1
|
Putthanbut N, Su PAB, Lee JY, Borlongan CV. Circadian rhythms in stem cells and their therapeutic potential. Stem Cell Res Ther 2025; 16:85. [PMID: 39988679 PMCID: PMC11849187 DOI: 10.1186/s13287-025-04178-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 01/23/2025] [Indexed: 02/25/2025] Open
Abstract
Circadian rhythms are present in almost all cells, but their existence in stem cells has remains not well established. Circadian clock appears to be closely associated with differentiated mature cells and rarely detected in immature embryonic stem cells. Recent evidence reveals the presence of circadian genes and rhythmic physiologic activities in stem cells as well as stem cell-derived extracellular vesicle (EV) characteristics. The circadian clock entails diverse physiologic and pathological mechanisms underlying cell fate. Integration of circadian rhythm to clinical applications, such as chronotherapy, chrono-biomarker, and environment modification, may facilitate therapeutic outcomes of stem cell-based regenerative medicine. Understanding circadian rhythms in stem cells can optimize stem cell-based therapies by determining the best times for harvesting and administering stem cells, thereby enhancing therapeutic efficacy. Further research into the circadian properties of stem cells will refine stem cell-based therapies, contributing to advancements in regenerative medicine.
Collapse
Affiliation(s)
- Napasiri Putthanbut
- Center of Aging and Brain Repair, Department of Neurosurgery, University of South Florida, Tampa, USA
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Salaya, Thailand
| | - Paul Alexis Bourgade Su
- Center of Aging and Brain Repair, Department of Neurosurgery, University of South Florida, Tampa, USA
- Centro de Investigación en Ciencias de La Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, Naucalpan, Mexico
| | - Jea-Young Lee
- Center of Aging and Brain Repair, Department of Neurosurgery, University of South Florida, Tampa, USA
| | - Cesario V Borlongan
- Center of Aging and Brain Repair, Department of Neurosurgery, University of South Florida, Tampa, USA.
| |
Collapse
|
2
|
Zhu (朱培) P, Chao CL, Steffeck AWT, Dang C, Hamlish NX, Pfrender EM, Jiang B, Peek CB. Circadian Dysfunction in the Skeletal Muscle Impairs Limb Perfusion and Muscle Regeneration in Peripheral Artery Disease. Arterioscler Thromb Vasc Biol 2025; 45:e30-e47. [PMID: 39633575 PMCID: PMC11753941 DOI: 10.1161/atvbaha.124.321772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Peripheral artery disease (PAD), caused by atherosclerosis, leads to limb ischemia, muscle damage, and impaired mobility in the lower extremities. Recent studies suggest that circadian rhythm disruptions can hinder vascular repair during ischemia, but the specific tissues involved and the impact on muscle health remain unclear. This study investigates the role of the skeletal muscle circadian clock in muscle adaptation to ischemic stress using a surgical mouse model of hindlimb ischemia. METHODS We performed secondary analysis of publicly available RNA-sequencing data sets derived from patients with PAD to identify the differential expression of circadian-related genes in endothelial cells and ischemic limb skeletal muscles. We used mice with specific genetic loss of the circadian clock activator, BMAL1 (brain and muscle ARNT-like 1), in adult skeletal muscle tissues (Bmal1muscle). Bmal1muscle mice and controls underwent femoral artery ligation surgery to induce hindlimb ischemia. Laser Doppler imaging was used to assess limb perfusion at various time points after the surgery. Muscle tissues were analyzed with RNA sequencing and histological examination to investigate PAD-related muscle pathologies. Additionally, we studied the role of BMAL1 in muscle fiber adaptation to hypoxia using RNA and assay for transposase-accessible chromatin with sequencing analyses in primary myotube culture model. RESULTS Disrupted expression of circadian rhythm-related genes was observed in existing RNA-sequencing data sets from endothelial cells and ischemic limb skeletal muscles derived from patients with PAD. Genetic loss of Bmal1 specifically in adult mouse skeletal muscle tissues delayed reperfusion recovery following induction of hindlimb ischemia. Histological examination of muscle tissues showed reduced regenerated myofiber number and a decreased proportion of type IIB fast-twitch myofibers in Bmal1muscle mouse muscles in the ischemic limbs but not in their contralateral nonischemic limbs. Transcriptomic analysis revealed abrogated metabolic, angiogenic, and myogenic pathways relevant to hypoxia adaptation in Bmal1muscle mouse muscles. These changes were corroborated in Bmal1-deficient cultured primary myotubes cultured under hypoxic conditions. CONCLUSIONS Circadian clock in skeletal muscle is crucial for the muscle's response to hypoxia during hindlimb ischemia. Targeting the muscle circadian clock may have therapeutic potential for enhancing muscle response to reduced blood flow and promoting recovery in conditions such as PAD.
Collapse
Affiliation(s)
- Pei Zhu (朱培)
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Calvin L Chao
- Department of Surgery, Division of Vascular Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Adam W T Steffeck
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Caitlyn Dang
- Department of Surgery, Division of Vascular Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Noah X Hamlish
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Eric M Pfrender
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Bin Jiang
- Department of Surgery, Division of Vascular Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Chicago, IL, USA
| | - Clara B Peek
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
3
|
Zhao W, Lu J, Yan H, Zhu J, Liu Y, Song X, Suo T, Miao L. Treatment of acute pharyngitis in rats with season tea decoctions from traditional Chinese medicine through a synergistic and subtle regulation of ARNTL and BHLHE40. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118924. [PMID: 39389396 DOI: 10.1016/j.jep.2024.118924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 10/05/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE While the seasonal variations in the human immune function and many infectious diseases are well-known, to develop therapeutic strategies regarding such seasonality is quite challenging. However, some traditional medical practices have already taken the seasonality into account, such as the "Season Tea" (ST) decoctions investigated in the present study. AIM OF THE STUDY We present a study of the ST decoctions from traditional Chinese medicine, which include four formulae designed for the four seasons, aiming to investigate their pharmacological commonality and distinction. MATERIALS AND METHODS A rat model of acute pharyngitis was utilized for the pharmacological study, and the effects of the ST decoctions were evaluated through histology, biomedical assays, microarray analysis, real-time quantitative PCR and Western blot. RESULTS The experimental data show that all of the four ST formulae display good pharmaceutical effects on acute pharyngitis, and circadian rhythm appears to be a significant pathway for investigating their pharmacological commonality and distinction. Specifically, while all of the four ST decoctions can regulate the circadian-rhythm-related genes ARNTL and BHLHE40, the regulation is along different directions with the modification of the supplements and the substrates in each ST formula. CONCLUSION These results indicate the correlation between the acute pharyngitis and circadian rhythm, and illustrate the possibility of synergistically and subtly regulating ARNTL and BHLHE40, which is significant for relevant drug development.
Collapse
Affiliation(s)
- Wei Zhao
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine Tianjin, 301617, China
| | - Jia Lu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine Tianjin, 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Huimin Yan
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Junjie Zhu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine Tianjin, 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yang Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine Tianjin, 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xinbo Song
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine Tianjin, 301617, China; College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Tongchuan Suo
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine Tianjin, 301617, China; College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Chinese Medicine Modernization, Tianjin, 301617, China; Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Lin Miao
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine Tianjin, 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Chinese Medicine Modernization, Tianjin, 301617, China.
| |
Collapse
|
4
|
Gentile F, Emdin M, Passino C, Montuoro S, Tognini P, Floras JS, O'Neill J, Giannoni A. The chronobiology of human heart failure: clinical implications and therapeutic opportunities. Heart Fail Rev 2025; 30:103-116. [PMID: 39392534 DOI: 10.1007/s10741-024-10447-1] [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] [Accepted: 09/29/2024] [Indexed: 10/12/2024]
Abstract
Circadian variation in cardiovascular and metabolic dynamics arises from interactions between intrinsic rhythms and extrinsic cues. By anticipating and accommodating adaptation to awakening and activity, their synthesis maintains homeostasis and maximizes efficiency, flexibility, and resilience. The dyssynchrony of cardiovascular load and energetic capacity arising from attenuation or loss of such rhythms is strongly associated with incident heart failure (HF). Once established, molecular, neurohormonal, and metabolic rhythms are frequently misaligned with each other and with extrinsic cycles, contributing to HF progression and adverse outcomes. Realignment of biological rhythms via lifestyle interventions, chronotherapy, and time-tailored autonomic modulation represents an appealing potential strategy for improving HF-related morbidity and mortality.
Collapse
Affiliation(s)
- Francesco Gentile
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa, Italy
- Division of Cardiology and Cardiovascular Medicine, Fondazione Monasterio, Pisa, Italy
| | - Michele Emdin
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa, Italy
- Division of Cardiology and Cardiovascular Medicine, Fondazione Monasterio, Pisa, Italy
| | - Claudio Passino
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa, Italy
- Division of Cardiology and Cardiovascular Medicine, Fondazione Monasterio, Pisa, Italy
| | - Sabrina Montuoro
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Paola Tognini
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa, Italy
| | - John S Floras
- University Health Network and Sinai Health Division of Cardiology, Toronto, ON, Canada
| | - John O'Neill
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Alberto Giannoni
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa, Italy.
- Division of Cardiology and Cardiovascular Medicine, Fondazione Monasterio, Pisa, Italy.
| |
Collapse
|
5
|
Moraes RCM, Roth JR, Mao H, Crawley SR, Xu BP, Watson JC, Melkani GC. Apolipoprotein E Induces Lipid Accumulation Through Dgat2 That Is Prevented with Time-Restricted Feeding in Drosophila. Genes (Basel) 2024; 15:1376. [PMID: 39596576 PMCID: PMC11594465 DOI: 10.3390/genes15111376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/19/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Apolipoprotein E (ApoE) is the leading genetic risk factor for late-onset Alzheimer's disease (AD), which is the leading cause of dementia worldwide. Most people have two ApoE-ε3 (ApoE3) alleles, while ApoE-ε2 (ApoE2) is protective from AD, and ApoE-ε4 (ApoE4) confers AD risk. How these alleles modulate AD risk is not clearly defined, and ApoE's role in lipid metabolism is also not fully known. Lipid droplets increase in AD. However, how ApoE contributes to lipid accumulation in the brain remains unknown. Methods: Here, we use Drosophila to study the effects of ApoE alleles on lipid accumulation in the brain and muscle in a cell-autonomous and non-cell-autonomous manner. Results: We report that pan-neuronal expression of each ApoE allele induces lipid accumulation specifically in the brain, but not in the muscle. However, this was not the case when expressed with muscle-specific drivers. ApoE2- and ApoE3-induced lipid accumulation is dependent on the expression of Dgat2, a key regulator of triacylglycerol production, while ApoE4 still induces lipid accumulation even with knock-down of Dgat2. Additionally, we find that implementation of time-restricted feeding (TRF), a dietary intervention in which food access only occurs in the active period (day), prevents ApoE-induced lipid accumulation in the brain of flies and modulates lipid metabolism genes. Conclusions: Altogether, our results demonstrate that ApoE induces lipid accumulation in the brain, that ApoE4 is unique in causing lipid accumulation independent of Dgat2, and that TRF prevents ApoE-induced lipid accumulation. These results support the idea that lipid metabolism is critical in AD, and that TRF could be a promising therapeutic approach to prevent ApoE-associated dysfunction in lipid metabolism.
Collapse
Affiliation(s)
- Ruan C. M. Moraes
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Psychiatry and Behavioral Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jonathan R. Roth
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Hailey Mao
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Savannah R. Crawley
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Brittney P. Xu
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - John C. Watson
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Girish C. Melkani
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- UAB Nathan Shock Center, 1300 University Boulevard, Birmingham, AL 35294, USA
| |
Collapse
|
6
|
Festus ID, Spilberg J, Young ME, Cain S, Khoshnevis S, Smolensky MH, Zaheer F, Descalzi G, Martino TA. Pioneering new frontiers in circadian medicine chronotherapies for cardiovascular health. Trends Endocrinol Metab 2024; 35:607-623. [PMID: 38458859 DOI: 10.1016/j.tem.2024.02.011] [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: 12/21/2023] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 03/10/2024]
Abstract
Cardiovascular disease (CVD) is a global health concern. Circadian medicine improves cardiovascular care by aligning treatments with our body's daily rhythms and their underlying cellular circadian mechanisms. Time-based therapies, or chronotherapies, show special promise in clinical cardiology. They optimize treatment schedules for better outcomes with fewer side effects by recognizing the profound influence of rhythmic body cycles. In this review, we focus on three chronotherapy areas (medication, light, and meal timing) with potential to enhance cardiovascular care. We also highlight pioneering research in the new field of rest, the gut microbiome, novel chronotherapies for hypertension, pain management, and small molecules that targeting the circadian mechanism.
Collapse
Affiliation(s)
- Ifene David Festus
- Centre for Cardiovascular Investigations, University of Guelph; Guelph, Ontario, Canada; Department of Biomedical Sciences, University of Guelph; Guelph, Ontario, Canada
| | - Jeri Spilberg
- Centre for Cardiovascular Investigations, University of Guelph; Guelph, Ontario, Canada; Department of Biomedical Sciences, University of Guelph; Guelph, Ontario, Canada
| | - Martin E Young
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sean Cain
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, Victoria, Australia
| | - Sepideh Khoshnevis
- Department of Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Michael H Smolensky
- Department of Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX, USA; Department of Internal Medicine, Division of Cardiology, McGovern School of Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Fariya Zaheer
- Department of Biomedical Sciences, University of Guelph; Guelph, Ontario, Canada
| | - Giannina Descalzi
- Department of Biomedical Sciences, University of Guelph; Guelph, Ontario, Canada
| | - Tami A Martino
- Centre for Cardiovascular Investigations, University of Guelph; Guelph, Ontario, Canada; Department of Biomedical Sciences, University of Guelph; Guelph, Ontario, Canada.
| |
Collapse
|
7
|
Farag HI, Murphy BA, Templeman JR, Hanlon C, Joshua J, Koch TG, Niel L, Shoveller AK, Bedecarrats GY, Ellison A, Wilcockson D, Martino TA. One Health: Circadian Medicine Benefits Both Non-human Animals and Humans Alike. J Biol Rhythms 2024; 39:237-269. [PMID: 38379166 PMCID: PMC11141112 DOI: 10.1177/07487304241228021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Circadian biology's impact on human physical health and its role in disease development and progression is widely recognized. The forefront of circadian rhythm research now focuses on translational applications to clinical medicine, aiming to enhance disease diagnosis, prognosis, and treatment responses. However, the field of circadian medicine has predominantly concentrated on human healthcare, neglecting its potential for transformative applications in veterinary medicine, thereby overlooking opportunities to improve non-human animal health and welfare. This review consists of three main sections. The first section focuses on the translational potential of circadian medicine into current industry practices of agricultural animals, with a particular emphasis on horses, broiler chickens, and laying hens. The second section delves into the potential applications of circadian medicine in small animal veterinary care, primarily focusing on our companion animals, namely dogs and cats. The final section explores emerging frontiers in circadian medicine, encompassing aquaculture, veterinary hospital care, and non-human animal welfare and concludes with the integration of One Health principles. In summary, circadian medicine represents a highly promising field of medicine that holds the potential to significantly enhance the clinical care and overall health of all animals, extending its impact beyond human healthcare.
Collapse
Affiliation(s)
- Hesham I. Farag
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
- Centre for Cardiovascular Investigations, University of Guelph, Guelph, ON, Canada
| | - Barbara A. Murphy
- School of Agriculture and Food Science, University College, Dublin, Ireland
| | - James R. Templeman
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
| | - Charlene Hanlon
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
- Department of Poultry Science, Auburn University, Auburn, Alabama, USA
| | - Jessica Joshua
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Thomas G. Koch
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Lee Niel
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada
| | - Anna K. Shoveller
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
| | | | - Amy Ellison
- School of Natural Sciences, Bangor University, Bangor, UK
| | - David Wilcockson
- Department of Life Sciences, Aberystwyth University, Aberystwyth, UK
| | - Tami A. Martino
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
- Centre for Cardiovascular Investigations, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
8
|
Eckle T, Bertazzo J, Khatua TN, Tabatabaei SRF, Bakhtiari NM, Walker LA, Martino TA. Circadian Influences on Myocardial Ischemia-Reperfusion Injury and Heart Failure. Circ Res 2024; 134:675-694. [PMID: 38484024 PMCID: PMC10947118 DOI: 10.1161/circresaha.123.323522] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 02/02/2024] [Accepted: 02/08/2024] [Indexed: 03/19/2024]
Abstract
The impact of circadian rhythms on cardiovascular function and disease development is well established, with numerous studies in genetically modified animals emphasizing the circadian molecular clock's significance in the pathogenesis and pathophysiology of myocardial ischemia and heart failure progression. However, translational preclinical studies targeting the heart's circadian biology are just now emerging and are leading to the development of a novel field of medicine termed circadian medicine. In this review, we explore circadian molecular mechanisms and novel therapies, including (1) intense light, (2) small molecules modulating the circadian mechanism, and (3) chronotherapies such as cardiovascular drugs and meal timings. These promise significant clinical translation in circadian medicine for cardiovascular disease. (4) Additionally, we address the differential functioning of the circadian mechanism in males versus females, emphasizing the consideration of biological sex, gender, and aging in circadian therapies for cardiovascular disease.
Collapse
Affiliation(s)
- Tobias Eckle
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Júlia Bertazzo
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Tarak Nath Khatua
- Centre for Cardiovascular Investigations, Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Seyed Reza Fatemi Tabatabaei
- Centre for Cardiovascular Investigations, Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Naghmeh Moori Bakhtiari
- Centre for Cardiovascular Investigations, Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Lori A Walker
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Tami A. Martino
- Centre for Cardiovascular Investigations, Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
9
|
Ferreira LL, Rosatto N, Marzullo P, Bellan M. Circadian variations in the elderly: A scoping review. Chronobiol Int 2024; 41:311-328. [PMID: 38501270 DOI: 10.1080/07420528.2024.2327456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/28/2024] [Accepted: 03/02/2024] [Indexed: 03/20/2024]
Abstract
The circadian clock plays important roles in several physiological processes. With aging, some of these circadian rhythms have been shown to be disrupted and suggested contributing to age-related diseases. The aim of this scoping review was to examine and map the existing evidence of circadian differences between young and older people in body fluid composition. Literature search was carried out on PubMed, Embase, Scopus and OpenGrey. The studies were screened based on inclusion and exclusion criteria by two independent reviewers and the results were summarized tabularly and narratively. The review process resulted in the identification of 1889 publications, of which 42 were eligible for inclusion. Forty-eight parameters or families of parameters were identified, including cortisol and melatonin, sex hormones, thyroid-related hormones, steroids and aldosterone. However, many were reported by only a single study. The results from the studies were heterogeneous. Even though the majority suggested the flattening of several circadian oscillations in the elderly population, this was not always observed for all the parameters analyzed, and some contradictory results were found. This review revealed a substantial number of publications that explored this research question, but further studies would be important to elucidate the clinical significance of these alterations.
Collapse
Affiliation(s)
- Luciana L Ferreira
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Nadia Rosatto
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Paolo Marzullo
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Mattia Bellan
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
- Department of Internal Medicine and Rheumatology Unit, Azienda Ospedaliero-Universitaria Maggiore Della Carità, Novara, Italy
- Center on Autoimmune and Allergic Diseases, Università del Piemonte Orientale, Novara, Italy
| |
Collapse
|
10
|
Yoo SH. Circadian regulation of cardiac muscle function and protein degradation. Chronobiol Int 2023; 40:4-12. [PMID: 34521283 PMCID: PMC8918439 DOI: 10.1080/07420528.2021.1957911] [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: 04/23/2021] [Revised: 07/12/2021] [Accepted: 07/13/2021] [Indexed: 10/20/2022]
Abstract
The circadian clock plays a fundamental role in physiology. In particular, the heart is a target organ where the clock orchestrates various aspects of cardiac function. At the molecular level, the clock machinery governs daily rhythms of gene expression. Such circadian regulation is in tune with the dynamic nature of heart structure and function, and provides the foundation for chronotherapeutic applications in cardiovascular diseases. In comparison, a regulatory role of the clock in cardiac protein degradation is poorly documented. Sarcomere is the structural and functional unit responsible for cardiac muscle contraction, and sarcomere components are closely regulated by protein folding and proteolysis. Emerging evidence supports a role of the circadian clock in governing sarcomere integrity and function. Particularly, recent studies uncovered a circadian regulation of a core sarcomere component TCAP. It is possible that circadian regulation of the cardiac muscle protein turnover is a key regulatory mechanism underlying cardiac remodeling in response to physiological and environmental stimuli. While the detailed regulatory mechanisms and the molecular links to cardiac (patho)physiology remain to be further studied, therapeutic strategies targeting circadian control in the heart may markedly enhance intervention outcomes against cardiovascular disease.
Collapse
Affiliation(s)
- Seung-Hee Yoo
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
11
|
Zhang Y, Liu L, Zhao X, Yan S, Zeng F, Zhou D. New insight into ischemic stroke: Circadian rhythm in post-stroke angiogenesis. Front Pharmacol 2022; 13:927506. [PMID: 36016550 PMCID: PMC9395980 DOI: 10.3389/fphar.2022.927506] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/07/2022] [Indexed: 11/29/2022] Open
Abstract
The circadian rhythm is an endogenous clock system that coordinates and optimizes various physiological and pathophysiological processes, which accord with the master and the peripheral clock. Increasing evidence indicates that endogenous circadian rhythm disruption is involved in the lesion volume and recovery of ischemic stroke. As a critical recovery mechanism in post-stroke, angiogenesis reestablishes the regional blood supply and enhances cognitive and behavioral abilities, which is mainly composed of the following processes: endothelial cell proliferation, migration, and pericyte recruitment. The available evidence revealed that the circadian governs many aspects of angiogenesis. This study reviews the mechanism by which circadian rhythms regulate the process of angiogenesis and its contribution to functional recovery in post-stroke at the aspects of the molecular level. A comprehensive understanding of the circadian clock regulating angiogenesis in post-stroke is expected to develop new strategies for the treatment of cerebral infarction.
Collapse
Affiliation(s)
- Yuxing Zhang
- The Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China
| | - Lijuan Liu
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xin Zhao
- The Medical School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Siyang Yan
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Fukang Zeng
- The Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China
| | - Desheng Zhou
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- *Correspondence: Desheng Zhou,
| |
Collapse
|
12
|
Blanco JR, Verdugo-Sivianes EM, Amiama A, Muñoz-Galván S. The circadian rhythm of viruses and its implications on susceptibility to infection. Expert Rev Anti Infect Ther 2022; 20:1109-1117. [PMID: 35546444 DOI: 10.1080/14787210.2022.2072296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Circadian genes have an impact on multiple hormonal, metabolic, and immunological pathways and have recently been implicated in some infectious diseases. AREAS COVERED We review aspects related to the current knowledge about circadian rhythm and viral infections, their consequences, and the potential therapeutic options. EXPERT OPINION Expert opinion: In order to address a problem, it is necessary to know the topic in depth. Although in recent years there has been a growing interest in the role of circadian rhythms, many relevant questions remain to be resolved. Thus, the mechanisms linking the circadian machinery against viral infections are poorly understood. In a clear approach to personalized precision medicine, in order to treat a disease in the most appropriate phase of the circadian rhythm, and in order to achieve the optimal efficacy, it is highly recommended to carry out studies that improve the knowledge about the circadian rhythm.
Collapse
Affiliation(s)
- José-Ramon Blanco
- Servicio de Enfermedades Infecciosas, Hospital Universitario San Pedro, Logroño, Spain.,Centro de Investigación Biomédica de La Rioja (CIBIR), Logroño, Spain
| | - Eva M Verdugo-Sivianes
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Sevilla, Spain.,CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Amiama
- Centro de Investigación Biomédica de La Rioja (CIBIR), Logroño, Spain
| | - Sandra Muñoz-Galván
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Sevilla, Spain.,CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
13
|
Padgett CA, Butcher JT, Haigh SB, Speese AC, Corley ZL, Rosewater CL, Sellers HG, Larion S, Mintz JD, Fulton DJR, Stepp DW. Obesity Induces Disruption of Microvascular Endothelial Circadian Rhythm. Front Physiol 2022; 13:887559. [PMID: 35600313 PMCID: PMC9119407 DOI: 10.3389/fphys.2022.887559] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Obese individuals are at significantly elevated risk of developing cardiovascular disease (CVD). Additionally, obesity has been associated with disrupted circadian rhythm, manifesting in abnormal sleeping and feeding patterns. To date, the mechanisms linking obesity, circadian disruption, and CVD are incompletely understood, and insight into novel mechanistic pathways is desperately needed to improve therapeutic potential and decrease morbidity and mortality. The objective of this study was to investigate the roles of metabolic and circadian disruptions in obesity and assess their contributions in promoting vascular disease. Lean (db/+) and obese (db/db) mice were subjected to 12 weeks of constant darkness to differentiate diurnal and circadian rhythms, and were assessed for changes in metabolism, gene expression, and vascular function. Expression of endothelial nitric oxide synthase (eNOS), an essential enzyme for vascular health, was blunted in obesity and correlated with the oscillatory loss of the novel regulator cezanne (OTUD7B). Lean mice subjected to constant darkness displayed marked reduction in vasodilatory capacity, while endothelial dysfunction of obese mice was not further compounded by diurnal insult. Endothelial gene expression of essential circadian clock components was altered in obesity, but imperfectly phenocopied in lean mice housed in constant darkness, suggesting overlapping but separate mechanisms driving endothelial dysfunction in obesity and circadian disruption. Taken together, these data provide insight into the nature of endothelial circadian rhythm in obesity and suggest a distinct mechanism by which obesity causes a unique circadian defect in the vasculature.
Collapse
Affiliation(s)
| | - Joshua T. Butcher
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, United States
| | | | | | | | | | | | - Sebastian Larion
- Division of Gastroenterology and Hepatology, Department of Medicine, College of Medicine, Medical University of South Carolina, Charleston, SC, United States
| | | | - David J. R. Fulton
- Vascular Biology Center, Augusta, GA, United States,Department of Pharmacology and Toxicology, Augusta, GA, United States
| | - David W. Stepp
- Vascular Biology Center, Augusta, GA, United States,Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States,*Correspondence: David W. Stepp,
| |
Collapse
|
14
|
Schroder EA, Ono M, Johnson SR, Rozmus ER, Burgess DE, Esser KA, Delisle BP. The role of the cardiomyocyte circadian clocks in ion channel regulation and cardiac electrophysiology. J Physiol 2022; 600:2037-2048. [PMID: 35301719 PMCID: PMC9980729 DOI: 10.1113/jp282402] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 03/04/2022] [Indexed: 11/08/2022] Open
Abstract
Daily variations in cardiac electrophysiology and the incidence for different types of arrhythmias reflect ≈24 h changes in the environment, behaviour and internal circadian rhythms. This article focuses on studies that use animal models to separate the impact that circadian rhythms, as well as changes in the environment and behaviour, have on 24 h rhythms in heart rate and ventricular repolarization. Circadian rhythms are initiated at the cellular level by circadian clocks, transcription-translation feedback loops that cycle with a periodicity of 24 h. Several studies now show that the circadian clock in cardiomyocytes regulates the expression of cardiac ion channels by multiple mechanisms; underlies time-of-day changes in sinoatrial node excitability/intrinsic heart rate; and limits the duration of the ventricular action potential waveform. However, the 24 h rhythms in heart rate and ventricular repolarization are primarily driven by autonomic signalling. A functional role for the cardiomyocyte circadian clock appears to buffer the heart against perturbations. For example, the cardiomyocyte circadian clock limits QT-interval prolongation (especially at slower heart rates), and it may facilitate the realignment of the 24 h rhythm in heart rate to abrupt changes in the light cycle. Additional studies show that modifying rhythmic behaviours (including feeding behaviour) can dramatically impact the 24 h rhythms in heart rate and ventricular repolarization. If these mechanisms are conserved, these studies suggest that targeting endogenous circadian mechanisms in the heart, as well as modifying the timing of certain rhythmic behaviours, could emerge as therapeutic strategies to support heart function against perturbations and regulate 24 h rhythms in cardiac electrophysiology.
Collapse
Affiliation(s)
- Elizabeth A. Schroder
- Department of Physiology, University of Kentucky, 800 Rose Street, MN508, Lexington, KY 40536-0298
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Kentucky, 740 S. Limestone Street, L543, Lexington, KY 40536-0284
| | - Makoto Ono
- Department of Physiology, University of Kentucky, 800 Rose Street, MN508, Lexington, KY 40536-0298
| | - Sidney R. Johnson
- Department of Physiology, University of Kentucky, 800 Rose Street, MN508, Lexington, KY 40536-0298
| | - Ezekiel R. Rozmus
- Department of Physiology, University of Kentucky, 800 Rose Street, MN508, Lexington, KY 40536-0298
| | - Don E. Burgess
- Department of Physiology, University of Kentucky, 800 Rose Street, MN508, Lexington, KY 40536-0298
| | - Karyn A. Esser
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA
| | - Brian P. Delisle
- Department of Physiology, University of Kentucky, 800 Rose Street, MN508, Lexington, KY 40536-0298
| |
Collapse
|
15
|
Modelling Female Physiology from Head to Toe: Impact of Sex Hormones, Menstrual Cycle, and Pregnancy. J Theor Biol 2022; 540:111074. [PMID: 35227731 DOI: 10.1016/j.jtbi.2022.111074] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 12/14/2022]
|
16
|
Liu Y, Wu Z, Cheng L, Zhang X, Yang H. The role of the intestinal microbiota in the pathogenesis of host depression and mechanism of TPs relieving depression. Food Funct 2021; 12:7651-7663. [PMID: 34286799 DOI: 10.1039/d1fo01091c] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Depression is a prevalent neuropsychiatric disease with a high recurrence rate, affecting over 350 million people worldwide. Intestinal flora disorders and gut-brain-axis (GBA) dysfunction may cause mental disorders. Alterations in the intestinal flora composition could increase the permeability of the gut barrier, activate systemic inflammation and immune responses, regulate the release and efficacy of monoamine neurotransmitters, alter the activity and function of the hypothalamic-pituitary-adrenal (HPA) axis, and modify the abundance of the brain-derived neurotrophic factor (BDNF); all of these showed a close correlation with the occurrence of depression. In addition, the disturbance of the intestinal flora is related to circadian rhythm disorders, which aggravate the symptoms of depression. Tea polyphenols (TPs) have been found to have antidepressant effects. Therefore, the close reciprocity between the intestinal flora and circadian rhythm provides a new opportunity for TPs to regulate depression relying on the intestinal flora. In this review, we discussed the relationship between intestinal flora dysbiosis and the pathogenesis of depression and the mechanism of TPs relieving depression via the GBA.
Collapse
Affiliation(s)
- Yang Liu
- Laboratory of Food Biotechnology, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo315211, PR China.
| | | | | | | | | |
Collapse
|
17
|
Kenig A, Kolben Y, Asleh R, Amir O, Ilan Y. Improving Diuretic Response in Heart Failure by Implementing a Patient-Tailored Variability and Chronotherapy-Guided Algorithm. Front Cardiovasc Med 2021; 8:695547. [PMID: 34458334 PMCID: PMC8385752 DOI: 10.3389/fcvm.2021.695547] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/21/2021] [Indexed: 01/12/2023] Open
Abstract
Heart failure is a major public health problem, which is associated with significant mortality, morbidity, and healthcare expenditures. A substantial amount of the morbidity is attributed to volume overload, for which loop diuretics are a mandatory treatment. However, the variability in response to diuretics and development of diuretic resistance adversely affect the clinical outcomes. Morevoer, there exists a marked intra- and inter-patient variability in response to diuretics that affects the clinical course and related adverse outcomes. In the present article, we review the mechanisms underlying the development of diuretic resistance. The role of the autonomic nervous system and chronobiology in the pathogenesis of congestive heart failure and response to therapy are also discussed. Establishing a novel model for overcoming diuretic resistance is presented based on a patient-tailored variability and chronotherapy-guided machine learning algorithm that comprises clinical, laboratory, and sensor-derived inputs, including inputs from pulmonary artery measurements. Inter- and intra-patient signatures of variabilities, alterations of biological clock, and autonomic nervous system responses are embedded into the algorithm; thus, it may enable a tailored dose regimen in a continuous manner that accommodates the highly dynamic complex system.
Collapse
Affiliation(s)
- Ariel Kenig
- Department of Medicine, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Yotam Kolben
- Department of Medicine, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Rabea Asleh
- Department of Cardiology, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Offer Amir
- Department of Cardiology, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, Israel
| | - Yaron Ilan
- Department of Medicine, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| |
Collapse
|
18
|
Gombert M, Codoñer-Franch P. Melatonin in Early Nutrition: Long-Term Effects on Cardiovascular System. Int J Mol Sci 2021; 22:6809. [PMID: 34202781 PMCID: PMC8269134 DOI: 10.3390/ijms22136809] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/21/2021] [Accepted: 06/21/2021] [Indexed: 02/07/2023] Open
Abstract
Breastfeeding protects against adverse cardiovascular outcomes in the long term. Melatonin is an active molecule that is present in the breast milk produced at night beginning in the first stages of lactation. This indoleamine appears to be a relevant contributor to the benefits of breast milk because it can affect infant health in several ways. The melatonin concentration in breast milk varies in a circadian pattern, making breast milk a chrononutrient. The consumption of melatonin can induce the first circadian stimulation in the infant's body at an age when his/her own circadian machinery is not functioning yet. This molecule is also a powerful antioxidant with the ability to act on infant cells directly as a scavenger and indirectly by lowering oxidant molecule production and enhancing the antioxidant capacity of the body. Melatonin also participates in regulating inflammation. Furthermore, melatonin can participate in shaping the gut microbiota composition, richness, and variation over time, also modulating which molecules are absorbed by the host. In all these ways, melatonin from breast milk influences weight gain in infants, limiting the development of obesity and comorbidities in the long term, and it can help shape the ideal cellular environment for the development of the infant's cardiovascular system.
Collapse
Affiliation(s)
- Marie Gombert
- Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, 46010 Valencia, Spain
| | - Pilar Codoñer-Franch
- Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, 46010 Valencia, Spain
- Service of Pediatrics, Hospital Universitario del Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46010 Valencia, Spain;
| |
Collapse
|
19
|
Clock-Modulating Activities of the Anti-Arrhythmic Drug Moricizine. Clocks Sleep 2021; 3:351-365. [PMID: 34206497 PMCID: PMC8293187 DOI: 10.3390/clockssleep3030022] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/09/2021] [Accepted: 06/21/2021] [Indexed: 12/12/2022] Open
Abstract
Dysregulated circadian functions contribute to various diseases, including cardiovascular disease. Much progress has been made on chronotherapeutic applications of drugs against cardiovascular disease (CVD); however, the direct effects of various medications on the circadian system are not well characterized. We previously conducted high-throughput chemical screening for clock modulators and identified an off-patent anti-arrhythmic drug, moricizine, as a clock-period lengthening compound. In Per2:LucSV reporter fibroblast cells, we showed that under both dexamethasone and forskolin synchronization, moricizine was able to increase the circadian period length, with greater effects seen with the former. Titration studies revealed a dose-dependent effect of moricizine to lengthen the period. In contrast, flecainide, another Class I anti-arrhythmic, showed no effects on circadian reporter rhythms. Real-time qPCR analysis in fibroblast cells treated with moricizine revealed significant circadian time- and/or treatment-dependent expression changes in core clock genes, consistent with the above period-lengthening effects. Several clock-controlled cardiac channel genes also displayed altered expression patterns. Using tissue explant culture, we showed that moricizine was able to significantly prolong the period length of circadian reporter rhythms in atrial ex vivo cultures. Using wild-type C57BL/6J mice, moricizine treatment was found to promote sleep, alter circadian gene expression in the heart, and show a slight trend of increasing free-running periods. Together, these observations demonstrate novel clock-modulating activities of moricizine, particularly the period-lengthening effects on cellular oscillators, which may have clinical relevance against heart diseases.
Collapse
|
20
|
Lopuszko A, Patrick Tan SZC, Munir W, Bashir M. Aortic aneurysm disease-Make room for chronobiology. J Card Surg 2021; 36:2496-2501. [PMID: 33797793 DOI: 10.1111/jocs.15548] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 03/02/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Aortic aneurysm (AA) is a common atherosclerotic condition, accounting for nearly 6000 deaths in England and up to 175,000 deaths globally each year. The pathological outward bulging of the aorta typically results from atherosclerosis or hereditary connective tissue disorders. AAs are usually asymptomatic until spontaneous rupture or detected on incidental screening. Eight in 10 patients do not survive the rupture and die either before reaching hospital or from complications following surgery. Similar to other cardiovascular pathologies, AA is thought to be subject to chronobiological patterns of varying incidence. METHODS We performed a literature review of the current literature to evaluate the association between circadian rhythms, seasonal variations, and genetic factors and the pathogenesis of AA, reviewing the impact of chronobiology. RESULTS The incidence of AA is found to peak in the early morning (6-11 a.m.) and colder months, and conversely troughs towards the evening and warmer months, exhibiting a similar pattern of chronobiological rhythm as other cerebrovascular pathologies, such as myocardial infarcts, or cerebrovascular strokes. CONCLUSION Literature suggests there exists a clear relationship between chronobiology and the incidence and pathogenesis of ruptured AA; incidence increases in the morning (6-11 a.m.), and during colder months (December-January). This is more pronounced in patients with Marfan syndrome, or vitamin D deficiency. The underlying pathophysiology and implications this has for chronotherapeutics, are also discussed. Our review shows a clear need for further research into the chronotherapeutic approach to preventing ruptured AA in the journey towards precision medicine.
Collapse
Affiliation(s)
- Aleksandra Lopuszko
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Sven Zhen Cian Patrick Tan
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Wahaj Munir
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Mohamad Bashir
- Vascular Surgery Department, Royal Blackburn Teaching Hospital, Blackburn, United Kingdom
| |
Collapse
|
21
|
Abstract
Circadian rhythms describe physiological systems that repeat themselves with a cycle of approximately 24 h. Our understanding of the cellular and molecular origins of these oscillations has improved dramatically, allowing us to appreciate the significant role these oscillations play in maintaining physiological homeostasis. Circadian rhythms allow living organisms to predict and efficiently respond to a dynamically changing environment, set by repetitive day/night cycles. Since circadian rhythms underlie almost every aspect of human physiology, it is unsurprising that they also influence the response of a living organism to disease, stress, and therapeutics. Therefore, not only do the mechanisms that maintain health and disrupt homeostasis depend on our internal circadian clock, but also the way drugs are perceived and function depends on these physiological rhythms. We present a holistic view of the therapeutic process, discussing components such as disease state, pharmacokinetics, and pharmacodynamics, as well as adverse reactions that are critically affected by circadian rhythms. We outline challenges and opportunities in moving toward personalized medicine approaches that explore and capitalize on circadian rhythms for the benefit of the patient.
Collapse
Affiliation(s)
- Yaakov Nahmias
- Center for Bioengineering, School of Computer Science and Engineering, Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Ioannis P Androulakis
- Department of Biomedical Engineering and Department of Chemical & Biochemical Engineering, Rutgers University, Piscataway, New Jersey 08854, USA; .,Department of Surgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey 08854, USA
| |
Collapse
|
22
|
Alkhodari M, Jelinek HF, Werghi N, Hadjileontiadis LJ, Khandoker AH. Estimating Left Ventricle Ejection Fraction Levels Using Circadian Heart Rate Variability Features and Support Vector Regression Models. IEEE J Biomed Health Inform 2021; 25:746-754. [PMID: 32750938 DOI: 10.1109/jbhi.2020.3002336] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
OBJECTIVES The purpose of this study was to set an optimal fit of the estimated LVEF at hourly intervals from 24-hour ECG recordings and compare it with the fit based on two gold-standard guidelines. METHODS Support vector regression (SVR) models were applied to estimate LVEF from ECG derived heart rate variability (HRV) data in one-hour intervals from 24-hour ECG recordings of patients with either preserved, mid-range, or reduced LVEF, obtained from the Intercity Digital ECG Alliance (IDEAL) study. A step-wise feature selection approach was used to ensure the best possible estimations of LVEF levels. RESULTS The experimental results have shown that the lowest Root Mean Square Error (RMSE) between the original and estimated LVEF levels was during 3-4 am, 5-6 am and 6-7 pm. CONCLUSION The observations suggest these hours as possible times for intervention and optimal treatment outcomes. In addition, LVEF classifications following the ACCF/AHA guidelines leads to a more accurate assessment of mid-range LVEF. SIGNIFICANCE This study paves the way to explore the use of HRV features in the prediction of LVEF percentages as an indicator of disease progression, which may lead to an automated classification process for CAD patients.
Collapse
|
23
|
Ruan W, Yuan X, Eltzschig HK. Circadian rhythm as a therapeutic target. Nat Rev Drug Discov 2021; 20:287-307. [PMID: 33589815 DOI: 10.1038/s41573-020-00109-w] [Citation(s) in RCA: 213] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2020] [Indexed: 12/20/2022]
Abstract
The circadian clock evolved in diverse organisms to integrate external environmental changes and internal physiology. The clock endows the host with temporal precision and robust adaptation to the surrounding environment. When circadian rhythms are perturbed or misaligned, as a result of jet lag, shiftwork or other lifestyle factors, adverse health consequences arise, and the risks of diseases such as cancer, cardiovascular diseases or metabolic disorders increase. Although the negative impact of circadian rhythm disruption is now well established, it remains underappreciated how to take advantage of biological timing, or correct it, for health benefits. In this Review, we provide an updated account of the circadian system and highlight several key disease areas with altered circadian signalling. We discuss environmental and lifestyle modifications of circadian rhythm and clock-based therapeutic strategies, including chronotherapy, in which dosing time is deliberately optimized for maximum therapeutic index, and pharmacological agents that target core clock components and proximal regulators. Promising progress in research, disease models and clinical applications should encourage a concerted effort towards a new era of circadian medicine.
Collapse
Affiliation(s)
- Wei Ruan
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA.,Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoyi Yuan
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Holger K Eltzschig
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
24
|
Circadian influence on inflammatory response during cardiovascular disease. Curr Opin Pharmacol 2020; 57:60-70. [PMID: 33340915 DOI: 10.1016/j.coph.2020.11.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/26/2020] [Accepted: 11/16/2020] [Indexed: 12/20/2022]
Abstract
Circadian rhythms follow a 24 h day and night cycle, regulate vital physiological processes, and are especially relevant to cardiovascular growth, renewal, repair, and remodeling. A recent flurry of clinical and experimental studies reveals a profound circadian influence on immune responses in cardiovascular disease. The first section of this review summarizes the importance of circadian rhythms for cardiovascular health and disease. The second section introduces the circadian nature of inflammatory responses. The third section combines these to elucidate a new role for the circadian system, influencing inflammation in heart disease, especially myocardial infarction. Particular focus is on circadian regulation of the NACHT, LRR, and PYD domains-containing protein 3 inflammasome, neutrophils, monocytes/macrophages, and T cells involved in cardiac repair. A role for biological sex is noted. The final section explores circadian influences on inflammation in other major cardiovascular conditions. Circadian regulation of inflammation has profound implications for benefitting the diagnosis, treatment, and prognosis of patients with cardiovascular disease.
Collapse
|
25
|
Zhou Z, Yuan J, Zhu D, Chen Y, Qian Z, Wang Y, Ge P, Wang Q, Hou X, Zou J. CLOCK-BMAL1 regulates circadian oscillation of ventricular arrhythmias in failing hearts through β1 adrenergic receptor. Am J Transl Res 2020; 12:6122-6135. [PMID: 33194018 PMCID: PMC7653582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 09/15/2020] [Indexed: 06/11/2023]
Abstract
The incidence of ventricular arrhythmias (VAs) in chronic heart failure (CHF) exhibits a notable circadian rhythm, for which the underlying mechanism has not yet been well defined. Thus, we aimed to investigate the role of cardiac core circadian genes on circadian VAs in CHF. First, a guinea pig CHF model was created by transaortic constriction. Circadian oscillation of core clock genes was evaluated by RT-PCR and was found to be unaltered in CHF (P > 0.05). Using programmed electrical stimulation in Langendorff-perfused failing hearts, we discovered that the CHF group exhibited increased VAs with greater incidence at CT3 compared to CT15 upon isoproterenol (ISO) stimulation. Circadian VAs was blunted by a β1-AR-selective blocker rather than a β2-AR-selective blocker. Circadian oscillation of β1-AR was retained in CHF (P > 0.05) and a 4-h phase delay between β1-AR and CLOCK-BMAL1 was recorded. Therefore, when CLOCK-BMAL1 was overexpressed using adenovirus infection, an induced overexpression of β1-AR also ensued, which resulted in prolonged action potential duration (APD) and enhanced arrhythmic response to ISO stimulation in cardiomyocytes (P < 0.05). Finally, chromatin immunoprecipitation and luciferase assays confirmed that CLOCK-BMAL1 binds to the enhancer of β1-AR gene and upregulates β1-AR expression. Therefore, in this study, we discovered that CLOCK-BMAL1 regulates the expression of β1-AR on a transcriptional level and subsequently modulates circadian VAs in CHF.
Collapse
Affiliation(s)
- Zihao Zhou
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Jiamin Yuan
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
- Department of Cardiology, The First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, China
| | - Didi Zhu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
- Department of Cardiology, Zhongda HospitalNanjing, Jiangsu, China
| | - Yanhong Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Zhiyong Qian
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Yao Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Peibin Ge
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Quanpeng Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Xiaofeng Hou
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Jiangang Zou
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
| |
Collapse
|
26
|
Monfredi O, Lakatta EG. Complexities in cardiovascular rhythmicity: perspectives on circadian normality, ageing and disease. Cardiovasc Res 2020; 115:1576-1595. [PMID: 31150049 DOI: 10.1093/cvr/cvz112] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 02/06/2019] [Accepted: 05/25/2019] [Indexed: 12/13/2022] Open
Abstract
Biological rhythms exist in organisms at all levels of complexity, in most organs and at myriad time scales. Our own biological rhythms are driven by energy emitted by the sun, interacting via our retinas with brain stem centres, which then send out complex messages designed to synchronize the behaviour of peripheral non-light sensing organs, to ensure optimal physiological responsiveness and performance of the organism based on the time of day. Peripheral organs themselves have autonomous rhythmic behaviours that can act independently from central nervous system control but is entrainable. Dysregulation of biological rhythms either through environment or disease has far-reaching consequences on health that we are only now beginning to appreciate. In this review, we focus on cardiovascular rhythms in health, with ageing and under disease conditions.
Collapse
Affiliation(s)
- Oliver Monfredi
- Division of Medicine, Department of Cardiology, The Johns Hopkins Hospital, 1800 Orleans Street, Baltimore, MD, USA.,Laboratory of Cardiovascular Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd, Baltimore, MD, USA
| | - Edward G Lakatta
- Laboratory of Cardiovascular Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd, Baltimore, MD, USA
| |
Collapse
|
27
|
Abstract
The Earth turns on its axis every 24 h; almost all life on the planet has a mechanism - circadian rhythmicity - to anticipate the daily changes caused by this rotation. The molecular clocks that control circadian rhythms are being revealed as important regulators of physiology and disease. In humans, circadian rhythms have been studied extensively in the cardiovascular system. Many cardiovascular functions, such as endothelial function, thrombus formation, blood pressure and heart rate, are now known to be regulated by the circadian clock. Additionally, the onset of acute myocardial infarction, stroke, arrhythmias and other adverse cardiovascular events show circadian rhythmicity. In this Review, we summarize the role of the circadian clock in all major cardiovascular cell types and organs. Second, we discuss the role of circadian rhythms in cardiovascular physiology and disease. Finally, we postulate how circadian rhythms can serve as a therapeutic target by exploiting or altering molecular time to improve existing therapies and develop novel ones.
Collapse
|
28
|
Lin SY. Thermoresponsive gating membranes embedded with liquid crystal(s) for pulsatile transdermal drug delivery: An overview and perspectives. J Control Release 2019; 319:450-474. [PMID: 31901369 DOI: 10.1016/j.jconrel.2019.12.046] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 12/27/2019] [Accepted: 12/28/2019] [Indexed: 01/08/2023]
Abstract
Due to the circadian rhythm regulation of almost every biological process in the human body, physiological and biochemical conditions vary considerably over the course of a 24-h period. Thus, optimal drug delivery and therapy should be effectively controlled to achieve the desired therapeutic plasma concentrations and therapeutic drug responses at the required time according to chronopharmacological concepts, rather than continuous maintenance of constant drug concentrations for an extended time period. For many drugs, it is not always necessary to constantly deliver a drug into the human body under disease conditions due to rhythmic variations. Pulsatile drug delivery systems (PDDSs) have been receiving more attention in pharmaceutical development by providing a predetermined lag period, followed by a fast or rate-controlled drug release after application. PDDSs are characterized by a programmed drug release, which may release a drug at repeatable pulses to match the biological and clinical needs of a given disease therapy. This review article focuses on thermoresponsive gating membranes embedded with liquid crystals (LCs) for transdermal drug delivery using PDDS technology. In addition, the principal rationale and the advanced approaches for the use of PDDSs, the marketed products of chronotherapeutic DDSs with pulsatile function designed by various PDDS technologies, pulsatile drug delivery designed with thermoresponsive polymers, challenges and opportunities of transdermal drug delivery, and novel approaches of LC systems for drug delivery are reviewed and discussed. A brief overview of all academic research articles concerning single LC- or binary LC-embedded thermoresponsive membranes with a switchable on-off permeation function through topical application by an external temperature control, which may modulate the dosing interval and administration time according to the therapeutic needs of the human body, is also compiled and presented. In the near future, since thermal-based approaches have become a well-accepted method to enhance transdermal delivery of different water-soluble drugs and macromolecules, a combination of the thermal-assisted approach with thermoresponsive LCs membranes will have the potential to improve PDDS applications but still poses a great challenge.
Collapse
Affiliation(s)
- Shan-Yang Lin
- Laboratory of Pharmaceutics and Biopharmaceutics, Department of Biotechnology and Pharmaceutical Technology, Yuanpei University of Medical Technology, No.306, Yuanpei Street, Hsin Chu 30015, Taiwan.
| |
Collapse
|
29
|
Ferreira LL, Cervantes M, Froufe HJC, Egas C, Cunha-Oliveira T, Sassone-Corsi P, Oliveira PJ. Doxorubicin persistently rewires cardiac circadian homeostasis in mice. Arch Toxicol 2019; 94:257-271. [PMID: 31768571 DOI: 10.1007/s00204-019-02626-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/13/2019] [Indexed: 12/29/2022]
Abstract
Circadian rhythms disruption can be the cause of chronic diseases. External cues, including therapeutic drugs, have been shown to modulate peripheral-circadian clocks. Since anthracycline cardiotoxicity is associated with loss of mitochondrial function and metabolic remodeling, we investigated whether the energetic failure induced by sub-chronic doxorubicin (DOX) treatment in juvenile mice was associated with persistent disruption of circadian regulators. Juvenile C57BL/6J male mice were subjected to a sub-chronic DOX treatment (4 weekly injections of 5 mg/kg DOX) and several cardiac parameters, as well as circadian-gene expression and acetylation patterns, were analyzed after 6 weeks of recovery time. Complementary experiments were performed with Mouse Embryonic Fibroblasts (MEFs) and Human Embryonic Kidney 293 cells. DOX-treated juvenile mice showed cardiotoxicity markers and persistent alterations of transcriptional- and signaling cardiac circadian homeostasis. The results showed a delayed influence of DOX on gene expression, accompanied by changes in SIRT1-mediated cyclic deacetylation. The mechanism behind DOX interference with the circadian clock was further studied in vitro, in which were observed alterations of circadian-gene expression and increased BMAL1 SIRT1-mediated deacetylation. In conclusion, DOX treatment in juvenile mice resulted in disruption of oscillatory molecular mechanisms including gene expression and acetylation profiles.
Collapse
Affiliation(s)
- Luciana L Ferreira
- Mitochondrial Toxicology and Experimental Therapeutics Laboratory (MitoXT), CNC, Center for Neuroscience and Cell Biology, University of Coimbra, UC Biotech Building (Lote 8A), Biocant Park, 3060-197, Cantanhede, Portugal
| | - Marlene Cervantes
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, University of California, Irvine, CA, 92697, USA
| | - Hugo J C Froufe
- Next Generation Sequencing Unit, Biocant, Biocant Park, Núcleo 04, Lote 8, Cantanhede, Portugal
| | - Conceição Egas
- Mitochondrial Toxicology and Experimental Therapeutics Laboratory (MitoXT), CNC, Center for Neuroscience and Cell Biology, University of Coimbra, UC Biotech Building (Lote 8A), Biocant Park, 3060-197, Cantanhede, Portugal.,Next Generation Sequencing Unit, Biocant, Biocant Park, Núcleo 04, Lote 8, Cantanhede, Portugal
| | - Teresa Cunha-Oliveira
- Mitochondrial Toxicology and Experimental Therapeutics Laboratory (MitoXT), CNC, Center for Neuroscience and Cell Biology, University of Coimbra, UC Biotech Building (Lote 8A), Biocant Park, 3060-197, Cantanhede, Portugal
| | - Paolo Sassone-Corsi
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, University of California, Irvine, CA, 92697, USA
| | - Paulo J Oliveira
- Mitochondrial Toxicology and Experimental Therapeutics Laboratory (MitoXT), CNC, Center for Neuroscience and Cell Biology, University of Coimbra, UC Biotech Building (Lote 8A), Biocant Park, 3060-197, Cantanhede, Portugal. .,Institute for Interdisciplinary Research (I.I.I.), University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
30
|
Liebert A, Bicknell B, Johnstone DM, Gordon LC, Kiat H, Hamblin MR. "Photobiomics": Can Light, Including Photobiomodulation, Alter the Microbiome? Photobiomodul Photomed Laser Surg 2019; 37:681-693. [PMID: 31596658 PMCID: PMC6859693 DOI: 10.1089/photob.2019.4628] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 06/18/2019] [Indexed: 12/14/2022] Open
Abstract
Objective: The objective of this review is to consider the dual effects of microbiome and photobiomodulation (PBM) on human health and to suggest a relationship between these two as a novel mechanism. Background: PBM describes the use of low levels of visible or near-infrared (NIR) light to heal and stimulate tissue, and to relieve pain and inflammation. In recent years, PBM has been applied to the head as an investigative approach to treat diverse brain diseases such as stroke, traumatic brain injury (TBI), Alzheimer's and Parkinson's diseases, and psychiatric disorders. Also, in recent years, increasing attention has been paid to the total microbial population that colonizes the human body, chiefly in the gut and the mouth, called the microbiome. It is known that the composition and health of the gut microbiome affects many diseases related to metabolism, obesity, cardiovascular disorders, autoimmunity, and even brain disorders. Materials and methods: A literature search was conducted for published reports on the effect of light on the microbiome. Results: Recent work by our research group has demonstrated that PBM (red and NIR light) delivered to the abdomen in mice, can alter the gut microbiome in a potentially beneficial way. This has also now been demonstrated in human subjects. Conclusions: In consideration of the known effects of PBM on metabolomics, and the now demonstrated effects of PBM on the microbiome, as well as other effects of light on the microbiome, including modulating circadian rhythms, the present perspective introduces a new term "photobiomics" and looks forward to the application of PBM to influence the microbiome in humans. Some mechanisms by which this phenomenon might occur are considered.
Collapse
Affiliation(s)
- Ann Liebert
- Australasian Research Institute, Wahroonga, Australia
- Department of Medicine, University of Sydney, Camperdown, Australia
| | - Brian Bicknell
- Faculty of Health Sciences, Australian Catholic University, North Sydney, Australia
| | | | - Luke C. Gordon
- Discipline of Physiology, University of Sydney, Camperdown, Australia
| | - Hosen Kiat
- Faculty of Medicine and Health Sciences, Macquarie University, Marsfield, Australia
- Faculty of Medicine, University of New South Wales, Kensington, Australia
| | - Michael R. Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts
- Department of Dermatology, Harvard Medical School, Boston, Massachusetts
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts
| |
Collapse
|
31
|
Chellappa SL, Vujovic N, Williams JS, Scheer FAJL. Impact of Circadian Disruption on Cardiovascular Function and Disease. Trends Endocrinol Metab 2019; 30:767-779. [PMID: 31427142 PMCID: PMC6779516 DOI: 10.1016/j.tem.2019.07.008] [Citation(s) in RCA: 177] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/07/2019] [Accepted: 07/08/2019] [Indexed: 10/26/2022]
Abstract
The circadian system, that is ubiquitous across species, generates ∼24 h rhythms in virtually all biological processes, and allows them to anticipate and adapt to the 24 h day/night cycle, thus ensuring optimal physiological function. Epidemiological studies show time-of-day variations in adverse cardiovascular (CV) events, and controlled laboratory studies demonstrate a circadian influence on key markers of CV function and risk. Furthermore, circadian misalignment, that is typically experienced by shift workers as well as by individuals who experience late eating, (social) jet lag, or circadian rhythm sleep-wake disturbances, increases CV risk factors. Therefore, understanding the mechanisms by which the circadian system regulates CV function, and which of these are affected by circadian disruption, may help to develop intervention strategies to mitigate CV risk.
Collapse
Affiliation(s)
- Sarah L Chellappa
- Medical Chronobiology Program, Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, MA 02115, USA; Division of Sleep Medicine, Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Nina Vujovic
- Medical Chronobiology Program, Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, MA 02115, USA; Division of Sleep Medicine, Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Jonathan S Williams
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Frank A J L Scheer
- Medical Chronobiology Program, Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, MA 02115, USA; Division of Sleep Medicine, Department of Medicine, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
32
|
Villanueva JE, Livelo C, Trujillo AS, Chandran S, Woodworth B, Andrade L, Le HD, Manor U, Panda S, Melkani GC. Time-restricted feeding restores muscle function in Drosophila models of obesity and circadian-rhythm disruption. Nat Commun 2019; 10:2700. [PMID: 31221967 PMCID: PMC6586848 DOI: 10.1038/s41467-019-10563-9] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 05/16/2019] [Indexed: 12/19/2022] Open
Abstract
Pathological obesity can result from genetic predisposition, obesogenic diet, and circadian rhythm disruption. Obesity compromises function of muscle, which accounts for a majority of body mass. Behavioral intervention that can counteract obesity arising from genetic, diet or circadian disruption and can improve muscle function holds untapped potential to combat the obesity epidemic. Here we show that Drosophila melanogaster (fruit fly) subject to obesogenic challenges exhibits metabolic disease phenotypes in skeletal muscle; sarcomere disorganization, mitochondrial deformation, upregulation of Phospho-AKT level, aberrant intramuscular lipid infiltration, and insulin resistance. Imposing time-restricted feeding (TRF) paradigm in which flies were fed for 12 h during the day counteracts obesity-induced dysmetabolism and improves muscle performance by suppressing intramuscular fat deposits, Phospho-AKT level, mitochondrial aberrations, and markers of insulin resistance. Importantly, TRF was effective even in an irregular lighting schedule mimicking shiftwork. Hence, TRF is an effective dietary intervention for combating metabolic dysfunction arising from multiple causes.
Collapse
Affiliation(s)
- Jesús E Villanueva
- Department of Biology, Molecular Biology Institute and Heart Institute, San Diego State University, San Diego, CA, 92182, USA
| | - Christopher Livelo
- Department of Biology, Molecular Biology Institute and Heart Institute, San Diego State University, San Diego, CA, 92182, USA
| | - Adriana S Trujillo
- Department of Biology, Molecular Biology Institute and Heart Institute, San Diego State University, San Diego, CA, 92182, USA
| | - Sahaana Chandran
- Department of Biology, Molecular Biology Institute and Heart Institute, San Diego State University, San Diego, CA, 92182, USA
| | - Brendon Woodworth
- Department of Biology, Molecular Biology Institute and Heart Institute, San Diego State University, San Diego, CA, 92182, USA
| | - Leo Andrade
- Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Hiep D Le
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Uri Manor
- Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Satchidananda Panda
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Girish C Melkani
- Department of Biology, Molecular Biology Institute and Heart Institute, San Diego State University, San Diego, CA, 92182, USA.
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA.
| |
Collapse
|
33
|
Khushhal A, Nichols S, Carroll S, Abt G, Ingle L. Insufficient exercise intensity for clinical benefit? Monitoring and quantification of a community-based Phase III cardiac rehabilitation programme: A United Kingdom perspective. PLoS One 2019; 14:e0217654. [PMID: 31194759 PMCID: PMC6563987 DOI: 10.1371/journal.pone.0217654] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/15/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND In recent years, criticism of the percentage range approach for individualised exercise prescription has intensified and we were concerned that sub-optimal exercise dose (especially intensity) may be in part responsible for the variability in the effectiveness of cardiac rehabilitation (CR) programmes in the United Kingdom (UK). The aim was to investigate the fidelity of a structured Phase III CR programme, by monitoring and quantifying exercise training intensity. DESIGN Observational study. METHODS The programme comprised 16 sessions over 8 weeks, where patients undertook an interval, circuit training approach within national guidelines for exercise prescription (40-70% heart rate reserve [HRR]). All patients wore an Apple Watch (Series 0 or 2, Watch OS2.0.1, Apple Inc., California, USA). We compared the mean % heart rate reserve (%HRR) achieved during the cardiovascular training component (%HRR-CV) of a circuit-based programme, with the %HRR during the active recovery phases (%HRR-AR) in a randomly selected cohort of patients attending standard CR. We then compared the mean %HRR-CV achieved with the minimal exercise intensity threshold during supervised exercise (40% HRR) recommended by national governing bodies. RESULTS Thirty cardiac patients (83% male; mean age [SD] 67 [10] years; BMI 28.3 [4.6] kg∙m-2) were recruited. We captured 332 individual training sessions. The mean %HRR-CV and %HRR-AR were 37 (10) %, and 31 (13) %, respectively. There was weak evidence to support the alternative hypothesis of a difference between the %HRR-CV and 40% HRR. There was very strong evidence to accept the alternative hypothesis that the mean %HRR-AR was lower than the mean %HRR-CV, median standardised effect size 1.1 (95%CI: 0.563 to 1.669), with a moderate to large effect. CONCLUSION Mean exercise training intensity was below the lower limit of the minimal training intensity guidelines for a Phase III CR programme. These findings may be in part responsible for previous reports highlighting the significant variability in effectiveness of UK CR services and poor CRF improvements observed from several prior investigations.
Collapse
Affiliation(s)
- Alaa Khushhal
- Department of Sport, Health & Exercise Science, University of Hull, Kingston-upon-Hull, United Kingdom
| | - Simon Nichols
- Centre for Sport and Exercise Science, Sheffield Hallam University, Sheffield, United Kingdom
| | - Sean Carroll
- Department of Sport, Health & Exercise Science, University of Hull, Kingston-upon-Hull, United Kingdom
| | - Grant Abt
- Department of Sport, Health & Exercise Science, University of Hull, Kingston-upon-Hull, United Kingdom
| | - Lee Ingle
- Department of Sport, Health & Exercise Science, University of Hull, Kingston-upon-Hull, United Kingdom
| |
Collapse
|
34
|
Alibhai FJ, Reitz CJ, Peppler WT, Basu P, Sheppard P, Choleris E, Bakovic M, Martino TA. Female ClockΔ19/Δ19 mice are protected from the development of age-dependent cardiomyopathy. Cardiovasc Res 2019; 114:259-271. [PMID: 28927226 DOI: 10.1093/cvr/cvx185] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 09/08/2017] [Indexed: 12/22/2022] Open
Abstract
Aims Circadian rhythms are important for healthy cardiovascular physiology and they are regulated by the molecular circadian mechanism. Previously, we showed that disruption of the circadian mechanism factor CLOCK in male ClockΔ19/Δ19 mice led to development of age-dependent cardiomyopathy. Here, we investigate the role of biological sex in protecting against heart disease in aging female ClockΔ19/Δ19 mice. Methods and results Female ClockΔ19/Δ19 mice are protected from the development of cardiomyopathy with age, as heart structure and function are similar to 18 months of age vs. female WT mice. We show that female ClockΔ19/Δ19 mice maintain normal glucose tolerance as compared with female WT. Tissue metabolic profiling revealed that aging female ClockΔ19/Δ19 mice maintain normal cardiac glucose uptake, whereas the male ClockΔ19/Δ19 mice have increased cardiac glucose uptake consistent with pathological remodelling. Shotgun lipidomics revealed differences in phospholipids that were sex and genotype specific, including cardiolipin CL76:11 that was increased and CL72:8 that was decreased in male ClockΔ19/Δ19 mice. Additionally, female ClockΔ19/Δ19 mice show increased activation of AKT signalling and preserved cytochrome c oxidase activity compared with male ClockΔ19/Δ19 mice, which can help to explain why they are protected from heart disease. To determine how this protection occurs in females even with the Clock mutation, we examined the effects of ovarian hormones. We show that ovarian hormones protect female ClockΔ19/Δ19 mice from heart disease as ovariectomized female ClockΔ19/Δ19 mice develop cardiac dilation, glucose intolerance and reduced cardiac cytochrome c oxidase; this phenotype is consistent with the age-dependent decline observed in male ClockΔ19/Δ19 mice. Conclusions These data demonstrate that ovarian hormones protect female ClockΔ19/Δ19 mice from the development of age-dependent cardiomyopathy even though Clock function is disturbed. Understanding the interaction of biological sex and the circadian mechanism in cardiac growth, renewal and remodelling opens new doors for understanding and treating heart disease.
Collapse
Affiliation(s)
- Faisal J Alibhai
- Department of Biomedical Sciences/OVC, Centre for Cardiovascular Investigations, University of Guelph, Room 1646B, University of Guelph, Guelph, ON N1G2W1, Canada
| | - Cristine J Reitz
- Department of Biomedical Sciences/OVC, Centre for Cardiovascular Investigations, University of Guelph, Room 1646B, University of Guelph, Guelph, ON N1G2W1, Canada
| | - Willem T Peppler
- Human Health and Nutritional Sciences, , University of Guelph, Room 1646B, University of Guelph, Guelph, ON N1G2W1, Canada
| | - Poulami Basu
- Human Health and Nutritional Sciences, , University of Guelph, Room 1646B, University of Guelph, Guelph, ON N1G2W1, Canada
| | - Paul Sheppard
- Department of Psychology and Neuroscience Program, University of Guelph, Room 1646B, University of Guelph, Guelph, ON N1G2W1, Canada
| | - Elena Choleris
- Department of Psychology and Neuroscience Program, University of Guelph, Room 1646B, University of Guelph, Guelph, ON N1G2W1, Canada
| | - Marica Bakovic
- Human Health and Nutritional Sciences, , University of Guelph, Room 1646B, University of Guelph, Guelph, ON N1G2W1, Canada
| | - Tami A Martino
- Department of Biomedical Sciences/OVC, Centre for Cardiovascular Investigations, University of Guelph, Room 1646B, University of Guelph, Guelph, ON N1G2W1, Canada
| |
Collapse
|
35
|
Duong ATH, Reitz CJ, Louth EL, Creighton SD, Rasouli M, Zwaiman A, Kroetsch JT, Bolz SS, Winters BD, Bailey CDC, Martino TA. The Clock Mechanism Influences Neurobiology and Adaptations to Heart Failure in Clock ∆19/∆19 Mice With Implications for Circadian Medicine. Sci Rep 2019; 9:4994. [PMID: 30899044 PMCID: PMC6428811 DOI: 10.1038/s41598-019-41469-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 03/05/2019] [Indexed: 02/07/2023] Open
Abstract
In this study we investigated the role of the circadian mechanism on cognition-relevant brain regions and neurobiological impairments associated with heart failure (HF), using murine models. We found that the circadian mechanism is an important regulator of healthy cognitive system neurobiology. Normal Clock∆19/∆19 mice had neurons with smaller apical dendrite trees in the medial prefrontal cortex (mPFC), and hippocampus, showed impaired visual-spatial memory, and exhibited lower cerebrovascular myogenic tone, versus wild types (WT). We then used the left anterior descending coronary artery ligation model to investigate adaptations in response to HF. Intriguingly, adaptations to neuron morphology, memory, and cerebrovascular tone occurred in differing magnitude and direction between Clock∆19/∆19 and WT mice, ultimately converging in HF. To investigate this dichotomous response, we performed microarrays and found genes crucial for growth and stress pathways that were altered in Clock∆19/∆19 mPFC and hippocampus. Thus these data demonstrate for the first time that (i) the circadian mechanism plays a role in neuron morphology and function; (ii) there are changes in neuron morphology and function in HF; (iii) CLOCK influences neurobiological gene adaptations to HF at a cellular level. These findings have clinical relevance as patients with HF often present with concurrent neurocognitive impairments. There is no cure for HF, and new understanding is needed to reduce morbidity and improve the quality of life for HF patients.
Collapse
Affiliation(s)
- Austin T H Duong
- Centre for Cardiovascular Investigations, Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Cristine J Reitz
- Centre for Cardiovascular Investigations, Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Emma L Louth
- Centre for Cardiovascular Investigations, Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | | | - Mina Rasouli
- Centre for Cardiovascular Investigations, Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Ashley Zwaiman
- Centre for Cardiovascular Investigations, Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Jeffrey T Kroetsch
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | | | - Boyer D Winters
- Department of Psychology, University of Guelph, Guelph, Ontario, Canada
| | - Craig D C Bailey
- Centre for Cardiovascular Investigations, Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada.
| | - Tami A Martino
- Centre for Cardiovascular Investigations, Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada.
| |
Collapse
|
36
|
Zhao X, Fan J, Wu P, Wei C, Chen Q, Ming Z, Yan J, Yang L. Chronic chemotherapy with paclitaxel nanoparticles induced apoptosis in lung cancer in vitro and in vivo. Int J Nanomedicine 2019; 14:1299-1309. [PMID: 30863062 PMCID: PMC6391150 DOI: 10.2147/ijn.s188049] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Aim Paclitaxel (PTX) is an effective antitumor drug. Previous research demonstrated that paclitaxel nanoparticles (PTX-NPs) exhibited the greatest antitumor effect at 15 hours after light onset (15 HALO), but the mechanism in chronic chemotherapy is still unknown. In our study, we investigated whether PTX-NPs regulated Period2 (Per2) during chronic chemotherapy to induce apoptosis in vivo and in vitro. Methods To improve the antitumor effect and reduce organ damage induced by PTX treatment, PTX-NPs were prepared using a film dispersion method. Then, A549 cells were treated with PTX-NPs at 0, 5, 10, 15, and 20 HALO. An annexin/PI V-FITC apoptosis kit was measured for apoptosis, and PI was analyzed for cell cycle. The relative mechanism was detected by RT-PCR and Western blotting. Tumor volume and weight were measured to evaluate the antitumor effect of the PTX-NPs, and H&E staining was performed to assess organ damage. Results Cell cycle analysis demonstrated that PTX-NPs blocked cell cycle in G2 phase and that the ratio of cell death was significantly increased in A549 cells, while the ratios of cells in G2 phase and of apoptotic cells were highest at 15 HALO. Evaluation of in vivo antitumor activity revealed that PTX-NPs inhibited tumor growth and decreased tumor weight at 15 HALO. RT-PCR and Western blotting demonstrated that PTX-NPs upregulated Per2 mRNA and protein expression, and the highest Per2 expression was observed at 15 HALO in vivo and in vitro. Meanwhile, Bax mRNA and protein expression was upregulated, while Bcl-2 mRNA and protein expression was downregulated after PTX-NPs treatment in vivo. Moreover, H&E staining revealed that PTX-NPs reduced liver damage at 15 HALO. Conclusion PTX-NPs exhibited the most effective antitumor activity and reduced liver damage at 15 HALO through upregulation of Per2 expression to induce apoptosis in vivo and in vitro.
Collapse
Affiliation(s)
- Xuefeng Zhao
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China, .,Department of Oncology, Zigong 1st People's Hospital, Zigong 643000, China,
| | - Juan Fan
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China,
| | - Peng Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China,
| | - Chengming Wei
- Department of Oncology, Zigong 1st People's Hospital, Zigong 643000, China,
| | - Qiongying Chen
- Department of Oncology, Zigong 3rd People's Hospital, Zigong 643000, China
| | - Zhi Ming
- Department of Oncology, Zigong 1st People's Hospital, Zigong 643000, China,
| | - Jun Yan
- Department of Oncology, Zigong 1st People's Hospital, Zigong 643000, China,
| | - Linglin Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China,
| |
Collapse
|
37
|
|
38
|
Glen Pyle W, Martino TA. Circadian rhythms influence cardiovascular disease differently in males and females: role of sex and gender. CURRENT OPINION IN PHYSIOLOGY 2018. [DOI: 10.1016/j.cophys.2018.05.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
39
|
Khaper N, Bailey CDC, Ghugre NR, Reitz C, Awosanmi Z, Waines R, Martino TA. Implications of disturbances in circadian rhythms for cardiovascular health: A new frontier in free radical biology. Free Radic Biol Med 2018; 119:85-92. [PMID: 29146117 DOI: 10.1016/j.freeradbiomed.2017.11.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 10/27/2017] [Accepted: 11/08/2017] [Indexed: 01/19/2023]
Abstract
Cell autonomous circadian "clock" mechanisms are present in virtually every organ, and generate daily rhythms that are important for normal physiology. This is especially relevant to the cardiovascular system, for example the circadian mechanism orchestrates rhythms in heart rate, blood pressure, cardiac contractility, metabolism, gene and protein abundance over the 24-h day and night cycles. Conversely, disturbing circadian rhythms (e.g. via shift work, sleep disorders) increases cardiovascular disease risk, and exacerbates cardiac remodelling and worsens outcome. Notably, reactive oxygen species (ROS) are important contributors to heart disease, especially the pathophysiologic damage that occurs after myocardial infarction (MI, heart attack). However, little is known about how the circadian mechanism, or rhythm desynchrony, is involved in these key pathologic stress responses. This review summarizes the current knowledge on circadian rhythms in the cardiovascular system, and the implications of rhythm disturbances for cardiovascular health. Furthermore, we highlight how free radical biology coincides with the pathogenesis of myocardial repair and remodelling after MI, and indicate a role for the circadian system in the oxidative stress pathways in the heart and brain after MI. This fusion of circadian biology with cardiac oxidative stress pathways is novel, and offers enormous potential for improving our understanding and treatment of heart disease.
Collapse
Affiliation(s)
- Neelam Khaper
- Medical Sciences Division, Northern Ontario School of Medicine, Lakehead University, 955 Oliver Road, Thunder Bay, Ontario, Canada P7B5E1
| | - Craig D C Bailey
- Centre for Cardiovascular Investigations, Department of Biomedical Sciences/OVC, University of Guelph, Guelph, Ontario, Canada N1G2W1
| | - Nilesh R Ghugre
- Schulich Heart Research Program, Sunnybrook Research Institute, Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada M4N 3M5
| | - Cristine Reitz
- Centre for Cardiovascular Investigations, Department of Biomedical Sciences/OVC, University of Guelph, Guelph, Ontario, Canada N1G2W1
| | - Zikra Awosanmi
- Centre for Cardiovascular Investigations, Department of Biomedical Sciences/OVC, University of Guelph, Guelph, Ontario, Canada N1G2W1
| | - Ryan Waines
- Centre for Cardiovascular Investigations, Department of Biomedical Sciences/OVC, University of Guelph, Guelph, Ontario, Canada N1G2W1
| | - Tami A Martino
- Centre for Cardiovascular Investigations, Department of Biomedical Sciences/OVC, University of Guelph, Guelph, Ontario, Canada N1G2W1.
| |
Collapse
|
40
|
Altered Circadian Timing System-Mediated Non-Dipping Pattern of Blood Pressure and Associated Cardiovascular Disorders in Metabolic and Kidney Diseases. Int J Mol Sci 2018; 19:ijms19020400. [PMID: 29385702 PMCID: PMC5855622 DOI: 10.3390/ijms19020400] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 01/12/2018] [Accepted: 01/20/2018] [Indexed: 12/15/2022] Open
Abstract
The morning surge in blood pressure (BP) coincides with increased cardiovascular (CV) events. This strongly suggests that an altered circadian rhythm of BP plays a crucial role in the development of CV disease (CVD). A disrupted circadian rhythm of BP, such as the non-dipping type of hypertension (i.e., absence of nocturnal BP decline), is frequently observed in metabolic disorders and chronic kidney disease (CKD). The circadian timing system, controlled by the central clock in the suprachiasmatic nucleus of the hypothalamus and/or by peripheral clocks in the heart, vasculature, and kidneys, modulates the 24 h oscillation of BP. However, little information is available regarding the molecular and cellular mechanisms of an altered circadian timing system-mediated disrupted dipping pattern of BP in metabolic disorders and CKD that can lead to the development of CV events. A more thorough understanding of this pathogenesis could provide novel therapeutic strategies for the management of CVD. This short review will address our and others' recent findings on the molecular mechanisms that may affect the dipping pattern of BP in metabolic dysfunction and kidney disease and its association with CV disorders.
Collapse
|
41
|
Lewis R, Hackfort BT, Schultz HD. Chronic Heart Failure Abolishes Circadian Rhythms in Resting and Chemoreflex Breathing. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1071:129-136. [PMID: 30357743 DOI: 10.1007/978-3-319-91137-3_16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Physiological systems often display 24 h rhythms that vary with the light/dark cycle. Disruption of circadian physiological rhythms have been linked to the progression of various cardiovascular diseases, and advances in the understanding of these rhythms have led to novel interventions and improved clinical outcomes. Although respiratory function has been known to vary between the light and dark periods, circadian rhythms in breathing have been understudied in clinical conditions. In the current study, we have begun to assess light/dark variations in respiration in chronic heart failure (CHF), a condition associated with abnormal resting and chemoreflex breathing as well as exercise intolerance. CHF was induced using coronary artery ligation and verified using echocardiography. Sham animals underwent a thoracotomy without coronary artery ligation. Tidal volume, respiratory frequency, and minute ventilation were all determined by whole body plethysmography under resting conditions and in response to chemoreflex challenges during the light and dark periods. Light/dark differences in voluntary exercise were assessed using a running wheel. The sham control group showed light/dark differences in resting and chemoreflex breathing, as well as arterial pressure, and these effects were eliminated in the CHF group. Both groups completed more rotations on the running wheel during the dark period compared to during the light period. The data suggest that CHF disrupts cardiovascular and respiratory circadian rhythms.
Collapse
Affiliation(s)
- Robert Lewis
- A.T. Still University School of Osteopathic Medicine, Mesa, AZ, USA.
| | - Bryan T Hackfort
- Department of Cellular and Integrative Physiology at the University of Nebraska Medical Center, Omaha, NE, USA
| | - Harold D Schultz
- Department of Cellular and Integrative Physiology at the University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
42
|
Chen Z, Yoo SH, Takahashi JS. Development and Therapeutic Potential of Small-Molecule Modulators of Circadian Systems. Annu Rev Pharmacol Toxicol 2017; 58:231-252. [PMID: 28968186 DOI: 10.1146/annurev-pharmtox-010617-052645] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Circadian timekeeping systems drive oscillatory gene expression to regulate essential cellular and physiological processes. When the systems are perturbed, pathological consequences ensue and disease risks rise. A growing number of small-molecule modulators have been reported to target circadian systems. Such small molecules, identified via high-throughput screening or derivatized from known scaffolds, have shown promise as drug candidates to improve biological timing and physiological outputs in disease models. In this review, we first briefly describe the circadian system, including the core oscillator and the cellular networks. Research progress on clock-modulating small molecules is presented, focusing on development strategies and biological efficacies. We highlight the therapeutic potential of small molecules in clock-related pathologies, including jet lag and shiftwork; various chronic diseases, particularly metabolic disease; and aging. Emerging opportunities to identify and exploit clock modulators as novel therapeutic agents are discussed.
Collapse
Affiliation(s)
- Zheng Chen
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA;
| | - Seung-Hee Yoo
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA;
| | - Joseph S Takahashi
- Department of Neuroscience and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
43
|
|
44
|
Boltze J, Nitzsche F, Jolkkonen J, Weise G, Pösel C, Nitzsche B, Wagner DC. Concise Review: Increasing the Validity of Cerebrovascular Disease Models and Experimental Methods for Translational Stem Cell Research. Stem Cells 2017; 35:1141-1153. [DOI: 10.1002/stem.2595] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 02/06/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Johannes Boltze
- Department of Translational Medicine and Cell Technology; Fraunhofer Research Institution for Marine Biotechnology and Cell Technology; Lübeck Germany
- Institute for Medical and Marine Biotechnology, University of Lübeck; Lübeck Germany
| | - Franziska Nitzsche
- Department of Cell Therapy; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
- Department of Radiology; McGowan Institute for Regenerative Medicine, University of Pittsburgh; Pennsylvania USA
| | - Jukka Jolkkonen
- Department of Neurology; Institute of Clinical Medicine, University of Eastern Finland; Kuopio Finland
| | - Gesa Weise
- Department of Cell Therapy; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
- Department of Neurology; University of Leipzig; Germany
| | - Claudia Pösel
- Department of Cell Therapy; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
| | - Björn Nitzsche
- Department of Cell Therapy; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
- Department of Nuclear Medicine; University Hospital Leipzig; Germany
| | - Daniel-Christoph Wagner
- Department of Cell Therapy; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
- Institute of Pathology, University Medical Center Mainz; Germany
| |
Collapse
|
45
|
Fan BS, Zhang EH, Cheng MH, Wu ZT, Han B, Yu JG. Diurnal Variation of the Peripheral Cholinergic Antiinflammatory Function in Mice. CNS Neurosci Ther 2016; 22:764-70. [PMID: 27306582 DOI: 10.1111/cns.12578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 05/03/2016] [Accepted: 05/13/2016] [Indexed: 11/28/2022] Open
Abstract
AIMS Cholinergic antiinflammatory (CAI) pathway functions importantly in inflammation via α7 nicotinic acetylcholine receptors (α7nAChR). The present work tested circadian rhythm in peripheral CAI activity and validities of CAI activity and glucocorticoids in chronotherapy for lipopolysaccharide (LPS)-induced shock. METHODS Vesicular acetylcholine transporter (VAChT) expressed in liver and kidney was examined every 3 h in C57BL/6 mice. Proinflammatory cytokines in serum and survival time in shock were monitored after LPS injection every 3 h. Mifepristone, antagonist of glucocorticoid receptors, and methyllycaconitine (MLA), antagonist of α7nAChR, were administrated before LPS to block antiinflammatory function of endogenous glucocorticoids and acetylcholine. RESULTS Both levels of tumor necrosis factor α, interleukin 1β, and interleukin 6 and mortality exhibited diurnal variations with prominent peaks when LPS was given at 15:00, and the minimum mortality occurred at 00:00. Expression of VAChT increased during resting period. MLA increased serum proinflammatory cytokines slightly, but not affected survival rate. Both differences in cytokines and in survival times between LPS injection at 15:00 and 00:00 were eliminated by mifepristone, but not by MLA. CONCLUSION Peripheral CAI pathway exerts more powerful antiinflammatory effect during resting period. Glucocorticoids appear to be efficient in chronotherapy for septic shock.
Collapse
Affiliation(s)
- Bo-Shi Fan
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| | | | - Ming-He Cheng
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| | - Zhao-Tang Wu
- Department of Physiology, Second Military Medical University, Shanghai, China
| | - Bing Han
- Department of Pharmacy, Minhang Hospital, Fudan University, Shanghai, China
| | - Jian-Guang Yu
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| |
Collapse
|
46
|
Liao D, Xu Y, Zou R, Wu L, Luo X, Li F, Lin P, Wang X, Xie Z, Wang C. The circadian rhythm of syncopal episodes in patients with neurally mediated syncope. Int J Cardiol 2016; 215:186-92. [PMID: 27128529 DOI: 10.1016/j.ijcard.2016.04.086] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 04/11/2016] [Indexed: 10/21/2022]
Abstract
OBJECTIVE To explore the circadian rhythm of neurally mediated syncope (NMS). METHODS 411 patients with NMS (165 males and 246 females aged from 3 to 68years) were included in the study. All subjects underwent head-up tilt test (HUTT) and were carefully asked about the number of syncopal attacks and the periods (morning 06:00am-12:00am, afternoon 12:00am-18:00pm, evening 18:00pm-24:00pm, night 00:00am-06:00am) in which episodes occurred in. RESULTS (1) Syncopal attacks of all patients tended to occur in the morning (P=0.010); there was a statistical difference in the frequency of episodes in four periods through the day in HUTT positive patients (P=0.001), but there was no significant change of episodes within a day in HUTT negative group; and there was no statistical difference in circadian syncope distribution between HUTT negative and HUTT positive group or among patients with different HUTT responses (the orthostatic hypotension (OH) and orthostatic hypertension (OHT) patients were excluded). (2) The syncopal attacks of morning hours occurred more in males than females, but the episodes in the evening occurred more in females than males (P=0.034). (3) The younger the patients were, the chance of syncopal attacks in the morning increased; the older the patients were, they may have more episodes at night (P<0.001). CONCLUSIONS A distinct circadian variation in the frequency of syncopal episodes exists, with a peak in the morning, and there were statistical differences in circadian rhythm of syncopal episodes regarding gender and age.
Collapse
Affiliation(s)
- Donglei Liao
- Department of Pediatric Cardiovasology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Institute of Pediatrics of Central South University, Changsha 410011, Hunan, China
| | - Yi Xu
- Department of Pediatric Cardiovasology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Institute of Pediatrics of Central South University, Changsha 410011, Hunan, China
| | - Runmei Zou
- Department of Pediatric Cardiovasology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Institute of Pediatrics of Central South University, Changsha 410011, Hunan, China
| | - Lijia Wu
- Department of Pediatric Cardiovasology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Institute of Pediatrics of Central South University, Changsha 410011, Hunan, China
| | - Xuemei Luo
- Department of Pediatric Cardiovasology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Institute of Pediatrics of Central South University, Changsha 410011, Hunan, China
| | - Fang Li
- Department of Pediatric Cardiovasology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Institute of Pediatrics of Central South University, Changsha 410011, Hunan, China
| | - Ping Lin
- Department of Pediatric Cardiovasology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Institute of Pediatrics of Central South University, Changsha 410011, Hunan, China
| | - Xiuying Wang
- Department of Pediatric Cardiovasology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Institute of Pediatrics of Central South University, Changsha 410011, Hunan, China
| | - Zhenwu Xie
- Department of Pediatric Cardiovasology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Institute of Pediatrics of Central South University, Changsha 410011, Hunan, China
| | - Cheng Wang
- Department of Pediatric Cardiovasology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Institute of Pediatrics of Central South University, Changsha 410011, Hunan, China.
| |
Collapse
|
47
|
Garbarino S, Magnavita N. Work Stress and Metabolic Syndrome in Police Officers. A Prospective Study. PLoS One 2015; 10:e0144318. [PMID: 26641879 PMCID: PMC4671563 DOI: 10.1371/journal.pone.0144318] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 11/15/2015] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE The aim of this longitudinal study was to evaluate the association between occupational stress and metabolic syndrome (MetS) in a rapid response police unit. METHOD Work-related stress was continuously monitored during the 5-year period with both the Demand-Control-Support (DCS) and the Effort-Reward Imbalance (ERI) models. Blood pressure, body mass index (BMI), waist circumference, triglycerides, HDL-cholesterol, and fasting blood glucose were measured at baseline in January 2009, and in January 2014. 234 out of 290 police officers (81%) completed the follow-up. RESULTS The majority of police officers had high stress levels. At follow-up, police officers in the highest quartile of stress had significantly higher mean levels of triglycerides, and lower levels of HDL-cholesterol than their colleagues in the lowest quartile. Police officers with high stress had an increased adjusted risk of developing MetS (aOR = 2.68; CI95% = 1.08-6.70), and hypertriglyceridemia (aOR = 7.86; CI95 = 1.29-48.04). Demand and Effort were significant predictors of MetS. CONCLUSION Our study supports the hypothesis that work-related stress induces MetS, particularly through its effects on blood lipids. Future longitudinal studies with continuous monitoring of stress levels will definitively confirm this hypothesis.
Collapse
Affiliation(s)
- Sergio Garbarino
- State Police Health Service Department, Ministry of the Interior, Rome, Genoa, Italy
- Department of Health Sciences, University of Genoa, Genoa, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics and Maternal-Infantile Sciences (DINOGMI), Genoa, Italy
| | - Nicola Magnavita
- Department of Public Health, Università Cattolica del Sacro Cuore, Roma, Italy
| |
Collapse
|
48
|
Sundar IK, Yao H, Sellix MT, Rahman I. Circadian molecular clock in lung pathophysiology. Am J Physiol Lung Cell Mol Physiol 2015; 309:L1056-75. [PMID: 26361874 DOI: 10.1152/ajplung.00152.2015] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 09/08/2015] [Indexed: 02/06/2023] Open
Abstract
Disrupted daily or circadian rhythms of lung function and inflammatory responses are common features of chronic airway diseases. At the molecular level these circadian rhythms depend on the activity of an autoregulatory feedback loop oscillator of clock gene transcription factors, including the BMAL1:CLOCK activator complex and the repressors PERIOD and CRYPTOCHROME. The key nuclear receptors and transcription factors REV-ERBα and RORα regulate Bmal1 expression and provide stability to the oscillator. Circadian clock dysfunction is implicated in both immune and inflammatory responses to environmental, inflammatory, and infectious agents. Molecular clock function is altered by exposomes, tobacco smoke, lipopolysaccharide, hyperoxia, allergens, bleomycin, as well as bacterial and viral infections. The deacetylase Sirtuin 1 (SIRT1) regulates the timing of the clock through acetylation of BMAL1 and PER2 and controls the clock-dependent functions, which can also be affected by environmental stressors. Environmental agents and redox modulation may alter the levels of REV-ERBα and RORα in lung tissue in association with a heightened DNA damage response, cellular senescence, and inflammation. A reciprocal relationship exists between the molecular clock and immune/inflammatory responses in the lungs. Molecular clock function in lung cells may be used as a biomarker of disease severity and exacerbations or for assessing the efficacy of chronotherapy for disease management. Here, we provide a comprehensive overview of clock-controlled cellular and molecular functions in the lungs and highlight the repercussions of clock disruption on the pathophysiology of chronic airway diseases and their exacerbations. Furthermore, we highlight the potential for the molecular clock as a novel chronopharmacological target for the management of lung pathophysiology.
Collapse
Affiliation(s)
- Isaac K Sundar
- Department of Environmental Medicine, Lung Biology and Disease Program, University of Rochester Medical Center, Rochester, New York; and
| | - Hongwei Yao
- Department of Environmental Medicine, Lung Biology and Disease Program, University of Rochester Medical Center, Rochester, New York; and
| | - Michael T Sellix
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Rochester Medical Center, Rochester, New York
| | - Irfan Rahman
- Department of Environmental Medicine, Lung Biology and Disease Program, University of Rochester Medical Center, Rochester, New York; and
| |
Collapse
|
49
|
Yang G, Wang H, Zhang E. Editorial: Therapeutic implications of circadian rhythms. Front Pharmacol 2015; 6:175. [PMID: 26347653 PMCID: PMC4541160 DOI: 10.3389/fphar.2015.00175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 08/03/2015] [Indexed: 11/13/2022] Open
Affiliation(s)
- Guangrui Yang
- The Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania Philadelphia, PA, USA
| | - Han Wang
- Center for Circadian Clocks, Soochow University Suzhou, China
| | - Erquan Zhang
- National Institute of Biological Sciences Beijing, China
| |
Collapse
|