METHODS AND RESULTS We defined 3 sleep duration subtypes of excessive daytime sleepiness: normal (6-9 hours), short (<6 hours), and long (>9 hours), and compared these with a nonsleepy, normal-sleep-duration reference group. We analyzed their associations with incident myocardial infarction (MI) and stroke using medical records of 355 901 UK Biobank participants and performed 2-sample Mendelian randomization for each outcome. Compared with healthy sleep, long-sleep EDS was associated with an 83% increased rate of MI (hazard ratio, 1.83 [95% CI, 1.21-2.77]) during 8.2-year median follow-up, adjusting for multiple health and sociodemographic factors. Mendelian randomization analysis provided supporting evidence of a causal role for a genetic long-sleep EDS subtype in MI (inverse-variance weighted β=1.995, P=0.001). In contrast, we did not find evidence that other subtypes of EDS were associated with incident MI or any associations with stroke (P>0.05). CONCLUSIONS Our study suggests the previous evidence linking EDS with increased cardiovascular disease risk may be primarily driven by the effect of its long-sleep subtype on higher risk of MI. Underlying mechanisms remain to be investigated but may involve sleep irregularity and circadian disruption, suggesting a need for novel interventions in this population.
Collapse
Affiliation(s)
- Matthew O. Goodman
- Division of Sleep and Circadian DisordersBrigham and Women’s HospitalBostonMA
- Department of Neurology and MedicineHarvard Medical School, Brigham and Women’s HospitalBostonMA
- Broad InstituteCambridgeMA
| | - Hassan S. Dashti
- Broad InstituteCambridgeMA
- Center for Genomic MedicineMassachusetts General Hospital and Harvard Medical SchoolBostonMA
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General HospitalBostonMA
| | - Jacqueline M. Lane
- Division of Sleep and Circadian DisordersBrigham and Women’s HospitalBostonMA
- Department of Neurology and MedicineHarvard Medical School, Brigham and Women’s HospitalBostonMA
- Broad InstituteCambridgeMA
- Center for Genomic MedicineMassachusetts General Hospital and Harvard Medical SchoolBostonMA
| | - Daniel P. Windred
- School of Psychological SciencesTurner Institute for Brain and Mental Health, Monash UniversityMelbourneVictoriaAustralia
| | - Angus Burns
- Broad InstituteCambridgeMA
- Center for Genomic MedicineMassachusetts General Hospital and Harvard Medical SchoolBostonMA
- School of Psychological SciencesTurner Institute for Brain and Mental Health, Monash UniversityMelbourneVictoriaAustralia
| | - Samuel E. Jones
- Institute for Molecular Medicine Finland (FIMM)University of HelsinkiFinland
- University of Exeter Medical SchoolExeterUnited Kingdom
| | - Tamar Sofer
- Division of Sleep and Circadian DisordersBrigham and Women’s HospitalBostonMA
- Department of Neurology and MedicineHarvard Medical School, Brigham and Women’s HospitalBostonMA
- Department of BiostatisticsHarvard T.H. Chan School of Public HealthBostonMA
| | - Shaun M. Purcell
- Division of Sleep and Circadian DisordersBrigham and Women’s HospitalBostonMA
- Department of Neurology and MedicineHarvard Medical School, Brigham and Women’s HospitalBostonMA
- Broad InstituteCambridgeMA
- Department of PsychiatryBrigham and Women’s HospitalBostonMA
| | - Xiaofeng Zhu
- Department of Population and Quantitative Health SciencesCase Western Reserve UniversityClevelandOH
| | - Hanna M. Ollila
- Broad InstituteCambridgeMA
- Center for Genomic MedicineMassachusetts General Hospital and Harvard Medical SchoolBostonMA
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General HospitalBostonMA
- Institute for Molecular Medicine Finland (FIMM)University of HelsinkiFinland
| | - Simon D. Kyle
- Sleep and Circadian Neuroscience Institute, Nuffield Department of Clinical NeurosciencesUniversity of OxfordUnited Kingdom
| | - Kai Spiegelhalder
- Department of Psychiatry and PsychotherapyMedical Centre–University of Freiburg, Faculty of Medicine, University of FreiburgFreiburgGermany
| | - Yuksel Peker
- Division of Sleep and Circadian DisordersBrigham and Women’s HospitalBostonMA
- Department of Neurology and MedicineHarvard Medical School, Brigham and Women’s HospitalBostonMA
- Department of Pulmonary MedicineKoç University School of MedicineIstanbulTurkey
- Sahlgrenska AcademyUniversity of GothenburgSweden
- Department of Clinical Sciences, Respiratory Medicine and Allergology, Faculty of MedicineLund UniversityLundSweden
- Division of Pulmonary, Allergy, and Critical Care MedicineUniversity of Pittsburgh School of MedicinePittsburghPA
| | - Tianyi Huang
- Department of Neurology and MedicineHarvard Medical School, Brigham and Women’s HospitalBostonMA
- Channing Division of Network MedicineBrigham and Women’s Hospital, Harvard Medical SchoolBostonMA
| | - Sean W. Cain
- School of Psychological SciencesTurner Institute for Brain and Mental Health, Monash UniversityMelbourneVictoriaAustralia
| | - Andrew J. K. Phillips
- School of Psychological SciencesTurner Institute for Brain and Mental Health, Monash UniversityMelbourneVictoriaAustralia
| | - Richa Saxena
- Broad InstituteCambridgeMA
- Center for Genomic MedicineMassachusetts General Hospital and Harvard Medical SchoolBostonMA
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General HospitalBostonMA
| | - Martin K. Rutter
- Division of Endocrinology, Diabetes & Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUnited Kingdom
- Diabetes, Endocrinology and Metabolism CentreManchester University NHS Foundation Trust, NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science CentreManchesterUnited Kingdom
| | - Susan Redline
- Division of Sleep and Circadian DisordersBrigham and Women’s HospitalBostonMA
- Department of Neurology and MedicineHarvard Medical School, Brigham and Women’s HospitalBostonMA
| | - Heming Wang
- Division of Sleep and Circadian DisordersBrigham and Women’s HospitalBostonMA
- Department of Neurology and MedicineHarvard Medical School, Brigham and Women’s HospitalBostonMA
- Broad InstituteCambridgeMA
| |
Collapse
|
9
|
Kane MS, Benavides GA, Osuma E, Johnson MS, Collins HE, He Y, Westbrook D, Litovsky SH, Mitra K, Chatham JC, Darley-Usmar V, Young ME, Zhang J. The interplay between sex, time of day, fasting status, and their impact on cardiac mitochondrial structure, function, and dynamics. Sci Rep 2023; 13:21638. [PMID: 38062139 PMCID: PMC10703790 DOI: 10.1038/s41598-023-49018-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 12/03/2023] [Indexed: 12/18/2023] Open
Abstract
Mitochondria morphology and function, and their quality control by mitophagy, are essential for heart function. We investigated whether these are influenced by time of the day (TOD), sex, and fed or fasting status, using transmission electron microscopy (EM), mitochondrial electron transport chain (ETC) activity, and mito-QC reporter mice. We observed peak mitochondrial number at ZT8 in the fed state, which was dependent on the intrinsic cardiac circadian clock, as hearts from cardiomyocyte-specific BMAL1 knockout (CBK) mice exhibit different TOD responses. In contrast to mitochondrial number, mitochondrial ETC activities do not fluctuate across TOD, but decrease immediately and significantly in response to fasting. Concurrent with the loss of ETC activities, ETC proteins were decreased with fasting, simultaneous with significant increases of mitophagy, mitochondrial antioxidant protein SOD2, and the fission protein DRP1. Fasting-induced mitophagy was lost in CBK mice, indicating a direct role of BMAL1 in regulating mitophagy. This is the first of its kind report to demonstrate the interactions between sex, fasting, and TOD on cardiac mitochondrial structure, function and mitophagy. These studies provide a foundation for future investigations of mitochondrial functional perturbation in aging and heart diseases.
Collapse
Affiliation(s)
- Mariame S Kane
- Department of Pathology, University of Alabama at Birmingham, 901 19th Street S., Birmingham, AL, BMRII-53435294-0017, USA
- Birmingham VA Health Care System (BVACS), Birmingham, USA
| | - Gloria A Benavides
- Department of Pathology, University of Alabama at Birmingham, 901 19th Street S., Birmingham, AL, BMRII-53435294-0017, USA
| | - Edie Osuma
- Department of Pathology, University of Alabama at Birmingham, 901 19th Street S., Birmingham, AL, BMRII-53435294-0017, USA
- Baylor College of Medicine, Houston, USA
| | - Michelle S Johnson
- Department of Pathology, University of Alabama at Birmingham, 901 19th Street S., Birmingham, AL, BMRII-53435294-0017, USA
| | - Helen E Collins
- Department of Pathology, University of Alabama at Birmingham, 901 19th Street S., Birmingham, AL, BMRII-53435294-0017, USA
- Department of Medicine, University of Louisville, Louisville, USA
| | - Yecheng He
- Department of Pathology, University of Alabama at Birmingham, 901 19th Street S., Birmingham, AL, BMRII-53435294-0017, USA
- Department of Clinical Medicine, Suzhou Vocational Health College, Suzhou, Jiangsu, China
| | - David Westbrook
- Department of Pathology, University of Alabama at Birmingham, 901 19th Street S., Birmingham, AL, BMRII-53435294-0017, USA
| | - Silvio H Litovsky
- Department of Pathology, University of Alabama at Birmingham, 901 19th Street S., Birmingham, AL, BMRII-53435294-0017, USA
| | - Kasturi Mitra
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
- Ashoka University, Sonipat, NCR (Delhi), India
| | - John C Chatham
- Department of Pathology, University of Alabama at Birmingham, 901 19th Street S., Birmingham, AL, BMRII-53435294-0017, USA
| | - Victor Darley-Usmar
- Department of Pathology, University of Alabama at Birmingham, 901 19th Street S., Birmingham, AL, BMRII-53435294-0017, USA
| | - Martin E Young
- Department of Medicine, University of Alabama at Birmingham, 703 19th St. S., ZRB 308, Birmingham, AL, 35294, USA.
| | - Jianhua Zhang
- Department of Pathology, University of Alabama at Birmingham, 901 19th Street S., Birmingham, AL, BMRII-53435294-0017, USA.
| |
Collapse
|
10
|
Arrieta A, Chapski DJ, Reese A, Kimball T, Song K, Rosa-Garrido M, Vondriska TM. Circadian Control of Histone Turnover During Cardiac Development and Growth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.14.567086. [PMID: 38014083 PMCID: PMC10680691 DOI: 10.1101/2023.11.14.567086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Rationale: During postnatal cardiac hypertrophy, cardiomyocytes undergo mitotic exit, relying on DNA replication-independent mechanisms of histone turnover to maintain chromatin organization and gene transcription. In other tissues, circadian oscillations in nucleosome occupancy influence clock-controlled gene expression, suggesting an unrecognized role for the circadian clock in temporal control of histone turnover and coordinate cardiomyocyte gene expression. Objective: To elucidate roles for the master circadian transcription factor, Bmal1, in histone turnover, chromatin organization, and myocyte-specific gene expression and cell growth in the neonatal period. Methods and Results: Bmal1 knockdown in neonatal rat ventricular myocytes (NRVM) decreased myocyte size, total cellular protein, and transcription of the fetal hypertrophic gene Nppb following treatment with increasing serum concentrations or the α-adrenergic agonist phenylephrine (PE). Bmal1 knockdown decreased expression of clock-controlled genes Per2 and Tcap, and salt-inducible kinase 1 (Sik1) which was identified via gene ontology analysis of Bmal1 targets upregulated in adult versus embryonic hearts. Epigenomic analyses revealed co-localized chromatin accessibility and Bmal1 localization in the Sik1 promoter. Bmal1 knockdown impaired Per2 and Sik1 promoter accessibility as measured by MNase-qPCR and impaired histone turnover indicated by metabolic labeling of acid-soluble chromatin fractions and immunoblots of total and chromatin-associated core histones. Sik1 knockdown basally increased myocyte size, while simultaneously impairing and driving Nppb and Per2 transcription, respectively. Conclusions: Bmal1 is required for neonatal myocyte growth, replication-independent histone turnover, and chromatin organization at the Sik1 promoter. Sik1 represents a novel clock-controlled gene that coordinates myocyte growth with hypertrophic and clock-controlled gene transcription.
Collapse
|
11
|
Chen R, Routh BN, Gaudet AD, Fonken LK. Circadian Regulation of the Neuroimmune Environment Across the Lifespan: From Brain Development to Aging. J Biol Rhythms 2023; 38:419-446. [PMID: 37357738 PMCID: PMC10475217 DOI: 10.1177/07487304231178950] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023]
Abstract
Circadian clocks confer 24-h periodicity to biological systems, to ultimately maximize energy efficiency and promote survival in a world with regular environmental light cycles. In mammals, circadian rhythms regulate myriad physiological functions, including the immune, endocrine, and central nervous systems. Within the central nervous system, specialized glial cells such as astrocytes and microglia survey and maintain the neuroimmune environment. The contributions of these neuroimmune cells to both homeostatic and pathogenic demands vary greatly across the day. Moreover, the function of these cells changes across the lifespan. In this review, we discuss circadian regulation of the neuroimmune environment across the lifespan, with a focus on microglia and astrocytes. Circadian rhythms emerge in early life concurrent with neuroimmune sculpting of brain circuits and wane late in life alongside increasing immunosenescence and neurodegeneration. Importantly, circadian dysregulation can alter immune function, which may contribute to susceptibility to neurodevelopmental and neurodegenerative diseases. In this review, we highlight circadian neuroimmune interactions across the lifespan and share evidence that circadian dysregulation within the neuroimmune system may be a critical component in human neurodevelopmental and neurodegenerative diseases.
Collapse
Affiliation(s)
- Ruizhuo Chen
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
| | - Brandy N. Routh
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
- Institute for Neuroscience, The University of Texas at Austin, Austin, Texas
| | - Andrew D. Gaudet
- Institute for Neuroscience, The University of Texas at Austin, Austin, Texas
- Department of Psychology, The University of Texas at Austin, Austin, Texas
- Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, Texas
| | - Laura K. Fonken
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
- Institute for Neuroscience, The University of Texas at Austin, Austin, Texas
| |
Collapse
|
12
|
Alam S, Revi GS, Kerick SE, Yang X, Robucci R, Banerjee N, Spangler DP. Anticipatory cardiac deceleration estimates cognitive performance in virtual reality beyond tonic heart period and heart period variability. Biol Psychol 2023; 181:108602. [PMID: 37295768 DOI: 10.1016/j.biopsycho.2023.108602] [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: 07/29/2022] [Revised: 06/05/2023] [Accepted: 06/05/2023] [Indexed: 06/12/2023]
Abstract
Anticipatory cardiac deceleration is the lengthening of heart period before an expected event. It appears to reflect preparation that supports rapid action. The current study sought to bolster anticipatory deceleration as a practical and unique estimator of performance efficiency. To this end, we examined relationships between deceleration and virtual reality performance under low and high time pressure. Importantly, we investigated whether deceleration separately estimates performance beyond basal heart period and basal high-frequency heart rate variability (other vagally influenced metrics related to cognition). Thirty participants completed an immersive virtual reality (VR) cognitive performance task across six longitudinal sessions. Anticipatory deceleration and basal heart period/heart period variability were quantified from electrocardiography collected during pre-task anticipatory countdowns and baseline periods, respectively. At the between-person level, we found that greater anticipatory declaration was related to superior accuracy and faster response times (RT). The relation between deceleration and accuracy was stronger under high relative to low time pressure, when good performance requires greater efficiency. Findings for heart period and heart period variability largely converge with the prior literature, but importantly, were statistically separate from deceleration effects on performance. Lastly, deceleration effects were detected using anticipatory periods that are more practical (shorter and more intermittent) than those typically employed. Taken together, findings suggest that anticipatory deceleration is a unique and practical correlate of cognitive-motor efficiency apart from heart period and heart period variability in virtual reality.
Collapse
Affiliation(s)
- Sazedul Alam
- Department of Computer Science and Electrical Engineering, University of Maryland - Baltimore County, Baltimore, MD, USA
| | - Gabriela S Revi
- Department of Biobehavioral Health, Pennsylvania State University, University Park, PA, USA
| | | | - Xiao Yang
- Department of Psychology, Old Dominion University, Norfolk, VA, USA
| | - Ryan Robucci
- Department of Computer Science and Electrical Engineering, University of Maryland - Baltimore County, Baltimore, MD, USA
| | - Nilanjan Banerjee
- Department of Computer Science and Electrical Engineering, University of Maryland - Baltimore County, Baltimore, MD, USA
| | - Derek P Spangler
- Department of Biobehavioral Health, Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
13
|
Sveric KM, Botan R, Dindane Z, Winkler A, Nowack T, Heitmann C, Schleußner L, Linke A. Single-Site Experience with an Automated Artificial Intelligence Application for Left Ventricular Ejection Fraction Measurement in Echocardiography. Diagnostics (Basel) 2023; 13:diagnostics13071298. [PMID: 37046515 PMCID: PMC10093353 DOI: 10.3390/diagnostics13071298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/28/2023] [Accepted: 03/28/2023] [Indexed: 03/31/2023] Open
Abstract
Left ventricular ejection fraction (LVEF) is a key parameter in evaluating left ventricular (LV) function using echocardiography (Echo), but its manual measurement by the modified biplane Simpson (MBS) method is time consuming and operator dependent. We investigated the feasibility of a server-based, commercially available and ready-to use-artificial intelligence (AI) application based on convolutional neural network methods that integrate fully automatic view selection and measurement of LVEF from an entire Echo exam into a single workflow. We prospectively enrolled 1083 consecutive patients who had been referred to Echo for diagnostic or therapeutic purposes. LVEF was measured independently using MBS and AI. Test–retest variability was assessed in 40 patients. The reliability, repeatability, and time efficiency of LVEF measurements were compared between the two methods. Overall, 889 Echos were analyzed by cardiologists with the MBS method and by the AI. Over the study period of 10 weeks, the feasibility of both automatic view classification and seamlessly measured LVEF rose to 81% without user involvement. LVEF, LV end-diastolic and end-systolic volumes correlated strongly between MBS and AI (R = 0.87, 0.89 and 0.93, p < 0.001 for all) with a mean bias of +4.5% EF, −12 mL and −11 mL, respectively, due to impaired image quality and the extent of LV function. Repeatability and reliability of LVEF measurement (n = 40, test–retest) by AI was excellent compared to MBS (coefficient of variation: 3.2% vs. 5.9%), although the median analysis time of the AI was longer than that of the operator-dependent MBS method (258 s vs. 171 s). This AI has succeeded in identifying apical LV views and measuring EF in one workflow with comparable results to the MBS method and shows excellent reproducibility. It offers realistic perspectives for fully automated AI-based measurement of LVEF in routine clinical settings.
Collapse
|
14
|
Joshi K, Das M, Sarma A, Arora MK, SInghal M, Kumar B. Insight on Cardiac Chronobiology and Latest Developments of Chronotherapeutic Antihypertensive Interventions for Better Clinical Outcomes. Curr Hypertens Rev 2023; 19:106-122. [PMID: 36624649 DOI: 10.2174/1573402119666230109142156] [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: 04/23/2022] [Revised: 11/10/2022] [Accepted: 11/15/2022] [Indexed: 01/11/2023]
Abstract
Cardiac circadian rhythms are an important regulator of body functions, including cardiac activities and blood pressure. Disturbance of circadian rhythm is known to trigger and aggravate various cardiovascular diseases. Thus, modulating the circadian rhythm can be used as a therapeutic approach to cardiovascular diseases. Through this work, we intend to discuss the current understanding of cardiac circadian rhythms, in terms of quantifiable parameters like BP and HR. We also elaborate on the molecular regulators and the molecular cascades along with their specific genetic aspects involved in modulating circadian rhythms, with specific reference to cardiovascular health and cardiovascular diseases. Along with this, we also presented the latest pharmacogenomic and metabolomics markers involved in chronobiological control of the cardiovascular system along with their possible utility in cardiovascular disease diagnosis and therapeutics. Finally, we reviewed the current expert opinions on chronotherapeutic approaches for utilizing the conventional as well as the new pharmacological molecules for antihypertensive chronotherapy.
Collapse
Affiliation(s)
- Kumud Joshi
- Department of Pharmacy, Lloyd Institute of Management and Technology, Greater Noida, India
| | - Madhubanti Das
- Department of Zoology, Gauhati University, Guwahati, Assam, India
| | - Anupam Sarma
- Advanced Drug Delivery Laboratory, GIPS, Girijananda Chowdhury University, Guwahati, Assam, India
| | - Mandeep K Arora
- School of Pharmacy and population health informatics, DIT University, Dehradun, India
| | - Manmohan SInghal
- School of Pharmacy and population health informatics, DIT University, Dehradun, India
| | - Bhavna Kumar
- School of Pharmacy and population health informatics, DIT University, Dehradun, India
| |
Collapse
|
15
|
Yu Y, Li W, Xu L, Wang Y. Circadian rhythm of plasminogen activator inhibitor-1 and cardiovascular complications in type 2 diabetes. Front Endocrinol (Lausanne) 2023; 14:1124353. [PMID: 37020596 PMCID: PMC10067678 DOI: 10.3389/fendo.2023.1124353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/27/2023] [Indexed: 03/22/2023] Open
Abstract
Cardiovascular complications are a common death cause in type 2 diabetes patients, as they are often combined. Plasminogen-activator Inhibitor 1 (PAI-1) participates in the development and progression of cardiovascular complications in diabetes. Insulin resistance increases PAI-1 production, and high PAI-1 levels lead to an environment conducive to thrombosis and earlier and more severe vascular disease. Current evidence also suggests that PAI-1 has a rhythmic profile of circadian fluctuations and acrophase in the morning within a single day, which might explain the high morning incidence of cardiovascular events. Thus, PAI-1 is a possible drug target. Although several PAI-1 inhibitors have been developed, none have yet been allowed for clinical use. Research on rhythm has also led to the concept of "chronotherapy", a rhythm-based drug regimen expected to improve the treatment of cardiovascular complications in diabetic patients. Herein, we searched several databases and reviewed relevant articles to describe the circadian rhythm characteristics and endogenous molecular mechanisms of PAI-1, its relationship with insulin resistance, the causes of cardiovascular complications caused by PAI-1, and the current development of PAI-1 inhibitors. We also summarized the possibility of using the circadian rhythm of PAI-1 to treat cardiovascular complications in diabetic patients.
Collapse
|
16
|
Vinod M, Berthier A, Maréchal X, Gheeraert C, Boutry R, Delhaye S, Annicotte JS, Duez H, Hovasse A, Cianférani S, Montaigne D, Eeckhoute J, Staels B, Lefebvre P. Timed use of digoxin prevents heart ischemia-reperfusion injury through a REV-ERBα-UPS signaling pathway. NATURE CARDIOVASCULAR RESEARCH 2022; 1:990-1005. [PMID: 38229609 PMCID: PMC7615528 DOI: 10.1038/s44161-022-00148-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 09/16/2022] [Indexed: 01/18/2024]
Abstract
Myocardial ischemia-reperfusion injury (MIRI) induces life-threatening damages to the cardiac tissue and pharmacological means to achieve cardioprotection are sorely needed. MIRI severity varies along the day-night cycle and is molecularly linked to components of the cellular clock including the nuclear receptor REV-ERBα, a transcriptional repressor. Here we show that digoxin administration in mice is cardioprotective when timed to trigger REV-ERBα protein degradation. In cardiomyocytes, digoxin increases REV-ERBα ubiquitinylation and proteasomal degradation, which depend on REV-ERBα ability to bind its natural ligand, heme. Inhibition of the membrane-bound Src tyrosine-kinase partially alleviated digoxin-induced REV-ERBα degradation. In untreated cardiomyocytes, REV-ERBα proteolysis is controlled by known (HUWE1, FBXW7, SIAH2) or novel (CBL, UBE4B) E3 ubiquitin ligases and the proteasome subunit PSMB5. Only SIAH2 and PSMB5 contributed to digoxin-induced degradation of REV-ERBα. Thus, controlling REV-ERBα proteostasis through the ubiquitin-proteasome system is an appealing cardioprotective strategy. Our data support the timed use of clinically-approved cardiotonic steroids in prophylactic cardioprotection.
Collapse
Affiliation(s)
- Manjula Vinod
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Alexandre Berthier
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Xavier Maréchal
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Céline Gheeraert
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Raphaёl Boutry
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 – RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000 Lille, France
| | - Stéphane Delhaye
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Jean-Sébastien Annicotte
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 – RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000 Lille, France
| | - Hélène Duez
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Agnès Hovasse
- Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), IPHC, Université de Strasbourg, CNRS, UMR7178, 25 Rue Becquerel, F-67087 Strasbourg, France
| | - Sarah Cianférani
- Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), IPHC, Université de Strasbourg, CNRS, UMR7178, 25 Rue Becquerel, F-67087 Strasbourg, France
| | - David Montaigne
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Jérôme Eeckhoute
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Bart Staels
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Philippe Lefebvre
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| |
Collapse
|
17
|
Krylov V, Machikhin A, Sizov D, Guryleva A, Sizova A, Zhdanova S, Tchougounov V, Burlakov A. Influence of hypomagnetic field on the heartbeat in zebrafish embryos. Front Physiol 2022; 13:1040083. [PMID: 36338501 PMCID: PMC9634549 DOI: 10.3389/fphys.2022.1040083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 10/12/2022] [Indexed: 12/04/2022] Open
Abstract
The magnetic environment may influence the functioning of the cardiovascular system. It was reported that low-frequency and static magnetic fields affect hemodynamics, heart rate, and heart rate variability in animals and humans. Moreover, recent data suggest that magnetic fields affect the circadian rhythms of physiological processes. The influence of the magnetic environment on heart functionating during early development has been studied insufficiently. We utilized transparent zebrafish embryos to evaluate the effect of the hypomagnetic field on the characteristics of cardiac function using a noninvasive optical approach based on photoplethysmographic microscopic imaging. The embryos were exposed to the geomagnetic and hypomagnetic fields from the second to the 116th hour post fertilization under a 16 h light/8 h dark cycle or constant illumination. The exposure of embryos to the hypomagnetic field in both lighting modes led to increased embryo mortality, the appearance of abnormal phenotypes, and a significant increase in the embryo’s heartbeat rate. The difference between maximal and minimal heartbeat intervals, maximal to minimal heartbeat intervals ratio, and the coefficient of variation of heartbeat rate were increased in the embryos exposed to the hypomagnetic field under constant illumination from 96 to 116 h post fertilization. The dynamics of heartbeat rate changes followed a circadian pattern in all studied groups except zebrafish exposed to the hypomagnetic field under constant illumination. The results demonstrate the importance of natural magnetic background for the early development of zebrafish. The possible mechanisms of observed effects are discussed.
Collapse
Affiliation(s)
- Viacheslav Krylov
- Scientific and Technological Center of Unique Instrumentation, Russian Academy of Sciences, Moscow, Russia
- Papanin Institute for Biology of Inland Waters, Russian Academy of Sciences, Borok, Russia
- *Correspondence: Viacheslav Krylov,
| | - Alexander Machikhin
- Scientific and Technological Center of Unique Instrumentation, Russian Academy of Sciences, Moscow, Russia
| | - Daniil Sizov
- Papanin Institute for Biology of Inland Waters, Russian Academy of Sciences, Borok, Russia
| | - Anastasia Guryleva
- Scientific and Technological Center of Unique Instrumentation, Russian Academy of Sciences, Moscow, Russia
| | - Anastasia Sizova
- Scientific and Technological Center of Unique Instrumentation, Russian Academy of Sciences, Moscow, Russia
- Papanin Institute for Biology of Inland Waters, Russian Academy of Sciences, Borok, Russia
| | - Svetlana Zhdanova
- Papanin Institute for Biology of Inland Waters, Russian Academy of Sciences, Borok, Russia
| | - Vladimir Tchougounov
- Papanin Institute for Biology of Inland Waters, Russian Academy of Sciences, Borok, Russia
| | - Alexander Burlakov
- Scientific and Technological Center of Unique Instrumentation, Russian Academy of Sciences, Moscow, Russia
- Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
18
|
Shirato K, Sato S. Macrophage Meets the Circadian Clock: Implication of the Circadian Clock in the Role of Macrophages in Acute Lower Respiratory Tract Infection. Front Cell Infect Microbiol 2022; 12:826738. [PMID: 35281442 PMCID: PMC8904936 DOI: 10.3389/fcimb.2022.826738] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/01/2022] [Indexed: 12/13/2022] Open
Abstract
The circadian rhythm is a biological system that creates daily variations of physiology and behavior with a 24-h cycle, which is precisely controlled by the molecular circadian clock. The circadian clock dominates temporal activity of physiological homeostasis at the molecular level, including endocrine secretion, metabolic, immune response, coupled with extrinsic environmental cues (e.g., light/dark cycles) and behavioral cues (e.g., sleep/wake cycles and feeding/fasting cycles). The other side of the clock is that the misaligned circadian rhythm contributes to the onset of a variety of diseases, such as cancer, metabolic diseases, and cardiovascular diseases, the acceleration of aging, and the development of systemic inflammation. The role played by macrophages is a key mediator between circadian disruption and systemic inflammation. At the molecular level, macrophage functions are under the direct control of the circadian clock, and thus the circadian misalignment remodels the phenotype of macrophages toward a ‘killer’ mode. Remarkably, the inflammatory macrophages induce systemic and chronic inflammation, leading to the development of inflammatory diseases and the dampened immune defensive machinery against infectious diseases such as COVID-19. Here, we discuss how the circadian clock regulates macrophage immune functions and provide the potential risk of misaligned circadian rhythms against inflammatory and infectious diseases.
Collapse
Affiliation(s)
- Ken Shirato
- Department of Molecular Predictive Medicine and Sport Science, Kyorin University School of Medicine, Mitaka, Japan
| | - Shogo Sato
- Center for Biological Clocks Research, Department of Biology, Texas A&M University, College Station, TX, United States
- *Correspondence: Shogo Sato,
| |
Collapse
|
19
|
Austad SN, Ballinger S, Buford TW, Carter CS, Smith DL, Darley-Usmar V, Zhang J. Targeting whole body metabolism and mitochondrial bioenergetics in the drug development for Alzheimer's disease. Acta Pharm Sin B 2022; 12:511-531. [PMID: 35256932 PMCID: PMC8897048 DOI: 10.1016/j.apsb.2021.06.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/26/2021] [Accepted: 06/16/2021] [Indexed: 02/07/2023] Open
Abstract
Aging is by far the most prominent risk factor for Alzheimer's disease (AD), and both aging and AD are associated with apparent metabolic alterations. As developing effective therapeutic interventions to treat AD is clearly in urgent need, the impact of modulating whole-body and intracellular metabolism in preclinical models and in human patients, on disease pathogenesis, have been explored. There is also an increasing awareness of differential risk and potential targeting strategies related to biological sex, microbiome, and circadian regulation. As a major part of intracellular metabolism, mitochondrial bioenergetics, mitochondrial quality-control mechanisms, and mitochondria-linked inflammatory responses have been considered for AD therapeutic interventions. This review summarizes and highlights these efforts.
Collapse
Key Words
- ACE2, angiotensin I converting enzyme (peptidyl-dipeptidase A) 2
- AD, Alzheimer's disease
- ADP, adenosine diphosphate
- ADRD, AD-related dementias
- Aβ, amyloid β
- CSF, cerebrospinal fluid
- Circadian regulation
- DAMPs
- DAMPs, damage-associated molecular patterns
- Diabetes
- ER, estrogen receptor
- ETC, electron transport chain
- FCCP, trifluoromethoxy carbonylcyanide phenylhydrazone
- FPR-1, formyl peptide receptor 1
- GIP, glucose-dependent insulinotropic polypeptide
- GLP-1, glucagon-like peptide-1
- HBP, hexoamine biosynthesis pathway
- HTRA, high temperature requirement A
- Hexokinase biosynthesis pathway
- I3A, indole-3-carboxaldehyde
- IRF-3, interferon regulatory factor 3
- LC3, microtubule associated protein light chain 3
- LPS, lipopolysaccharide
- LRR, leucine-rich repeat
- MAVS, mitochondrial anti-viral signaling
- MCI, mild cognitive impairment
- MRI, magnetic resonance imaging
- MRS, magnetic resonance spectroscopy
- Mdivi-1, mitochondrial division inhibitor 1
- Microbiome
- Mitochondrial DNA
- Mitochondrial electron transport chain
- Mitochondrial quality control
- NLRP3, leucine-rich repeat (LRR)-containing protein (NLR)-like receptor family pyrin domain containing 3
- NOD, nucleotide-binding oligomerization domain
- NeuN, neuronal nuclear protein
- PET, fluorodeoxyglucose (FDG)-positron emission tomography
- PKA, protein kinase A
- POLβ, the base-excision repair enzyme DNA polymerase β
- ROS, reactive oxygen species
- Reactive species
- SAMP8, senescence-accelerated mice
- SCFAs, short-chain fatty acids
- SIRT3, NAD-dependent deacetylase sirtuin-3
- STING, stimulator of interferon genes
- STZ, streptozotocin
- SkQ1, plastoquinonyldecyltriphenylphosphonium
- T2D, type 2 diabetes
- TCA, Tricarboxylic acid
- TLR9, toll-like receptor 9
- TMAO, trimethylamine N-oxide
- TP, tricyclic pyrone
- TRF, time-restricted feeding
- cAMP, cyclic adenosine monophosphate
- cGAS, cyclic GMP/AMP synthase
- hAPP, human amyloid precursor protein
- hPREP, human presequence protease
- i.p., intraperitoneal
- mTOR, mechanistic target of rapamycin
- mtDNA, mitochondrial DNA
- αkG, alpha-ketoglutarate
Collapse
Affiliation(s)
- Steven N. Austad
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Scott Ballinger
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Thomas W. Buford
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Christy S. Carter
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Daniel L. Smith
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Victor Darley-Usmar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jianhua Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
20
|
Li MD, Xin H, Yuan Y, Yang X, Li H, Tian D, Zhang H, Zhang Z, Han TL, Chen Q, Duan G, Ju D, Chen K, Deng F, He W. Circadian Clock-Controlled Checkpoints in the Pathogenesis of Complex Disease. Front Genet 2021; 12:721231. [PMID: 34557221 PMCID: PMC8452875 DOI: 10.3389/fgene.2021.721231] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 08/16/2021] [Indexed: 12/26/2022] Open
Abstract
The circadian clock coordinates physiology, metabolism, and behavior with the 24-h cycles of environmental light. Fundamental mechanisms of how the circadian clock regulates organ physiology and metabolism have been elucidated at a rapid speed in the past two decades. Here we review circadian networks in more than six organ systems associated with complex disease, which cluster around metabolic disorders, and seek to propose critical regulatory molecules controlled by the circadian clock (named clock-controlled checkpoints) in the pathogenesis of complex disease. These include clock-controlled checkpoints such as circadian nuclear receptors in liver and muscle tissues, chemokines and adhesion molecules in the vasculature. Although the progress is encouraging, many gaps in the mechanisms remain unaddressed. Future studies should focus on devising time-dependent strategies for drug delivery and engagement in well-characterized organs such as the liver, and elucidating fundamental circadian biology in so far less characterized organ systems, including the heart, blood, peripheral neurons, and reproductive systems.
Collapse
Affiliation(s)
- Min-Dian Li
- Department of Cardiology and the Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Haoran Xin
- Department of Cardiology and the Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yinglin Yuan
- Medical Center of Hematology, The Xinqiao Hospital of Army Medical University, Chongqing, China
| | - Xinqing Yang
- Department of Anesthesiology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hongli Li
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dingyuan Tian
- Department of Cardiology and the Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hua Zhang
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhihui Zhang
- Department of Cardiology and the Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Ting-Li Han
- Department of Obstetrics and Gynaecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qing Chen
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Guangyou Duan
- Department of Anesthesiology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Dapeng Ju
- Department of Anesthesiology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Ka Chen
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Army Medical University, Chongqing, China
| | - Fang Deng
- Key Laboratory of Extreme Environmental Medicine, Department of Pathophysiology, College of High Altitude Military Medicine, Ministry of Education of China, Army Medical University (Third Military Medical University), Chongqing, China.,Key Laboratory of High Altitude Medicine, PLA, Army Medical University (Third Military Medical University), Chongqing, China
| | - Wenyan He
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | | |
Collapse
|
21
|
Abstract
Circadian rhythm evolved to allow organisms to coordinate intrinsic physiological functions in anticipation of recurring environmental changes. The importance of this coordination is exemplified by the tight temporal control of cardiac metabolism. Levels of metabolites, metabolic flux, and response to nutrients all oscillate in a time-of-day-dependent fashion. While these rhythms are affected by oscillatory behavior (feeding/fasting, wake/sleep) and neurohormonal changes, recent data have unequivocally demonstrated an intrinsic circadian regulation at the tissue and cellular level. The circadian clock - through a network of a core clock, slave clock, and effectors - exerts intricate temporal control of cardiac metabolism, which is also integrated with environmental cues. The combined anticipation and adaptability that the circadian clock enables provide maximum advantage to cardiac function. Disruption of the circadian rhythm, or dyssynchrony, leads to cardiometabolic disorders seen not only in shift workers but in most individuals in modern society. In this Review, we describe current findings on rhythmic cardiac metabolism and discuss the intricate regulation of circadian rhythm and the consequences of rhythm disruption. An in-depth understanding of the circadian biology in cardiac metabolism is critical in translating preclinical findings from nocturnal-animal models as well as in developing novel chronotherapeutic strategies.
Collapse
Affiliation(s)
- Lilei Zhang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Mukesh K Jain
- Case Cardiovascular Research Institute, Department of Medicine.,Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, and.,School of Medicine; Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
22
|
Hayter EA, Wehrens SMT, Van Dongen HPA, Stangherlin A, Gaddameedhi S, Crooks E, Barron NJ, Venetucci LA, O'Neill JS, Brown TM, Skene DJ, Trafford AW, Bechtold DA. Distinct circadian mechanisms govern cardiac rhythms and susceptibility to arrhythmia. Nat Commun 2021; 12:2472. [PMID: 33931651 PMCID: PMC8087694 DOI: 10.1038/s41467-021-22788-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 03/26/2021] [Indexed: 02/02/2023] Open
Abstract
Electrical activity in the heart exhibits 24-hour rhythmicity, and potentially fatal arrhythmias are more likely to occur at specific times of day. Here, we demonstrate that circadian clocks within the brain and heart set daily rhythms in sinoatrial (SA) and atrioventricular (AV) node activity, and impose a time-of-day dependent susceptibility to ventricular arrhythmia. Critically, the balance of circadian inputs from the autonomic nervous system and cardiomyocyte clock to the SA and AV nodes differ, and this renders the cardiac conduction system sensitive to decoupling during abrupt shifts in behavioural routine and sleep-wake timing. Our findings reveal a functional segregation of circadian control across the heart's conduction system and inherent susceptibility to arrhythmia.
Collapse
Affiliation(s)
- Edward A Hayter
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Sophie M T Wehrens
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Hans P A Van Dongen
- Sleep and Performance Research Center, Washington State University, Spokane, WA, USA
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | | | - Shobhan Gaddameedhi
- Department of Biological Sciences, Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA
| | - Elena Crooks
- Sleep and Performance Research Center, Washington State University, Spokane, WA, USA
- Department of Physical Therapy, Eastern Washington University, Spokane, WA, USA
| | - Nichola J Barron
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Luigi A Venetucci
- Unit of Clinical Physiology, Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | | | - Timothy M Brown
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Debra J Skene
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Andrew W Trafford
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Unit of Clinical Physiology, Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - David A Bechtold
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
23
|
Khannous-Lleiffe O, Willis JR, Saus E, Cabrera-Aguilera I, Almendros I, Farré R, Gozal D, Farré N, Gabaldón T. A Mouse Model Suggests That Heart Failure and Its Common Comorbidity Sleep Fragmentation Have No Synergistic Impacts on the Gut Microbiome. Microorganisms 2021; 9:microorganisms9030641. [PMID: 33808770 PMCID: PMC8003359 DOI: 10.3390/microorganisms9030641] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/11/2021] [Accepted: 03/15/2021] [Indexed: 12/19/2022] Open
Abstract
Heart failure (HF) is a common condition associated with a high rate of hospitalizations and adverse outcomes. HF is characterized by impairments of either the cardiac ventricular filling, ejection of blood capacity or both. Sleep fragmentation (SF) involves a series of short sleep interruptions that lead to fatigue and contribute to cognitive impairments and dementia. Both conditions are known to be associated with increased inflammation and dysbiosis of the gut microbiota. In the present study, mice were distributed into four groups, and subjected for four weeks to either HF, SF, both HF and SF, or left unperturbed as controls. We used 16S metabarcoding to assess fecal microbiome composition before and after the experiments. Evidence for distinct alterations in several bacterial groups and an overall decrease in alpha diversity emerged in HF and SF treatment groups. Combined HF and SF conditions, however, showed no synergism, and observed changes were not always additive, suggesting preliminarily that some of the individual effects of either HF or SF cancel each other out when applied concomitantly.
Collapse
Affiliation(s)
- Olfat Khannous-Lleiffe
- Barcelona Supercomputing Centre (BSC-CNS), 08034 Barcelona, Spain; (O.K.-L.); (J.R.W.); (E.S.)
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Jesse R. Willis
- Barcelona Supercomputing Centre (BSC-CNS), 08034 Barcelona, Spain; (O.K.-L.); (J.R.W.); (E.S.)
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Ester Saus
- Barcelona Supercomputing Centre (BSC-CNS), 08034 Barcelona, Spain; (O.K.-L.); (J.R.W.); (E.S.)
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Ignacio Cabrera-Aguilera
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain; (I.C.-A.); (I.A.); (R.F.)
- Department of Human Movement Sciences, Faculty of Health Sciences, School of Kinesiology, Universidad de Talca, Talca 3460000, Chile
| | - Isaac Almendros
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain; (I.C.-A.); (I.A.); (R.F.)
- CIBER de Enfermedades Respiratorias, 28029 Madrid, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, 08036 Barcelona, Spain
| | - Ramon Farré
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain; (I.C.-A.); (I.A.); (R.F.)
- CIBER de Enfermedades Respiratorias, 28029 Madrid, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, 08036 Barcelona, Spain
| | - David Gozal
- Department of Child Health and Child Health Research Institute, The University of Missouri School of Medicine, Columbia, MO 65212, USA;
| | - Nuria Farré
- Heart Failure Unit, Department of Cardiology, Hospital del Mar (Parc de Salut Mar), 08003 Barcelona, Spain
- Heart Diseases Biomedical Research Group, IMIM (Hospital del Mar Medical Research Institute), 08003 Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
- Correspondence: (N.F.); (T.G.)
| | - Toni Gabaldón
- Barcelona Supercomputing Centre (BSC-CNS), 08034 Barcelona, Spain; (O.K.-L.); (J.R.W.); (E.S.)
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
- Correspondence: (N.F.); (T.G.)
| |
Collapse
|
24
|
Xin H, Deng F, Zhou M, Huang R, Ma X, Tian H, Tan Y, Chen X, Deng D, Shui G, Zhang Z, Li MD. A multi-tissue multi-omics analysis reveals distinct kineztics in entrainment of diurnal transcriptomes by inverted feeding. iScience 2021; 24:102335. [PMID: 33889826 PMCID: PMC8050734 DOI: 10.1016/j.isci.2021.102335] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/26/2021] [Accepted: 03/16/2021] [Indexed: 02/07/2023] Open
Abstract
Time of eating synchronizes circadian rhythms of metabolism and physiology. Inverted feeding can uncouple peripheral circadian clocks from the central clock located in the suprachiasmatic nucleus. However, system-wide changes of circadian metabolism and physiology entrained to inverted feeding in peripheral tissues remain largely unexplored. Here, we performed a 24-h global profiling of transcripts and metabolites in mouse peripheral tissues to study the transition kinetics during inverted feeding, and revealed distinct kinetics in phase entrainment of diurnal transcriptomes by inverted feeding, which graded from fat tissue (near-completely entrained), liver, kidney, to heart. Phase kinetics of tissue clocks tracked with those of transcriptomes and were gated by light-related cues. Integrated analysis of transcripts and metabolites demonstrated that fatty acid oxidation entrained completely to inverted feeding in heart despite the slow kinetics/resistance of the heart clock to entrainment by feeding. This multi-omics resource defines circadian signatures of inverted feeding in peripheral tissues (www.CircaMetDB.org.cn). A multi-omics analysis of food entrainment in mouse peripheral tissues Inverted feeding rhythm entrains diurnal transcriptomes with distinct kinetics Phase kinetics of tissue clocks is conditioned by constant light Cardiac metabolism entrains to feeding fast with slow kinetics of the heart clock
Collapse
Affiliation(s)
- Haoran Xin
- Department of Cardiology and the Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Fang Deng
- Department of Pathophysiology, College of High Altitude Military Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Meiyu Zhou
- Department of Cardiology and the Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Rongfeng Huang
- Department of Cardiology and the Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xiaogen Ma
- Department of Cardiology and the Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - He Tian
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yan Tan
- Department of Pathophysiology, College of High Altitude Military Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xinghua Chen
- Department of Cardiology and the Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Dan Deng
- Department of Cardiology and the Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zhihui Zhang
- Department of Cardiology and the Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Min-Dian Li
- Department of Cardiology and the Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| |
Collapse
|
25
|
Fong CK, Hearn T, Webb SE, Frøland Steindal IA, Miller AL, Whitmore D. Daily rhythms in heartbeat rate are intrinsic to the zebrafish heart. Curr Biol 2021; 31:R239-R240. [PMID: 33689719 DOI: 10.1016/j.cub.2021.01.106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
It is a well-established fact that different tissues within the body contain their own circadian clocks or pacemakers, where it is proposed that the clock controls the local, daily cell biology of that organ.1,2 In mammals, these peripheral clocks work in concert with and are entrained by rhythmic signals arising from the suprachiasmatic nucleus (SCN) in the hypothalamus of the animal, among other systemic cues.2 In the case of zebrafish, the circadian system appears to be highly decentralized with each tissue not only having an internal circadian clock, but also being directly light entrained.1 Several years ago, we showed that the zebrafish heart contains its own circadian pacemaker at the gene expression level.1 This is also the case in mammals, where the circadian clock controls approximately 10% of the genes expressed in the heart.3 However, heart rate itself is generally thought to be regulated by several well-described autonomic cues, neurotransmitters, and hormones. In this study, we report that, for larval zebrafish hearts, the daily change in heartbeat rate is not only clock-controlled in vivo, but that this rhythm also persists in vitro, indicating that the cardiac circadian clock itself can directly drive this major physiological oscillation.
Collapse
Affiliation(s)
- Chun Ki Fong
- Division of Life Science and State Key Laboratory for Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Tim Hearn
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Sarah E Webb
- Division of Life Science and State Key Laboratory for Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | | | - Andrew L Miller
- Division of Life Science and State Key Laboratory for Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.
| | - David Whitmore
- Department of Cell and Developmental Biology, University College London, London, UK; College of Public Health, Medical and Veterinary Sciences, Department of Molecular and Cell Biology, James Cook University, Townsville, QLD 4811, Australia.
| |
Collapse
|
26
|
Zhang H, Dai J, Tian D, Xiao L, Xue H, Guo Q, Zhang X, Teng X, Jin S, Wu Y. Hydrogen Sulfide Restored the Diurnal Variation in Cardiac Function of Aging Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8841575. [PMID: 33747351 PMCID: PMC7943277 DOI: 10.1155/2021/8841575] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 02/06/2021] [Accepted: 02/12/2021] [Indexed: 12/21/2022]
Abstract
The present study was performed to investigate whether H2S could restore the diurnal variation in cardiac function of aging mice and explore the potential mechanisms. We found that ejection fraction (EF) and fractional shortening (FS) in 3-month-old mice exhibited diurnal variations over a 24-hour period. However, the diurnal variations were disrupted in 18-month-old mice, and there was a decline in EF and FS. In addition, the plasma malondialdehyde (MDA) levels were increased, and H2S concentrations and superoxide dismutase (SOD) activities were decreased in 18-month-old mice. Then, CSE KO mice were used to determine if there was a relationship between endogenous H2S and diurnal variations in EF and FS. There was no difference in 12-hour averaged EF and FS between dark and light periods in CSE KO mice accompanying increased MDA levels and decreased SOD activities in plasma, indicating that deficiency of endogenous H2S blunted diurnal variations of cardiac function. To determine whether oxidative stress disrupted the diurnal variations in cardiac function, D-galactose-induced subacute aging mice were employed. After 3-month D-gal treatment, both 12-hour averaged EF and FS in dark or light periods were decreased; meanwhile, there was no difference in 12-hour averaged EF and FS between dark and light periods. After 3-month NaHS treatment in the D-gal group, the plasma MDA levels were decreased and SOD activities were increased. The EF and FS were lower during the 12-hour light period than those during the 12-hour dark period which was fit to sine curves in the D-gal+NaHS group. Identical findings were also observed in 18-month-old mice. In conclusion, our studies revealed that the disrupted diurnal variation in cardiac function was associated with increased oxidative stress and decreased H2S levels in aging mice. H2S could restore the diurnal variation in cardiac function of aging mice by reducing oxidative stress.
Collapse
Affiliation(s)
- Huaxing Zhang
- School of Basic Medical Sciences, Hebei Medical University, Hebei 050017, China
| | - Jing Dai
- Department of Clinical Diagnostics, Hebei Medical University, Hebei 050017, China
| | - Danyang Tian
- Department of Physiology, Hebei Medical University, Hebei 050017, China
| | - Lin Xiao
- Department of Physiology, Hebei Medical University, Hebei 050017, China
| | - Hongmei Xue
- Department of Physiology, Hebei Medical University, Hebei 050017, China
| | - Qi Guo
- Department of Physiology, Hebei Medical University, Hebei 050017, China
| | - Xiangjian Zhang
- Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, 050017 Hebei, China
| | - Xu Teng
- Department of Physiology, Hebei Medical University, Hebei 050017, China
| | - Sheng Jin
- Department of Physiology, Hebei Medical University, Hebei 050017, China
| | - Yuming Wu
- Department of Physiology, Hebei Medical University, Hebei 050017, China
- Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, 050017 Hebei, China
| |
Collapse
|
27
|
Diekman CO, Wei N. Circadian Rhythms of Early Afterdepolarizations and Ventricular Arrhythmias in a Cardiomyocyte Model. Biophys J 2020; 120:319-333. [PMID: 33285114 DOI: 10.1016/j.bpj.2020.11.2264] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/25/2020] [Accepted: 11/10/2020] [Indexed: 11/30/2022] Open
Abstract
Sudden cardiac arrest is a malfunction of the heart's electrical system, typically caused by ventricular arrhythmias, that can lead to sudden cardiac death (SCD) within minutes. Epidemiological studies have shown that SCD and ventricular arrhythmias are more likely to occur in the morning than in the evening, and laboratory studies indicate that these daily rhythms in adverse cardiovascular events are at least partially under the control of the endogenous circadian timekeeping system. However, the biophysical mechanisms linking molecular circadian clocks to cardiac arrhythmogenesis are not fully understood. Recent experiments have shown that L-type calcium channels exhibit circadian rhythms in both expression and function in guinea pig ventricular cardiomyocytes. We developed an electrophysiological model of these cells to simulate the effect of circadian variation in L-type calcium conductance. In our simulations, we found that there is a circadian pattern in the occurrence of early afterdepolarizations (EADs), which are abnormal depolarizations during the repolarization phase of a cardiac action potential that can trigger fatal ventricular arrhythmias. Specifically, the model produces EADs in the morning, but not at other times of day. We show that the model exhibits a codimension-2 Takens-Bogdanov bifurcation that serves as an organizing center for different types of EAD dynamics. We also simulated a two-dimensional spatial version of this model across a circadian cycle. We found that there is a circadian pattern in the breakup of spiral waves, which represents ventricular fibrillation in cardiac tissue. Specifically, the model produces spiral wave breakup in the morning, but not in the evening. Our computational study is the first, to our knowledge, to propose a link between circadian rhythms and EAD formation and suggests that the efficacy of drugs targeting EAD-mediated arrhythmias may depend on the time of day that they are administered.
Collapse
Affiliation(s)
- Casey O Diekman
- Department of Mathematical Sciences, New Jersey Institute of Technology, Newark, New Jersey; EPSRC Centre for Predictive Modelling in Healthcare, Living Systems Institute, University of Exeter, Exeter, United Kingdom.
| | - Ning Wei
- Department of Mathematics, Purdue University, West Lafayette, Indiana
| |
Collapse
|
28
|
Xu W, Jain MK, Zhang L. Molecular link between circadian clocks and cardiac function: a network of core clock, slave clock, and effectors. Curr Opin Pharmacol 2020; 57:28-40. [PMID: 33189913 DOI: 10.1016/j.coph.2020.10.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/27/2020] [Accepted: 10/06/2020] [Indexed: 02/08/2023]
Abstract
The circadian rhythm has a strong influence on both cardiac physiology and disease in humans. Preclinical studies primarily using tissue-specific transgenic mouse models have contributed to our understanding of the molecular mechanism of the circadian clock in the cardiovascular system. The core clock driven by CLOCK:BMAL1 complex functions as a universal timing machinery that primarily sets the pace in all mammalian cell types. In one specific cell or tissue type, core clock may control a secondary transcriptional oscillator, conceptualized as slave clock, which confers the oscillatory expression of tissue-specific effectors. Here, we discuss a core clock-slave clock-effectors network, which links the molecular clock to cardiac function.
Collapse
Affiliation(s)
- Weiyi Xu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mukesh K Jain
- Case Cardiovascular Research Institute, Department of Medicine, Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, USA; School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Lilei Zhang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
29
|
Mia S, Kane MS, Latimer MN, Reitz CJ, Sonkar R, Benavides GA, Smith SR, Frank SJ, Martino TA, Zhang J, Darley-Usmar VM, Young ME. Differential effects of REV-ERBα/β agonism on cardiac gene expression, metabolism, and contractile function in a mouse model of circadian disruption. Am J Physiol Heart Circ Physiol 2020; 318:H1487-H1508. [PMID: 32357113 PMCID: PMC7311693 DOI: 10.1152/ajpheart.00709.2019] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Cell-autonomous circadian clocks have emerged as temporal orchestrators of numerous biological processes. For example, the cardiomyocyte circadian clock modulates transcription, translation, posttranslational modifications, ion homeostasis, signaling cascades, metabolism, and contractility of the heart over the course of the day. Circadian clocks are composed of more than 10 interconnected transcriptional modulators, all of which have the potential to influence the cardiac transcriptome (and ultimately cardiac processes). These transcriptional modulators include BMAL1 and REV-ERBα/β; BMAL1 induces REV-ERBα/β, which in turn feeds back to inhibit BMAL1. Previous studies indicate that cardiomyocyte-specific BMAL1-knockout (CBK) mice exhibit a dysfunctional circadian clock (including decreased REV-ERBα/β expression) in the heart associated with abnormalities in cardiac mitochondrial function, metabolism, signaling, and contractile function. Here, we hypothesized that decreased REV-ERBα/β activity is responsible for distinct phenotypical alterations observed in CBK hearts. To test this hypothesis, CBK (and littermate control) mice were administered with the selective REV-ERBα/β agonist SR-9009 (100 mg·kg-1·day-1 for 8 days). SR-9009 administration was sufficient to normalize cardiac glycogen synthesis rates, cardiomyocyte size, interstitial fibrosis, and contractility in CBK hearts (without influencing mitochondrial complex activities, nor normalizing substrate oxidation and Akt/mTOR/GSK3β signaling). Collectively, these observations highlight a role for REV-ERBα/β as a mediator of a subset of circadian clock-controlled processes in the heart.
Collapse
Affiliation(s)
- Sobuj Mia
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Mariame S Kane
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama.,Division of Molecular Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Mary N Latimer
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Cristine J Reitz
- Centre for Cardiovascular Investigations, Department of Biomedical Science, University of Guelph, Guelph, Ontario, Canada
| | - Ravi Sonkar
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Gloria A Benavides
- Division of Molecular Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Samuel R Smith
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Stuart J Frank
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama.,Endocrinology Section, Birmingham Veterans Affairs Medical Center Medical Service, Birmingham, Alabama
| | - Tami A Martino
- Centre for Cardiovascular Investigations, Department of Biomedical Science, University of Guelph, Guelph, Ontario, Canada
| | - Jianhua Zhang
- Division of Molecular Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Victor M Darley-Usmar
- Division of Molecular Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Martin E Young
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
30
|
Shen W, Zhang W, Ye W, Wang H, Zhang Q, Shen J, Hong Q, Li X, Wen G, Wei T, Zhang J. SR9009 induces a REV-ERB dependent anti-small-cell lung cancer effect through inhibition of autophagy. Theranostics 2020; 10:4466-4480. [PMID: 32292508 PMCID: PMC7150483 DOI: 10.7150/thno.42478] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/25/2020] [Indexed: 12/13/2022] Open
Abstract
Rationale: The circadian clock coordinates cell proliferation and metabolism and impacts the progression of some diseases, particularly cancer. Pharmacological modulation of the circadian machinery may be an effective therapeutic approach for treating cancer. SR9009 is a specific synthetic agonist of the REV-ERBs, essential circadian clock components. However, the potential efficacy and antitumor mechanism of this drug in small-cell lung cancer (SCLC) remains poorly understood. Methods: Here, we used chemosensitive cells (H69 and H446) and the corresponding chemoresistant cells (H69AR and H446DDP) to assess the efficacy of the REV-ERB agonist SR9009 for the treatment of SCLC in vitro and further validated the antitumor effect in subcutaneous tumor models of SCLC. Then, we determined whether REV-ERBα was correlated with the anti-SCLC effect of SR9009. Chromatin immunoprecipitation (ChIP) sequencing assays were conducted to identify potential DNA sequences directly regulated by REV-ERBα. Autophagy regulation by REV-ERBα and its possible mechanism in SR9009-based SCLC therapy were analyzed. Results: Here, we showed that the REV-ERB agonist SR9009 is specifically lethal to both chemosensitive and chemoresistant SCLC cells. REV-ERBα was involved in the antitumor effect of SR9009 in SCLC. The core autophagy gene Atg5 was identified as a direct downstream target of REV-ERBα and was suppressed by the REV-ERB agonist SR9009 in SCLC. Furthermore, the interaction of REV-ERBα with this autophagy gene impaired autophagy activity, leading to SR9009 cytotoxicity in SCLC cells. Principal conclusions: Our study provided a novel viewpoint indicating that the REV-ERB agonist SR9009 could be a novel and promising therapeutic strategy in first- or second-line SCLC treatment. The anti-SCLC effect of SR9009 is mediated by REV-ERB dependent suppression of autophagy via direct repression of the autophagy gene Atg5.
Collapse
|