1
|
Lai Y, Liu S, Song C, Long T, Song L, Jiang M. An update on the role and mechanisms of periodontitis in cardiovascular diseases. Cell Signal 2025; 132:111770. [PMID: 40164419 DOI: 10.1016/j.cellsig.2025.111770] [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: 02/03/2025] [Revised: 03/18/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death worldwide. Despite extensive studies into the causes and therapies for CVDs, their incidence and prevalence continue to increase. Periodontitis is a multifactorial, chronic inflammatory disease related to systemic health. Current research suggests that periodontitis may be an unconventional risk factor for CVDs and it may increase the risk of CVDs such as atherosclerosis, coronary heart disease, myocardial infarction, hypertension, heart failure as well as cardiomyopathy. For all these reasons, it is quite plausible that prevention of periodontitis has an impact on the onset or progression of CVDs. Therefore, in this review, we investigated the association between periodontitis caused by oral microorganisms and different CVDs. In addition, we discuss the various mechanisms by which periodontitis contributes to the onset and progression of CVDs. Our review aims to raise global awareness of periodontitis, particularly its role in CVDs, provide a basis for the prevention and treatment of CVDs and offer potential therapeutic targets.
Collapse
Affiliation(s)
- Yuping Lai
- The Huankui Academy, Jiangxi Medical College, Nanchang University, 999 Xuefu Road, Nanchang, Jiangxi 330031, China
| | - Sibo Liu
- The Queen Mary school, Jiangxi Medical College, Nanchang University, 999 Xuefu Road, Nanchang, Jiangxi 330031, China
| | - Chenxin Song
- The Second Clinical Medical College, Jiangxi Medical College, Nanchang University, 999 Xuefu Road, Nanchang, Jiangxi 330031, China
| | - Ting Long
- Center of Stomatology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, Jiangxi Province 330000, China; The Institute of Periodontal Disease, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, Jiangxi Province 330000, China; JXHC Key Laboratory of Periodontology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, Jiangxi Province 330000, China
| | - Li Song
- Center of Stomatology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, Jiangxi Province 330000, China; The Institute of Periodontal Disease, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, Jiangxi Province 330000, China; JXHC Key Laboratory of Periodontology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, Jiangxi Province 330000, China.
| | - Meixiu Jiang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, 999 Xuefu Road, Nanchang, Jiangxi 330031, China.
| |
Collapse
|
2
|
Savvidis C, Kallistrou E, Kouroglou E, Dionysopoulou S, Gavriiloglou G, Ragia D, Tsiama V, Proikaki S, Belis K, Ilias I. Circadian rhythm disruption and endocrine-related tumors. World J Clin Oncol 2024; 15:818-834. [PMID: 39071458 PMCID: PMC11271730 DOI: 10.5306/wjco.v15.i7.818] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 06/20/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
This review delved into the intricate relationship between circadian clocks and physiological processes, emphasizing their critical role in maintaining homeostasis. Orchestrated by interlocked clock genes, the circadian timekeeping system regulates fundamental processes like the sleep-wake cycle, energy metabolism, immune function, and cell proliferation. The central oscillator in the hypothalamic suprachiasmatic nucleus synchronizes with light-dark cycles, while peripheral tissue clocks are influenced by cues such as feeding times. Circadian disruption, linked to modern lifestyle factors like night shift work, correlates with adverse health outcomes, including metabolic syndrome, cardiovascular diseases, infections, and cancer. We explored the molecular mechanisms of circadian clock genes and their impact on metabolic disorders and cancer pathogenesis. Specific associations between circadian disruption and endocrine tumors, spanning breast, ovarian, testicular, prostate, thyroid, pituitary, and adrenal gland cancers, are highlighted. Shift work is associated with increased breast cancer risk, with PER genes influencing tumor progression and drug resistance. CLOCK gene expression correlates with cisplatin resistance in ovarian cancer, while factors like aging and intermittent fasting affect prostate cancer. Our review underscored the intricate interplay between circadian rhythms and cancer, involving the regulation of the cell cycle, DNA repair, metabolism, immune function, and the tumor microenvironment. We advocated for integrating biological timing into clinical considerations for personalized healthcare, proposing that understanding these connections could lead to novel therapeutic approaches. Evidence supports circadian rhythm-focused therapies, particularly chronotherapy, for treating endocrine tumors. Our review called for further research to uncover detailed connections between circadian clocks and cancer, providing essential insights for targeted treatments. We emphasized the importance of public health interventions to mitigate lifestyle-related circadian disruptions and underscored the critical role of circadian rhythms in disease mechanisms and therapeutic interventions.
Collapse
Affiliation(s)
- Christos Savvidis
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | - Efthymia Kallistrou
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | - Eleni Kouroglou
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | - Sofia Dionysopoulou
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | | | - Dimitra Ragia
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | - Vasiliki Tsiama
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | - Stella Proikaki
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | - Konstantinos Belis
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | - Ioannis Ilias
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| |
Collapse
|
3
|
El Jamal N, Brooks TG, Cohen J, Townsend RR, Sosa GRD, Shah V, Nelson RG, Drawz PE, Rao P, Bhat Z, Chang A, Yang W, FitzGerald GA, Skarke C. Prognostic utility of rhythmic components in 24-h ambulatory blood pressure monitoring for the risk stratification of chronic kidney disease patients with cardiovascular co-morbidity. J Hum Hypertens 2024; 38:420-429. [PMID: 38212425 PMCID: PMC11076200 DOI: 10.1038/s41371-023-00884-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/23/2023] [Accepted: 12/06/2023] [Indexed: 01/13/2024]
Abstract
Chronic kidney disease (CKD) represents a significant global burden. Hypertension is a modifiable risk factor for rapid progression of CKD. We extend the risk stratification by introducing the non-parametric determination of rhythmic components in 24-h profiles of ambulatory blood pressure monitoring (ABPM) in the Chronic Renal Insufficiency Cohort (CRIC) and the African American Study for Kidney Disease and Hypertension (AASK) cohort using Cox proportional hazards models. We find that rhythmic profiling of BP through JTK_CYCLE analysis identifies subgroups of CRIC participants that were more likely to die due to cardiovascular causes. While our fully adjusted model shows a trend towards a significant association between absent cyclic components and cardiovascular death in the full CRIC cohort (HR: 1.71,95% CI: 0.99-2.97, p = 0.056), CRIC participants with a history of cardiovascular disease (CVD) and absent cyclic components in their BP profile had at any time a 3.4-times higher risk of cardiovascular death than CVD patients with cyclic components present in their BP profile (HR: 3.37, 95% CI: 1.45-7.87, p = 0.005). This increased risk was not explained by the dipping or non-dipping pattern in ABPM. Due to the large differences in patient characteristics, the results do not replicate in the AASK cohort. This study suggests rhythmic blood pressure components as a potential novel biomarker to unmask excess risk among CKD patients with prior cardiovascular disease.
Collapse
Affiliation(s)
- Nadim El Jamal
- Institute for Translational Medicine and Therapeutics (ITMAT), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Thomas G Brooks
- Institute for Translational Medicine and Therapeutics (ITMAT), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jordana Cohen
- Renal-Electrolyte and Hypertension Division, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Raymond R Townsend
- Renal-Electrolyte and Hypertension Division, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Vallabh Shah
- Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM, USA
- Department of Biochemistry, School of Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Robert G Nelson
- The Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| | - Paul E Drawz
- Division of Nephrology and Hypertension, University of Minnesota, Minneapolis, MN, USA
| | - Panduranga Rao
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Zeenat Bhat
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Alexander Chang
- Kidney Health Research Institute, Department of Population Health Sciences, Geisinger, Danville, PA, USA
| | - Wei Yang
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Garret A FitzGerald
- Institute for Translational Medicine and Therapeutics (ITMAT), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Carsten Skarke
- Institute for Translational Medicine and Therapeutics (ITMAT), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
4
|
Tyagi SC, Pushpakumar S, Sen U, Akinterinwa OE, Zheng Y, Mokshagundam SPL, Kalra DK, Singh M. Role of circadian clock system in the mitochondrial trans-sulfuration pathway and tissue remodeling. Can J Physiol Pharmacol 2024; 102:105-115. [PMID: 37979203 DOI: 10.1139/cjpp-2023-0186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2023]
Abstract
Previous studies from our laboratory revealed that the gaseous molecule hydrogen sulfide (H2S), a metabolic product of epigenetics, involves trans-sulfuration pathway for ensuring metabolism and clearance of homocysteine (Hcy) from body, thereby mitigating the skeletal muscle's pathological remodeling. Although the master circadian clock regulator that is known as brain and muscle aryl hydrocarbon receptor nuclear translocator like protein 1 (i.e., BMAL 1) is associated with S-adenosylhomocysteine hydrolase (SAHH) and Hcy metabolism but how trans-sulfuration pathway is influenced by the circadian clock remains unexplored. We hypothesize that alterations in the functioning of circadian clock during sleep and wake cycle affect skeletal muscle's biology. To test this hypothesis, we measured serum matrix metalloproteinase (MMP) activities using gelatin gels for analyzing the MMP-2 and MMP-9. Further, employing casein gels, we also studied MMP-13 that is known to be influenced by the growth arrest and DNA damage-45 (GADD45) protein during sleep and wake cycle. The wild type and cystathionine β synthase-deficient (CBS-/+) mice strains were treated with H2S and subjected to measurement of trans-sulfuration factors from skeletal muscle tissues. The results suggested highly robust activation of MMPs in the wake mice versus sleep mice, which appears somewhat akin to the "1-carbon metabolic dysregulation", which takes place during remodeling of extracellular matrix during muscular dystrophy. Interestingly, the levels of trans-sulfuration factors such as CBS, cystathionine γ lyase (CSE), methyl tetrahydrofolate reductase (MTHFR), phosphatidylethanolamine N-methyltransferase (PEMT), and Hcy-protein bound paraoxonase 1 (PON1) were attenuated in CBS-/+ mice. However, treatment with H2S mitigated the attenuation of the trans-sulfuration pathway. In addition, levels of mitochondrial peroxisome proliferator-activated receptor-gamma coactivator 1-α (PGC 1-α) and mitofusin-2 (MFN-2) were significantly improved by H2S intervention. Our findings suggest participation of the circadian clock in trans-sulfuration pathway that affects skeletal muscle remodeling and mitochondrial regeneration.
Collapse
Affiliation(s)
- Suresh C Tyagi
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Sathnur Pushpakumar
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Utpal Sen
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Oluwaseun E Akinterinwa
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Yuting Zheng
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Sri Prakash L Mokshagundam
- Division of Endocrinology, Metabolism and Diabetes and Robley Rex VA Medical Center, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Dinesh K Kalra
- Division of Cardiovascular Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Mahavir Singh
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
5
|
Khan S, Siddique R, Liu Y, Yong VW, Xue M. Towards improving the prognosis of stroke through targeting the circadian clock system. Int J Biol Sci 2024; 20:403-413. [PMID: 38169640 PMCID: PMC10758097 DOI: 10.7150/ijbs.88370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 10/17/2023] [Indexed: 01/05/2024] Open
Abstract
Rhythmicity of the circadian system is a 24-hour period, driven by transcription-translation feedback loops of circadian clock genes. The central circadian pacemaker in mammals is located in the hypothalamic suprachiasmatic nucleus (SCN), which controls peripheral circadian clocks. In general, most physiological processes are regulated by the circadian system, which is modulated by environmental cues such as exposure to light and/or dark, temperature, and the timing of sleep/wake and food intake. The chronic circadian disruption caused by shift work, jetlag, and/or irregular sleep-wake cycles has long-term health consequences. Its dysregulation contributes to the risk of psychiatric disorders, sleep abnormalities, hypothyroidism and hyperthyroidism, cancer, and obesity. A number of neurological conditions may be worsened by changes in the circadian clock via the SCN pacemaker. For stroke, different physiological activities such as sleep/wake cycles are disrupted due to alterations in circadian rhythms. Moreover, the immunological processes that affect the evolution and recovery processes of stroke are regulated by the circadian clock or core-clock genes. Thus, disrupted circadian rhythms may increase the severity and consequences of stroke, while readjustment of circadian clock machinery may accelerate recovery from stroke. In this manuscript, we discuss the relationship between stroke and circadian rhythms, particularly on stroke development and its recovery process. We focus on immunological and/or molecular processes linking stroke and the circadian system and suggest the circadian rhythm as a target for designing effective therapeutic strategies in stroke.
Collapse
Affiliation(s)
- Suliman Khan
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rabeea Siddique
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Liu
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - V. Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
6
|
Jamal NE, Brooks TG, Cohen J, Townsend RR, de Sosa GR, Shah V, Nelson RG, Drawz PE, Rao P, Bhat Z, Chang A, Yang W, FitzGerald GA, Skarke C. Prognostic utility of rhythmic components in 24-hour ambulatory blood pressure monitoring for the risk stratification of chronic kidney disease patients with cardiovascular co-morbidity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.02.23289413. [PMID: 37205602 PMCID: PMC10187452 DOI: 10.1101/2023.05.02.23289413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Background Chronic kidney disease (CKD) represents a significant global burden. Hypertension is a modifiable risk factor for rapid progression of CKD. Methods We extend the risk stratification by introducing the non-parametric determination of rhythmic components in 24-hour profiles of ambulatory blood pressure monitoring (ABPM) in the African American Study for Kidney Disease and Hypertension (AASK) cohort and the Chronic Renal Insufficiency Cohort (CRIC) using Cox proportional hazards models. Results We find that rhythmic profiling of BP through JTK_Cycle analysis identifies subgroups of CRIC participants at advanced risk of cardiovascular death. CRIC participants with a history of cardiovascular disease (CVD) and absent cyclic components in their BP profile had at any time a 3.4-times higher risk of cardiovascular death than CVD patients with cyclic components present in their BP profile (HR: 3.38, 95% CI: 1.45-7.88, p=0.005). This substantially increased risk was independent of whether ABPM followed a dipping or non-dipping pattern whereby non-dipping or reverse dipping were not significantly associated with cardiovascular death in patients with prior CVD (p>0.1). In the AASK cohort, unadjusted models demonstrate a higher risk in reaching end stage renal disease among participants without rhythmic ABPM components (HR:1.80, 95% CI: 1.10-2.96); however, full adjustment abolished this association. Conclusions This study proposes rhythmic blood pressure components as a novel biomarker to unmask excess risk among CKD patients with prior cardiovascular disease.
Collapse
Affiliation(s)
- Nadim E1 Jamal
- Institute for Translational Medicine and Therapeutics (ITMAT), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Thomas G. Brooks
- Institute for Translational Medicine and Therapeutics (ITMAT), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jordana Cohen
- Renal-Electrolyte and Hypertension Division, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Raymond R. Townsend
- Renal-Electrolyte and Hypertension Division, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Vallabh Shah
- Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM, USA
- Department of Biochemistry, School of Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Robert G. Nelson
- The Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| | - Paul E. Drawz
- Division of Nephrology and Hypertension, University of Minnesota, Minneapolis, MN, USA
| | - Panduranga Rao
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Zeenat Bhat
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Alexander Chang
- Kidney Health Research Institute, Department of Population Health Sciences, Geisinger, Danville, PA, USA
| | - Wei Yang
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Garret A. FitzGerald
- Institute for Translational Medicine and Therapeutics (ITMAT), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Carsten Skarke
- Institute for Translational Medicine and Therapeutics (ITMAT), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | |
Collapse
|
7
|
Gumz ML, Shimbo D, Abdalla M, Balijepalli RC, Benedict C, Chen Y, Earnest DJ, Gamble KL, Garrison SR, Gong MC, Hogenesch JB, Hong Y, Ivy JR, Joe B, Laposky AD, Liang M, MacLaughlin EJ, Martino TA, Pollock DM, Redline S, Rogers A, Dan Rudic R, Schernhammer ES, Stergiou GS, St-Onge MP, Wang X, Wright J, Oh YS. Toward Precision Medicine: Circadian Rhythm of Blood Pressure and Chronotherapy for Hypertension - 2021 NHLBI Workshop Report. Hypertension 2023; 80:503-522. [PMID: 36448463 PMCID: PMC9931676 DOI: 10.1161/hypertensionaha.122.19372] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Healthy individuals exhibit blood pressure variation over a 24-hour period with higher blood pressure during wakefulness and lower blood pressure during sleep. Loss or disruption of the blood pressure circadian rhythm has been linked to adverse health outcomes, for example, cardiovascular disease, dementia, and chronic kidney disease. However, the current diagnostic and therapeutic approaches lack sufficient attention to the circadian rhythmicity of blood pressure. Sleep patterns, hormone release, eating habits, digestion, body temperature, renal and cardiovascular function, and other important host functions as well as gut microbiota exhibit circadian rhythms, and influence circadian rhythms of blood pressure. Potential benefits of nonpharmacologic interventions such as meal timing, and pharmacologic chronotherapeutic interventions, such as the bedtime administration of antihypertensive medications, have recently been suggested in some studies. However, the mechanisms underlying circadian rhythm-mediated blood pressure regulation and the efficacy of chronotherapy in hypertension remain unclear. This review summarizes the results of the National Heart, Lung, and Blood Institute workshop convened on October 27 to 29, 2021 to assess knowledge gaps and research opportunities in the study of circadian rhythm of blood pressure and chronotherapy for hypertension.
Collapse
Affiliation(s)
- Michelle L Gumz
- Department of Physiology and Aging; Center for Integrative Cardiovascular and Metabolic Disease, Department of Medicine, Division of Nephrology, Hypertension and Renal Transplantation, University of Florida, Gainesville, FL (M.L.G.)
| | - Daichi Shimbo
- Department of Medicine, The Columbia Hypertension Center, Columbia University Irving Medical Center, New York, NY (D.S.)
| | - Marwah Abdalla
- Department of Medicine, Center for Behavioral Cardiovascular Health, Columbia University Irving Medical Center, New York, NY (M.A.)
| | - Ravi C Balijepalli
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD (R.C.B., Y.H., J.W., Y.S.O.)
| | - Christian Benedict
- Department of Pharmaceutical Biosciences, Molecular Neuropharmacology, Uppsala University, Sweden (C.B.)
| | - Yabing Chen
- Department of Pathology, University of Alabama at Birmingham, and Research Department, Birmingham VA Medical Center, AL (Y.C.)
| | - David J Earnest
- Department of Neuroscience & Experimental Therapeutics, Texas A&M University, Bryan, TX (D.J.E.)
| | - Karen L Gamble
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, AL (K.L.G.)
| | - Scott R Garrison
- Department of Family Medicine, University of Alberta, Canada (S.R.G.)
| | - Ming C Gong
- Department of Physiology, University of Kentucky, Lexington, KY (M.C.G.)
| | | | - Yuling Hong
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD (R.C.B., Y.H., J.W., Y.S.O.)
| | - Jessica R Ivy
- University/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, United Kingdom (J.R.I.)
| | - Bina Joe
- Department of Physiology and Pharmacology and Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, OH (B.J.)
| | - Aaron D Laposky
- National Center on Sleep Disorders Research, Division of Lung Diseases, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD (A.D.L.)
| | - Mingyu Liang
- Center of Systems Molecular Medicine, Department of Physiology, Medical College of Wisconsin, Milwaukee, WI (M.L.)
| | - Eric J MacLaughlin
- Department of Pharmacy Practice, Texas Tech University Health Sciences Center, Amarillo, TX (E.J.M.)
| | - Tami A Martino
- Center for Cardiovascular Investigations, Department of Biomedical Sciences, University of Guelph, Ontario, Canada (T.A.M.)
| | - David M Pollock
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, AL (D.M.P.)
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (S.R.)
| | - Amy Rogers
- Division of Molecular and Clinical Medicine, University of Dundee, United Kingdom (A.R.)
| | - R Dan Rudic
- Department of Pharmacology and Toxicology, Augusta University, GA (R.D.R.)
| | - Eva S Schernhammer
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (E.S.S.)
| | - George S Stergiou
- Hypertension Center, STRIDE-7, National and Kapodistrian University of Athens, School of Medicine, Third Department of Medicine, Sotiria Hospital, Athens, Greece (G.S.S.)
| | - Marie-Pierre St-Onge
- Division of General Medicine, Department of Medicine, Columbia University Irving Medical Center' New York, NY (M.-P.S.-O.)
| | - Xiaoling Wang
- Georgia Prevention Institute, Department of Medicine, Augusta University, GA (X.W.)
| | - Jacqueline Wright
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD (R.C.B., Y.H., J.W., Y.S.O.)
| | - Young S Oh
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD (R.C.B., Y.H., J.W., Y.S.O.)
| |
Collapse
|
8
|
Huo M, Cao X, Zhang H, Lau CW, Hong H, Chen FM, Huang Y, Chawla A, Tian XY. Loss of myeloid Bmal1 exacerbates hypertensive vascular remodelling through interaction with STAT6 in mice. Cardiovasc Res 2022; 118:2859-2874. [PMID: 34726702 DOI: 10.1093/cvr/cvab336] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 09/07/2021] [Indexed: 01/11/2023] Open
Abstract
AIMS In addition to its involvement of inflammatory responses, limited information is available on the phenotype and behaviour of vascular macrophages during hypertensive vascular remodelling. Here, we aim at studying the contribution of BMAL1 to the pro-fibrotic macrophage phenotype in the vasculature during hypertension, which leads to enhanced vascular remodelling and promoted blood pressure increase. METHODS AND RESULTS Wild type Bmal1f/f and myeloid cell selective Bmal1 knockout Bmal1f/f; LysMCre/+ mice were infused with AngII for 4 weeks to induce hypertension. AngII-induced blood pressure increase, vascular media thickness and vascular dysfunction were enhanced in Bmal1f/f; LysMCre/+ mice, accompanied with a pro-fibrotic M2 phenotype of the vascular macrophages. Bmal1f/f; LysMCre/+ mice also have more up-regulations of MMP9 and MMP13 expression in the vascular wall, accompanied by enhanced collagen deposition after AngII infusion. Loss of Bmal1 in bone marrow-derived macrophages enhanced STAT6 activation induced by IL4, and the subsequent MMP13 up-regulation and activity. In macrophages, loss of Bmal1 enhanced the phosphorylation and nuclear translocation of STAT6 triggered by IL4, through possibly a direct interaction between BMAL1 and STAT6. To further determine whether IL4-induced signalling in macrophage contributes to enhanced vascular remodelling in hypertensive mice, we showed that deletion of myeloid IL4Rα in Il4raf/f; LysMCre/+ mice attenuated blood pressure increase and hypertensive vascular remodelling after AngII infusion. CONCLUSIONS Our results suggested a tonic effect of BMAL1 deletion on hypertensive vascular remodelling. BMAL1 might inhibit IL4-STAT6 signalling in macrophages through the interaction with STAT6 to reduce STAT6 activation and target gene transcription, especially MMP9 and MMP13, contributing to vascular remodelling.
Collapse
Affiliation(s)
- Mingyu Huo
- School of Biomedical Sciences, Faculty of Medicine, CUHK Shenzhen Research Institute, Heart and Vascular Institute, Rm208, LIBSB, Chinese University of Hong Kong, Shatin, NT. Hong Kong SAR, China
- Digestive Medicine Centre, The Seventh Affiliated Hospital of Sun Yat-sen University, Zhenyuan Rd, Guangming (New) Dist., Shenzhen, China, 518107
| | - Xiaoyun Cao
- School of Biomedical Sciences, Faculty of Medicine, CUHK Shenzhen Research Institute, Heart and Vascular Institute, Rm208, LIBSB, Chinese University of Hong Kong, Shatin, NT. Hong Kong SAR, China
| | - Hongsong Zhang
- School of Biomedical Sciences, Faculty of Medicine, CUHK Shenzhen Research Institute, Heart and Vascular Institute, Rm208, LIBSB, Chinese University of Hong Kong, Shatin, NT. Hong Kong SAR, China
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Changle Rd, Qinhuai District, Nanjing, Jiangsu, China, 210029
| | - Chi Wai Lau
- School of Biomedical Sciences, Faculty of Medicine, CUHK Shenzhen Research Institute, Heart and Vascular Institute, Rm208, LIBSB, Chinese University of Hong Kong, Shatin, NT. Hong Kong SAR, China
| | - Huiling Hong
- School of Biomedical Sciences, Faculty of Medicine, CUHK Shenzhen Research Institute, Heart and Vascular Institute, Rm208, LIBSB, Chinese University of Hong Kong, Shatin, NT. Hong Kong SAR, China
| | - Francis M Chen
- School of Life Sciences, MMW 505, Chinese University of Hong Kong, Shatin, NT. Hong Kong SAR, China
| | - Yu Huang
- School of Biomedical Sciences, Faculty of Medicine, CUHK Shenzhen Research Institute, Heart and Vascular Institute, Rm208, LIBSB, Chinese University of Hong Kong, Shatin, NT. Hong Kong SAR, China
| | - Ajay Chawla
- Department of Physiology, Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA 94143-0795
| | - Xiao Yu Tian
- School of Biomedical Sciences, Faculty of Medicine, CUHK Shenzhen Research Institute, Heart and Vascular Institute, Rm208, LIBSB, Chinese University of Hong Kong, Shatin, NT. Hong Kong SAR, China
| |
Collapse
|
9
|
Yu Z, Liu Z, Jiao L, Zhang S, Nie L, Wang Y, Zhou L, Wang Y, Liu Z, Liu Z, Xu X, Li Z, Zhou Y, Zhou H, Li R, Peng C, Yu L, Jiang H. Bmal1 knockdown in the left stellate ganglion inhibits neural activity and prevents ventricular arrhythmias after myocardial ischemia. Front Cardiovasc Med 2022; 9:937608. [PMID: 36247430 PMCID: PMC9556266 DOI: 10.3389/fcvm.2022.937608] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 08/25/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives The neural activity of the left stellate ganglion (LSG) is closely related to the occurrence of ventricular arrhythmias (VAs). Bmal1 modulates genes associated with neural activity in the central nervous system. However, few studies indicated the role of Bmal1 in the LSG and the subsequent effect on the heart. Therefore, we aimed to investigate the influence of Bmal1 knockdown in the LSG on its neural activity and cardiac electrophysiology and to explore the mechanisms. Materials and methods We used adeno-associated virus (AAV) to knock down Bmal1 in the LSG. Male beagles were randomized into the Bmal1 knockdown group and the control group. After 4 weeks of injection, the LSG function, neural activity, left ventricular effective refractory period (ERP), and action potential duration (APD) were measured. Electrocardiography for 1 h was recorded for VAs analysis after myocardial ischemia. Nerve growth factor (NGF) and c-fos in the LSG were quantified by immunofluorescence. Transcriptomic analysis was performed to assess the gene expression in the LSG. Results Bmal1 was sufficiently knocked down by AAV. Compared with the control group, heart rate variability (HRV) in the knockdown group was altered. Bmal1 knockdown inhibited neural activity and function of LSG. It also prolonged ERP as well as APD90. Ischemia-induced VAs were significantly reduced. Nerve growth factor (NGF) and c-fos in the LSG were reduced. Bmal1 knockdown led to the expression changes of genes associated with neural activity in the LSG. Conclusion Bmal1 knockdown in the LSG suppresses neural activity and prevents ventricular arrhythmias after myocardial ischemia.
Collapse
Affiliation(s)
- Zhongyang Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zhihao Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Liying Jiao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Song Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Liqing Nie
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yueyi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Liping Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yuhong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zhihao Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zihan Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Xiao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zeyan Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yuyang Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Huixin Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Rui Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Chen Peng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Lilei Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
- Lilei Yu,
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
- *Correspondence: Hong Jiang,
| |
Collapse
|
10
|
The role of temporal changes of pro-inflammatory cytokines in the development of adverse cardiac remodeling after ST-elevation myocardial infarction. Adv Cardiol 2022; 18:217-227. [PMID: 36751290 PMCID: PMC9885240 DOI: 10.5114/aic.2022.120938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/10/2022] [Indexed: 11/18/2022]
Abstract
Introduction Increasing evidence supports the view that pro-inflammatory cytokines play a role in fibrosis after myocardial infarction (MI). It has been suggested that interleukin (IL)-12p40, a pro-inflammatory cytokine, can induce interferon γ (IFN-γ) and matrix metalloproteinase (MMP). However, the role of IL-12p40 in adverse cardiac remodeling (AR) after ST-elevation MI (STEMI) is unclear. Aim To examine the role of temporal changes of pro-inflammatory cytokines in the development of post-STEMI AR. Material and methods A total of 43 patients with STEMI for the first time ever were prospectively analyzed. In cardiac magnetic resonance imaging at 6 months after STEMI, a decrease of left ventricular end-diastolic volume by ≥ 12% was defined as reverse cardiac remodeling (RR), and a 12% increase was defined as AR. Cytokine concentrations were measured on the first day (baseline) and 2 weeks after STEMI. Results Mean IL-12p40 (59.1 ±14.5 vs. 46.7 ±9.1 pq/ml, p = 0.001), median IFN-γ (20.4 vs. 16.2 pq/ml, p = 0.048) and median MMP-2 (33866 vs. 20691 pq/ml, p = 0.011) baseline concentrations were higher in AR than RR. In patients with AR, IL-12p40 level was lower at 2 weeks than baseline (p < 0.001). There was a positive correlation between the baseline concentrations of IL-12p40, IFN-γ, MMP-2, C-reactive protein and infarct size (p < 0.05). Increased IL-12p40 and MMP-2 baseline levels were independently associated with AR (OR = 1.14, p = 0.010; OR = 1.08, p = 0.035). Conclusions In the initial phase of MI, greater release of pro-inflammatory cytokines was associated with increased MMP-2 levels. Elevated expression of IL-12 and MMP-2 had an independent association with AR. This may be related to the excessive inflammatory response in the initial phase of MI.
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
|
Lin C, Xu L, Tang X, Li X, Lu C, Cheng Q, Jiang J, Shen Y, Yan D, Qian R, Fu W, Guo D. Clock Gene Bmal1 Disruption in Vascular Smooth Muscle Cells Worsens Carotid Atherosclerotic Lesions. Arterioscler Thromb Vasc Biol 2022; 42:565-579. [PMID: 35236106 DOI: 10.1161/atvbaha.121.316480] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Clock system disruptions are associated with cardiovascular diseases. We previously demonstrated Bmal1 (brain muscle aryl nuclear translocase like-1) expression is significantly attenuated in plaque-derived vascular smooth muscle cells (VSMCs). However, the influence of Bmal1 disruption in VSMCs and its molecular targets are still unclear. Here, we aim to define how Bmal1 disruption in VSMCs influences the atherosclerosis lesions. METHODS The relationship among Bmal1, neurological symptoms, and plaque stability was investigated. VSMC Bmal1-/- and VSMC Bmal1+/+mice were generated and injected with adeno associated virus encoding mutant proprotein convertase subtilisin/kexin type 9 to induce atherosclerosis. Carotid artery ligation and cuff placement were performed in these mice to confirm the role of Bmal1 in atherosclerosis progression. The relevant molecular mechanisms were then explored. RESULTS Bmal1 expression in the carotid plague was significantly lower in symptomatic patients as well as in unstable plaques. Moreover, Bmal1 reduction is an independent risk factor for neurological symptoms and plaque instability. Besides, VSMC Bmal1-/- mice exhibit aggravated atherosclerotic lesions. Further study demonstrated that Bmal1 downregulation in VSMCs increased VSMC migration, monocyte transmigration, reactive oxygen species levels, and VSMCs apoptosis. As for the mechanism, we revealed that Bmal1 suppresses VSMCs migration by inhibiting RAC1 activity in 2 ways: by activating the transcription of RhoGDIα and by interacting with RAC1. Besides, Bmal1 was shown to preserve antioxidant function in VSMCs by activating Nrf2 (nuclear factor erythroid 2-related factor 2) and Bcl-2 transcription. CONCLUSIONS Bmal1 disruption in VSMCs worsens atherosclerosis by promoting VSMC migration and monocyte transmigration and impairing antioxidant function. Therefore, Bmal1 may be a potential therapeutic target and biomarker of atherosclerosis in the future.
Collapse
Affiliation(s)
- Changpo Lin
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China (C.L., X.T., J.J., Y.S., D.Y., W.F., D.G.).,National Clinical Research Center for Interventional Medicine, Shanghai, China (C.L., X.T., J.J., Y.S., D.Y., W.F., D.G.).,Shanghai Clinical Research Center for Interventional Medicine, China (C.L., X.T., J.J., Y.S., D.Y., W.F., D.G.)
| | - Lirong Xu
- National Clinical Research Center for Interventional Medicine, Shanghai, China (C.L., X.T., J.J., Y.S., D.Y., W.F., D.G.).,Shanghai Clinical Research Center for Interventional Medicine, China (C.L., X.T., J.J., Y.S., D.Y., W.F., D.G.)
| | - Xiao Tang
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China (C.L., X.T., J.J., Y.S., D.Y., W.F., D.G.)
| | - Xiaobo Li
- Department of Pathology, School of Basic Medical Science, Shanghai University of Traditional Chinese Medicine, China (L.X.).,Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, China (X.L., C.L., Q.C., R.Q.)
| | - Chao Lu
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, China (X.L., C.L., Q.C., R.Q.)
| | - Qianyun Cheng
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, China (X.L., C.L., Q.C., R.Q.)
| | - Junhao Jiang
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China (C.L., X.T., J.J., Y.S., D.Y., W.F., D.G.).,National Clinical Research Center for Interventional Medicine, Shanghai, China (C.L., X.T., J.J., Y.S., D.Y., W.F., D.G.).,Shanghai Clinical Research Center for Interventional Medicine, China (C.L., X.T., J.J., Y.S., D.Y., W.F., D.G.)
| | - Yang Shen
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China (C.L., X.T., J.J., Y.S., D.Y., W.F., D.G.).,National Clinical Research Center for Interventional Medicine, Shanghai, China (C.L., X.T., J.J., Y.S., D.Y., W.F., D.G.).,Shanghai Clinical Research Center for Interventional Medicine, China (C.L., X.T., J.J., Y.S., D.Y., W.F., D.G.)
| | - Dong Yan
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China (C.L., X.T., J.J., Y.S., D.Y., W.F., D.G.).,National Clinical Research Center for Interventional Medicine, Shanghai, China (C.L., X.T., J.J., Y.S., D.Y., W.F., D.G.).,Shanghai Clinical Research Center for Interventional Medicine, China (C.L., X.T., J.J., Y.S., D.Y., W.F., D.G.)
| | - Ruizhe Qian
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, China (X.L., C.L., Q.C., R.Q.)
| | - Weiguo Fu
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China (C.L., X.T., J.J., Y.S., D.Y., W.F., D.G.).,National Clinical Research Center for Interventional Medicine, Shanghai, China (C.L., X.T., J.J., Y.S., D.Y., W.F., D.G.).,Shanghai Clinical Research Center for Interventional Medicine, China (C.L., X.T., J.J., Y.S., D.Y., W.F., D.G.)
| | - Daqiao Guo
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China (C.L., X.T., J.J., Y.S., D.Y., W.F., D.G.).,National Clinical Research Center for Interventional Medicine, Shanghai, China (C.L., X.T., J.J., Y.S., D.Y., W.F., D.G.).,Shanghai Clinical Research Center for Interventional Medicine, China (C.L., X.T., J.J., Y.S., D.Y., W.F., D.G.)
| |
Collapse
|
13
|
Bryant AJ, Ebrahimi E, Nguyen A, Wolff CA, Gumz ML, Liu AC, Esser KA. A wrinkle in time: circadian biology in pulmonary vascular health and disease. Am J Physiol Lung Cell Mol Physiol 2022; 322:L84-L101. [PMID: 34850650 PMCID: PMC8759967 DOI: 10.1152/ajplung.00037.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
An often overlooked element of pulmonary vascular disease is time. Cellular responses to time, which are regulated directly by the core circadian clock, have only recently been elucidated. Despite an extensive collection of data regarding the role of rhythmic contribution to disease pathogenesis (such as systemic hypertension, coronary artery, and renal disease), the roles of key circadian transcription factors in pulmonary hypertension remain understudied. This is despite a large degree of overlap in the pulmonary hypertension and circadian rhythm fields, not only including shared signaling pathways, but also cell-specific effects of the core clock that are known to result in both protective and adverse lung vessel changes. Therefore, the goal of this review is to summarize the current dialogue regarding common pathways in circadian biology, with a specific emphasis on its implications in the progression of pulmonary hypertension. In this work, we emphasize specific proteins involved in the regulation of the core molecular clock while noting the circadian cell-specific changes relevant to vascular remodeling. Finally, we apply this knowledge to the optimization of medical therapy, with a focus on sleep hygiene and the role of chronopharmacology in patients with this disease. In dissecting the unique relationship between time and cellular biology, we aim to provide valuable insight into the practical implications of considering time as a therapeutic variable. Armed with this information, physicians will be positioned to more efficiently use the full four dimensions of patient care, resulting in improved morbidity and mortality of pulmonary hypertension patients.
Collapse
Affiliation(s)
- Andrew J. Bryant
- 1Department of Medicine, University of Florida College of Medicine, Gainesville, Florida
| | - Elnaz Ebrahimi
- 1Department of Medicine, University of Florida College of Medicine, Gainesville, Florida
| | - Amy Nguyen
- 1Department of Medicine, University of Florida College of Medicine, Gainesville, Florida
| | - Christopher A. Wolff
- 2Department of Physiology, University of Florida College of Medicine, Gainesville, Florida
| | - Michelle L. Gumz
- 2Department of Physiology, University of Florida College of Medicine, Gainesville, Florida
| | - Andrew C. Liu
- 2Department of Physiology, University of Florida College of Medicine, Gainesville, Florida
| | - Karyn A. Esser
- 2Department of Physiology, University of Florida College of Medicine, Gainesville, Florida
| |
Collapse
|
14
|
The Vascular Circadian Clock in Chronic Kidney Disease. Cells 2021; 10:cells10071769. [PMID: 34359937 PMCID: PMC8306728 DOI: 10.3390/cells10071769] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/29/2021] [Accepted: 07/09/2021] [Indexed: 12/11/2022] Open
Abstract
Chronic kidney disease is associated with extremely high cardiovascular mortality. The circadian rhythms (CR) have an impact on vascular function. The disruption of CR causes serious health problems and contributes to the development of cardiovascular diseases. Uremia may affect the master pacemaker of CR in the hypothalamus. A molecular circadian clock is also expressed in peripheral tissues, including the vasculature, where it regulates the different aspects of both vascular physiology and pathophysiology. Here, we address the impact of CKD on the intrinsic circadian clock in the vasculature. The expression of the core circadian clock genes in the aorta is disrupted in CKD. We propose a novel concept of the disruption of the circadian clock system in the vasculature of importance for the pathology of the uremic vasculopathy.
Collapse
|
15
|
Han Q, Bagi Z, Rudic RD. Review: Circadian clocks and rhythms in the vascular tree. Curr Opin Pharmacol 2021; 59:52-60. [PMID: 34111736 DOI: 10.1016/j.coph.2021.04.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 04/23/2021] [Accepted: 04/28/2021] [Indexed: 12/17/2022]
Abstract
The progression of vascular disease is influenced by many factors including aging, gender, diet, hypertension, and poor sleep. The intrinsic vascular circadian clock and the timing it imparts on the vasculature both conditions and is conditioned by all these variables. Circadian rhythms and their molecular components are rhythmically cycling in each endothelial cell, smooth muscle cell, in each artery, arteriole, vein, venule, and capillary. New research continues to tackle how circadian clocks act in the vasculature, describing influences in experimental and human disease, identifying potential target genes, compensatory molecules, that ultimately reveal a complexity that is vascular-bed-specific, cell-type-specific, and even single-cell-specific. Though we are yet to achieve a complete understanding, here we survey recent observations that are shedding more light on the nature of the interaction between circadian rhythms and the vascular system with implications for blood vessel disease.
Collapse
Affiliation(s)
- Qimei Han
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Zsolt Bagi
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Raducu Daniel Rudic
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
16
|
Chen Z, Xiong ZF, Liu X. Research progress on the interaction between circadian clock and early vascular aging. Exp Gerontol 2021; 146:111241. [PMID: 33453324 DOI: 10.1016/j.exger.2021.111241] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/15/2022]
Abstract
Considerable researches implicate that the circadian clock regulates the responsive rhythms of organs and sets the orderly aging process of cells indirectly. It influences an array of diverse biological process including intestinal flora, peripheral inflammatory responses, and redox homeostasis. People with sleep disoders and other kinds of circadian disruptions are prone to have vascular aging earlier. Meanwhile, those people are always faced with chronic vascular inflammation. It has not been elucidated that the specific mechanism of the interaction between the circadian system and early vascular aging. To explore the biphasic relationship between vascular aging and the circadian system, we summarize what is linking circadian clock with early vascular aging through four major prospect: inflammatory process, oxidative stress response, intestinal flora, and cellular senescence. Meanwhile, we discuss the hypothesis that the deterioration of circadian rhythms may exacerbate the process of early vascular aging, leading to the cardiovascular diseases. It will help us to provide new ideas for understanding the process of vascular aging and exploring the possible ways to design personalized chronotherapies.
Collapse
Affiliation(s)
- Zhuoying Chen
- Department of Geriatrics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China
| | - Zhi-Fan Xiong
- Department of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China
| | - Xiangjie Liu
- Department of Geriatrics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China.
| |
Collapse
|
17
|
Huang H, Li Z, Ruan Y, Feng W, Chen J, Li X, Ouyang L, Huang H. Circadian rhythm disorder: a potential inducer of vascular calcification? J Physiol Biochem 2020; 76:513-524. [PMID: 32945991 DOI: 10.1007/s13105-020-00767-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 09/14/2020] [Indexed: 12/24/2022]
Abstract
Over the past decades, circadian rhythm has drawn a great attention in cardiovascular diseases. The expressions of rhythm genes fluctuate in accordance with the diurnal changes of vascular physiology, which highlights the pivotal effect of vascular clock. Recent researches show that the circadian clock can directly regulate the synthetic and secretory function of endothelial cells and phenotypic switch of vascular smooth muscle cells to adjust vascular relaxation and contraction. Importantly, dysfunction of vascular cells is involved in vascular calcification. Secretion of osteogenic cytokines and calcified vesicles in the vessel, osteogenic phenotype switch of vascular smooth muscle cells are all implicated in the calcification process. Moreover, circadian rhythm disorder can lead to abnormal hormone secretion, oxidative stress, inflammatory reaction, and autophagy, all of which should not be ignored in vascular calcification. Vascular senescence is another pathogenetic mechanism in vascular calcification. Accelerated vascular senescence may act as an important intermediate factor to promote vascular calcification in circadian rhythm disorders. In this review, we elaborate the potential effect of circadian rhythm disorder in vascular calcification and try to provide a new direction in the prevention of vascular calcification.
Collapse
Affiliation(s)
- Haoran Huang
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, No. 3025, Shennan Middle Road, Futian District, Shenzhen, 518000, China
- Department of Pediatric Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhaohuai Li
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, No. 3025, Shennan Middle Road, Futian District, Shenzhen, 518000, China
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yuyi Ruan
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, No. 3025, Shennan Middle Road, Futian District, Shenzhen, 518000, China
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Weijing Feng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jie Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoxue Li
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, No. 3025, Shennan Middle Road, Futian District, Shenzhen, 518000, China
| | - Liu Ouyang
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, No. 3025, Shennan Middle Road, Futian District, Shenzhen, 518000, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hui Huang
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, No. 3025, Shennan Middle Road, Futian District, Shenzhen, 518000, China.
| |
Collapse
|
18
|
Xie M, Tang Q, Nie J, Zhang C, Zhou X, Yu S, Sun J, Cheng X, Dong N, Hu Y, Chen L. BMAL1-Downregulation Aggravates Porphyromonas Gingivalis-Induced Atherosclerosis by Encouraging Oxidative Stress. Circ Res 2020; 126:e15-e29. [PMID: 32078488 DOI: 10.1161/circresaha.119.315502] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
RATIONALE Atherosclerotic cardiovascular diseases are the leading cause of mortality worldwide. Atherosclerotic cardiovascular diseases are considered as chronic inflammation processes. In addition to risk factors associated with the cardiovascular system itself, pathogenic bacteria such as the periodontitis-associated Porphyromonas gingivalis (P gingivalis) are also closely correlated with the development of atherosclerosis, but the underlying mechanisms are still elusive. OBJECTIVE To elucidate the mechanisms of P gingivalis-accelerated atherosclerosis and explore novel therapeutic strategies of atherosclerotic cardiovascular diseases. METHODS AND RESULTS Bmal1-/- (brain and muscle Arnt-like protein 1) mice, ApoE-/- mice, Bmal1-/-ApoE-/- mice, conditional endothelial cell Bmal1 knockout mice (Bmal1fl/fl; Tek-Cre mice), and the corresponding jet-legged mouse model were used. Pgingivalis accelerates atherosclerosis progression by triggering arterial oxidative stress and inflammatory responses in ApoE-/- mice, accompanied by the perturbed circadian clock. Circadian clock disruption boosts P gingivalis-induced atherosclerosis progression. The mechanistic dissection shows that P gingivalis infection activates the TLRs-NF-κB signaling axis, which subsequently recruits DNMT-1 to methylate the BMAL1 promoter and thus suppresses BMAL1 transcription. The downregulation of BMAL1 releases CLOCK, which phosphorylates p65 and further enhances NF-κB signaling, elevating oxidative stress and inflammatory response in human aortic endothelial cells. Besides, the mouse model exhibits that joint administration of metronidazole and melatonin serves as an effective strategy for treating atherosclerotic cardiovascular diseases. CONCLUSIONS P gingivalis accelerates atherosclerosis via the NF-κB-BMAL1-NF-κB signaling loop. Melatonin and metronidazole are promising auxiliary medications toward atherosclerotic cardiovascular diseases.
Collapse
Affiliation(s)
- Mengru Xie
- From the Department of Stomatology (M.X., Q.T., J.N., X.Z., S.Y., J.S., L.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China (M.X., Q.T., J.N., X.Z., S.Y., J.S., L.C.)
| | - Qingming Tang
- From the Department of Stomatology (M.X., Q.T., J.N., X.Z., S.Y., J.S., L.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China (M.X., Q.T., J.N., X.Z., S.Y., J.S., L.C.)
| | - Jiaming Nie
- From the Department of Stomatology (M.X., Q.T., J.N., X.Z., S.Y., J.S., L.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China (M.X., Q.T., J.N., X.Z., S.Y., J.S., L.C.)
| | - Chao Zhang
- Department of Cardiovascular Surgery (C.Z., N.D.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Zhou
- From the Department of Stomatology (M.X., Q.T., J.N., X.Z., S.Y., J.S., L.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China (M.X., Q.T., J.N., X.Z., S.Y., J.S., L.C.)
| | - Shaoling Yu
- From the Department of Stomatology (M.X., Q.T., J.N., X.Z., S.Y., J.S., L.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China (M.X., Q.T., J.N., X.Z., S.Y., J.S., L.C.)
| | - Jiwei Sun
- From the Department of Stomatology (M.X., Q.T., J.N., X.Z., S.Y., J.S., L.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China (M.X., Q.T., J.N., X.Z., S.Y., J.S., L.C.)
| | - Xiang Cheng
- Department of Cardiology (X.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nianguo Dong
- Department of Cardiovascular Surgery (C.Z., N.D.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Hu
- Institute of Hematology (Y.H.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lili Chen
- From the Department of Stomatology (M.X., Q.T., J.N., X.Z., S.Y., J.S., L.C.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China (M.X., Q.T., J.N., X.Z., S.Y., J.S., L.C.)
| |
Collapse
|
19
|
Motohashi H, Tahara Y, Whittaker DS, Wang HB, Yamaji T, Wakui H, Haraguchi A, Yamazaki M, Miyakawa H, Hama K, Sasaki H, Sakai T, Hirooka R, Takahashi K, Takizawa M, Makino S, Aoyama S, Colwell CS, Shibata S. The circadian clock is disrupted in mice with adenine-induced tubulointerstitial nephropathy. Kidney Int 2020; 97:728-740. [PMID: 31948598 DOI: 10.1016/j.kint.2019.09.032] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 09/21/2019] [Accepted: 09/26/2019] [Indexed: 12/13/2022]
Abstract
Chronic Kidney Disease (CKD) is increasing in incidence and has become a worldwide health problem. Sleep disorders are prevalent in patients with CKD raising the possibility that these patients have a disorganized circadian timing system. Here, we examined the effect of adenine-induced tubulointerstitial nephropathy on the circadian system in mice. Compared to controls, adenine-treated mice showed serum biochemistry evidence of CKD as well as increased kidney expression of inflammation and fibrosis markers. Mice with CKD exhibited fragmented sleep behavior and locomotor activity, with lower degrees of cage activity compared to mice without CKD. On a molecular level, mice with CKD exhibited low amplitude rhythms in their central circadian clock as measured by bioluminescence in slices of the suprachiasmatic nucleus of PERIOD 2::LUCIFERASE mice. Whole animal imaging indicated that adenine treated mice also exhibited dampened oscillations in intact kidney, liver, and submandibular gland. Consistently, dampened circadian oscillations were observed in several circadian clock genes and clock-controlled genes in the kidney of the mice with CKD. Finally, mice with a genetically disrupted circadian clock (Clock mutants) were treated with adenine and compared to wild type control mice. The treatment evoked worse kidney damage as indicated by higher deposition of gelatinases (matrix metalloproteinase-2 and 9) and adenine metabolites in the kidney. Adenine also caused non-dipping hypertension and lower heart rate. Thus, our data indicate that central and peripheral circadian clocks are disrupted in the adenine-treated mice, and suggest that the disruption of the circadian clock accelerates CKD progression.
Collapse
Affiliation(s)
- Hiroaki Motohashi
- Laboratory of Physiology and Pharmacology, School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, Japan
| | - Yu Tahara
- Laboratory of Physiology and Pharmacology, School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, Japan; Department of Psychiatry and Biobehavioral Sciences, Semel Institute, University of California, Los Angeles, Los Angeles, California, USA
| | - Daniel S Whittaker
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, University of California, Los Angeles, Los Angeles, California, USA
| | - Huei-Bin Wang
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, University of California, Los Angeles, Los Angeles, California, USA
| | - Takahiro Yamaji
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hiromichi Wakui
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Atsushi Haraguchi
- Laboratory of Physiology and Pharmacology, School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, Japan
| | - Mayu Yamazaki
- Laboratory of Physiology and Pharmacology, School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, Japan
| | - Hiroki Miyakawa
- Laboratory of Physiology and Pharmacology, School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, Japan
| | - Koki Hama
- Laboratory of Physiology and Pharmacology, School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, Japan
| | - Hiroyuki Sasaki
- Laboratory of Physiology and Pharmacology, School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, Japan
| | - Tomoko Sakai
- Laboratory of Physiology and Pharmacology, School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, Japan
| | - Rina Hirooka
- Laboratory of Physiology and Pharmacology, School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, Japan
| | - Kengo Takahashi
- Laboratory of Physiology and Pharmacology, School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, Japan
| | - Miku Takizawa
- Laboratory of Physiology and Pharmacology, School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, Japan
| | - Saneyuki Makino
- Laboratory of Physiology and Pharmacology, School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, Japan
| | - Shinya Aoyama
- Laboratory of Physiology and Pharmacology, School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, Japan
| | - Christopher S Colwell
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, University of California, Los Angeles, Los Angeles, California, USA
| | - Shigenobu Shibata
- Laboratory of Physiology and Pharmacology, School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, Japan.
| |
Collapse
|
20
|
Wu X, Chen L, Zeb F, Li C, Jiang P, Chen A, Xu C, Haq IU, Feng Q. Clock-Bmal1 mediates MMP9 induction in acrolein-promoted atherosclerosis associated with gut microbiota regulation. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 252:1455-1463. [PMID: 31265956 DOI: 10.1016/j.envpol.2019.06.042] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/28/2019] [Accepted: 06/11/2019] [Indexed: 06/09/2023]
Abstract
Circadian rhythm is believed to play important roles in atherosclerosis. The gut microbiota is found to be closely related to atherogenesis, and shows compositional and functional circadian oscillation. However, it's still unclarified whether circadian clock and intestinal microbiota are involved in the progression of atherosclerosis induced by environmental pollutant acrolein. Herein, patients with atherosclerosis showed higher MMP9, a promising biomarker for atherosclerosis, and lower Bmal1 and Clock expression in the plasma. Interestingly, acrolein exposure contributed to the increased MMP9, decreased Clock and Bmal1, and activated MAPK pathways in human umbilical vein endothelial cells (HUVECs). We found that knockdown of Clock or Bmal1 lead to upregulation of MMP9 in HUVECs, and that Clock and Bmal1 expression was elevated while MAPK pathways were blocked. Atherosclerotic apolipoproteinE-deficient mice consumed a high-fat diet were used and treated with acrolein (3 mg/kg/day) in the drinking water for 12 weeks. Upregulation of MMP9, and downregulation of Clock and Bmal1 were also observed in plasma of the mice. Besides, acrolein feeding altered gut microbiota composition at a phylum level especially for an increased Firmicutes and a decreased Bacteroidetes. Additionally, gut microbiota showed correlation with atherosclerotic plaque, MMP9 and Bmal1 levels. Therefore, our findings indicated that acrolein increased the expression of MMP9 through MAPK regulating circadian clock, which was associated with gut microbiota regulation in atherosclerosis. Circadian rhythms and gut microbiota might be promising targets in the prevention of cardiovascular disease caused by environmental pollutants.
Collapse
Affiliation(s)
- Xiaoyue Wu
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Lijun Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Falak Zeb
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Chaofeng Li
- Department of Cardiology, The Second Affiliated Hospital of Southeast University, Nanjing, 210000, China
| | - Pan Jiang
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Aochang Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Chuyue Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Ijaz Ul Haq
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Qing Feng
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
21
|
Abstract
Matrix metalloproteinases (MMPs) and their endogenous inhibitors have been studied in the myocardium for the past 2 decades. An incomplete knowledge base and experimental design issues with inhibitors have hampered attempts at translation, but clinical interest remains high because of strong associations between MMPs and outcomes after myocardial infarction (MI) as well as mechanistic studies showing MMP involvement at multiple stages of the MI wound-healing process. This Review focuses on how our understanding of MMPs has evolved from a one-dimensional early focus on measuring MMP activity, monitoring MMP:inhibitor ratios, and evaluating one MMP-substrate pair to the current use of systems biology approaches to integrate the whole MMP repertoire of roles in the left ventricular response to MI. MMP9 is used as an example MMP to explain these concepts and to provide a template for examining MMPs as mechanistic mediators of cardiac remodelling.
Collapse
Affiliation(s)
- Merry L Lindsey
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA. .,Research Service,, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS, USA.
| |
Collapse
|
22
|
Winter C, Soehnlein O, Maegdefessel L. TIMPing the Aorta: Smooth Muscle Cell-Specific Deletion of BMAL1 Limits Murine Abdominal Aortic Aneurysm Development. Arterioscler Thromb Vasc Biol 2019; 38:982-983. [PMID: 29695531 DOI: 10.1161/atvbaha.118.310857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Carla Winter
- From the Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University Munich, Germany (C.W., O.S.)
| | - Oliver Soehnlein
- From the Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University Munich, Germany (C.W., O.S.).,Department of Physiology and Pharmacology (O.S.).,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Germany (O.S., L.M.)
| | - Lars Maegdefessel
- Department of Medicine Solna, Center for Molecular Medicine (L.M.), Karolinska Institute, Stockholm, Sweden .,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Germany (O.S., L.M.).,Department of Vascular and Endovascular Surgery, Technical University Munich, Germany (L.M.)
| |
Collapse
|
23
|
Xu Y, Pi W, Rudic RD. Old and New Roles and Evolving Complexities of Cardiovascular Clocks. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2019; 92:283-290. [PMID: 31249489 PMCID: PMC6585526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The cardiovascular (CV) system has been established to be significantly influenced by the molecular components of circadian rhythm. Oscillations of circadian rhythm occur within the circulation to affect thrombosis and blood pressure and within CV tissues including arteries, heart, and kidney to control function. Physiologic and molecular oscillations of circadian rhythm have been well connected via global, tissue-specific, and transgenic reporter mouse models of key core clock signals such as Bmal1, Period, and Clock, which can produce both pathology and protection with their mutation. With different nuances of CV clock action continuing to emerge in studies of the cardiovascular system, new questions are raised in both new and old mouse model system observations that underscore the importance, complexity, and continued study of the circadian clock mechanism in cardiovascular disease.
Collapse
Affiliation(s)
| | | | - R. D. Rudic
- To whom all correspondence should be addressed: Dan Rudic, Augusta University, 1120 15th Street, Augusta, GA, 30912, CB3620; Tel:706 721-7649, Fax 706 721-2347, E-mail:
| |
Collapse
|
24
|
Li HX. The role of circadian clock genes in tumors. Onco Targets Ther 2019; 12:3645-3660. [PMID: 31190867 PMCID: PMC6526167 DOI: 10.2147/ott.s203144] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 04/10/2019] [Indexed: 12/12/2022] Open
Abstract
Circadian rhythms are generated via variations in the expression of clock genes that are organized into a complex transcriptional–translational autoregulatory network and regulate the diverse physiological and behavioral activities that are required to adapt to periodic environmental changes. Aberrant clock gene expression is associated with a heightened risk of diseases that affect all aspects of human health, including cancers. Within the past several years, a number of studies have indicated that clock genes contribute to carcinogenesis by altering the expression of clock-controlled and tumor-related genes downstream of many cellular pathways. This review comprehensively summarizes how clock genes affect the development of tumors and their prognosis. In addition, the review provides a full description of the current state of oral cancer research that aims to optimize cancer diagnosis and treatment modalities.
Collapse
Affiliation(s)
- Han-Xue Li
- Department of Preventive Dentistry, Stomatological Hospital of Chongqing Medical University, Chongqing 400015, People's Republic of China
| |
Collapse
|
25
|
Rabinovich-Nikitin I, Lieberman B, Martino TA, Kirshenbaum LA. Circadian-Regulated Cell Death in Cardiovascular Diseases. Circulation 2019; 139:965-980. [DOI: 10.1161/circulationaha.118.036550] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Inna Rabinovich-Nikitin
- The Institute of Cardiovascular Sciences, St Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Canada (I.R.-N., B.L., L.A.K.)
| | - Brooke Lieberman
- The Institute of Cardiovascular Sciences, St Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Canada (I.R.-N., B.L., L.A.K.)
| | - Tami A. Martino
- Centre for Cardiovascular Investigations, Biomedical Sciences/Ontario Veterinary College, University of Guelph, Canada (T.A.M.)
| | - Lorrie A. Kirshenbaum
- The Institute of Cardiovascular Sciences, St Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Canada (I.R.-N., B.L., L.A.K.)
| |
Collapse
|
26
|
Liu SL, Bajpai A, Hawthorne EA, Bae Y, Castagnino P, Monslow J, Puré E, Spiller KL, Assoian RK. Cardiovascular protection in females linked to estrogen-dependent inhibition of arterial stiffening and macrophage MMP12. JCI Insight 2019; 4:e122742. [PMID: 30626744 PMCID: PMC6485356 DOI: 10.1172/jci.insight.122742] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 11/20/2018] [Indexed: 12/21/2022] Open
Abstract
Arterial stiffening is a consequence of aging and a cholesterol-independent risk factor for cardiovascular disease (CVD). Arterial stiffening and CVD show a sex bias, with men more susceptible than premenopausal women. How arterial stiffness and sex interact at a molecular level to confer risk of CVD is not well understood. Here, we used the sexual dimorphism in LDLR-null mice to show that the protective effect of female sex on atherosclerosis is linked to reduced aortic stiffness and reduced expression of matrix metalloproteinase-12 (MMP12) by lesional macrophages. Deletion of MMP12 in LDLR-null mice attenuated the male sex bias for both arterial stiffness and atherosclerosis, and these effects occurred despite high serum cholesterol. Mechanistically, we found that oxidized LDL stimulates secretion of MMP12 in human as well as mouse macrophages. Estrogen antagonizes this effect by downregulating MMP12 expression. Our data support cholesterol-independent causal relationships between estrogen, oxidized LDL-induced secretion of macrophage MMP12, and arterial stiffness that protect against atherosclerosis in females and emphasize that reduced MMP12 functionality can confer atheroprotection to males.
Collapse
Affiliation(s)
- Shu-lin Liu
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Anamika Bajpai
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania, USA
| | - Elizabeth A. Hawthorne
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Engineering MechanoBiology and
| | - Yongho Bae
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Paola Castagnino
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Engineering MechanoBiology and
| | - James Monslow
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ellen Puré
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kara L. Spiller
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania, USA
| | - Richard K. Assoian
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Engineering MechanoBiology and
| |
Collapse
|
27
|
Crnko S, Cour M, Van Laake LW, Lecour S. Vasculature on the clock: Circadian rhythm and vascular dysfunction. Vascul Pharmacol 2018; 108:1-7. [PMID: 29778521 DOI: 10.1016/j.vph.2018.05.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 04/23/2018] [Accepted: 05/10/2018] [Indexed: 01/08/2023]
Abstract
The master mammalian circadian clock (i.e. central clock), located in the suprachiasmatic nucleus of the hypothalamus, orchestrates the synchronization of the daily behavioural and physiological rhythms to better adapt the organism to the external environment in an anticipatory manner. This central clock is entrained by a variety of signals, the best established being light and food. However, circadian cycles are not simply the consequences of these two cues but are generated by endogenous circadian clocks. Indeed, clock machinery is found in mainly all tissues and cell types, including cells of the vascular system such as endothelial cells, fibroblasts, smooth muscle cells and stem cells. This machinery physiologically contributes to modulate the daily vascular function, and its disturbance therefore plays a major role in the pathophysiology of vascular dysfunction. Therapies targeting the circadian rhythm may therefore be of benefit against vascular disease.
Collapse
Affiliation(s)
- Sandra Crnko
- Division Heart and Lungs and Regenerative Medicine Center, University Medical Center Utrecht, The Netherlands
| | - Martin Cour
- Hatter Institute for Cardiovascular research in Africa and Lionel Opie Preclinical Imaging Core Facility, Faculty of Health Sciences, University of Cape Town, South Africa
| | - Linda W Van Laake
- Division Heart and Lungs and Regenerative Medicine Center, University Medical Center Utrecht, The Netherlands
| | - Sandrine Lecour
- Hatter Institute for Cardiovascular research in Africa and Lionel Opie Preclinical Imaging Core Facility, Faculty of Health Sciences, University of Cape Town, South Africa.
| |
Collapse
|
28
|
Lutshumba J, Liu S, Zhong Y, Hou T, Daugherty A, Lu H, Guo Z, Gong MC. Deletion of BMAL1 in Smooth Muscle Cells Protects Mice From Abdominal Aortic Aneurysms. Arterioscler Thromb Vasc Biol 2018; 38:1063-1075. [PMID: 29437576 PMCID: PMC5920729 DOI: 10.1161/atvbaha.117.310153] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 01/25/2018] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Abdominal aortic aneurysm (AAA) has high mortality rate when ruptured, but currently, there is no proven pharmacological therapy for AAA because of our poor understanding of its pathogenesis. The current study explored a novel role of smooth muscle cell (SMC) BMAL1 (brain and muscle Arnt-like protein-1)-a transcription factor known to regulate circadian rhythm-in AAA development. APPROACH AND RESULTS SMC-selective deletion of BMAL1 potently protected mice from AAA induced by (1) MR (mineralocorticoid receptor) agonist deoxycorticosterone acetate or aldosterone plus high salt intake and (2) angiotensin II infusion in hypercholesterolemia mice. Aortic BMAL1 was upregulated by deoxycorticosterone acetate-salt, and deletion of BMAL1 in SMCs selectively upregulated TIMP4 (tissue inhibitor of metalloproteinase 4) and suppressed deoxycorticosterone acetate-salt-induced MMP (matrix metalloproteinase) activation and elastin breakages. Moreover, BMAL1 bound to the Timp4 promoter and suppressed Timp4 transcription. CONCLUSIONS These results reveal an important, but previously unexplored, role of SMC BMAL1 in AAA. Moreover, these results identify TIMP4 as a novel target of BMAL1, which may mediate the AAA protective effect of SMC BMAL1 deletion.
Collapse
MESH Headings
- ARNTL Transcription Factors/deficiency
- ARNTL Transcription Factors/genetics
- Aldosterone
- Angiotensin II
- Animals
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/prevention & control
- Binding Sites
- Desoxycorticosterone Acetate
- Dilatation, Pathologic
- Disease Models, Animal
- Elastin/metabolism
- Male
- Matrix Metalloproteinase 2/metabolism
- Matrix Metalloproteinase 9/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Promoter Regions, Genetic
- Sodium Chloride, Dietary
- Tissue Inhibitor of Metalloproteinases/genetics
- Tissue Inhibitor of Metalloproteinases/metabolism
- Transcription, Genetic
- Tissue Inhibitor of Metalloproteinase-4
Collapse
Affiliation(s)
- Jenny Lutshumba
- From the Department of Physiology (J.L., Y.Z., A.D., H.L., M.C.G.)
| | - Shu Liu
- Department of Pharmacology and Nutritional Sciences (S.L., T.H., Z.G.), University of Kentucky, Lexington
| | - Yu Zhong
- From the Department of Physiology (J.L., Y.Z., A.D., H.L., M.C.G.)
| | | | - Alan Daugherty
- From the Department of Physiology (J.L., Y.Z., A.D., H.L., M.C.G.)
| | - Hong Lu
- From the Department of Physiology (J.L., Y.Z., A.D., H.L., M.C.G.)
- Department of Pharmacology and Nutritional Sciences (S.L., T.H., Z.G.), University of Kentucky, Lexington
| | - Zhenheng Guo
- Department of Pharmacology and Nutritional Sciences (S.L., T.H., Z.G.), University of Kentucky, Lexington
- Department of Research and Development, Lexington VA Medical Center, KY (Z.G.)
| | - Ming C Gong
- From the Department of Physiology (J.L., Y.Z., A.D., H.L., M.C.G.)
| |
Collapse
|
29
|
Soler A, Hunter I, Joseph G, Hutcheson R, Hutcheson B, Yang J, Zhang FF, Joshi SR, Bradford C, Gotlinger KH, Maniyar R, Falck JR, Proctor S, Schwartzman ML, Gupte SA, Rocic P. Elevated 20-HETE in metabolic syndrome regulates arterial stiffness and systolic hypertension via MMP12 activation. J Mol Cell Cardiol 2018; 117:88-99. [PMID: 29428638 PMCID: PMC5877315 DOI: 10.1016/j.yjmcc.2018.02.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/08/2018] [Accepted: 02/07/2018] [Indexed: 11/24/2022]
Abstract
Arterial stiffness plays a causal role in development of systolic hypertension. 20-hydroxyeicosatetraeonic acid (20-HETE), a cytochrome P450 (CYP450)-derived arachidonic acid metabolite, is known to be elevated in resistance arteries in hypertensive animal models and loosely associated with obesity in humans. However, the role of 20-HETE in the regulation of large artery remodeling in metabolic syndrome has not been investigated. We hypothesized that elevated 20-HETE in metabolic syndrome increases matrix metalloproteinase 12 (MMP12) activation leading to increased degradation of elastin, increased large artery stiffness and increased systolic blood pressure. 20-HETE production was increased ~7 fold in large, conduit arteries of metabolic syndrome (JCR:LA-cp, JCR) vs. normal Sprague-Dawley (SD) rats. This correlated with increased elastin degradation (~7 fold) and decreased arterial compliance (~75% JCR vs. SD). 20-HETE antagonists blocked elastin degradation in JCR rats concomitant with blocking MMP12 activation. 20-HETE antagonists normalized, and MMP12 inhibition (pharmacological and MMP12-shRNA-Lnv) significantly improved (~50% vs. untreated JCR) large artery compliance in JCR rats. 20-HETE antagonists also decreased systolic (182 ± 3 mmHg JCR, 145 ± 3 mmHg JCR + 20-HETE antagonists) but not diastolic blood pressure in JCR rats. Whereas diastolic pressure was fully angiotensin II (Ang II)-dependent, systolic pressure was only partially Ang II-dependent, and large artery stiffness was Ang II-independent. Thus, 20-HETE-dependent regulation of systolic blood pressure may be a unique feature of metabolic syndrome related to high 20-HETE production in large, conduit arteries, which results in increased large artery stiffness and systolic blood pressure. These findings may have implications for management of systolic hypertension in patients with metabolic syndrome.
Collapse
Affiliation(s)
- Amanda Soler
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Ian Hunter
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Gregory Joseph
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Rebecca Hutcheson
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Brenda Hutcheson
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Jenny Yang
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Frank Fan Zhang
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Sachindra Raj Joshi
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Chastity Bradford
- Department of Biology, Tuskegee University, Tuskegee, AL 36088, United States
| | - Katherine H Gotlinger
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Rachana Maniyar
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - John R Falck
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Spencer Proctor
- Metabolic and Cardiovascular Diseases Laboratory, Alberta Institute for Human Nutrition, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | | | - Sachin A Gupte
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Petra Rocic
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States.
| |
Collapse
|
30
|
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
|
31
|
Nanoudis S, Pikilidou M, Yavropoulou M, Zebekakis P. The Role of MicroRNAs in Arterial Stiffness and Arterial Calcification. An Update and Review of the Literature. Front Genet 2017; 8:209. [PMID: 29312437 PMCID: PMC5733083 DOI: 10.3389/fgene.2017.00209] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Accepted: 11/28/2017] [Indexed: 12/20/2022] Open
Abstract
Arterial stiffness is an independent risk factor for fatal and non-fatal cardiovascular events, such as systolic hypertension, coronary artery disease, stroke, and heart failure. Moreover it reflects arterial aging which in many cases does not coincide with chronological aging, a fact that is in large attributed to genetic factors. In addition to genetic factors, microRNAs (miRNAs) seem to largely affect arterial aging either by advancing or by regressing arterial stiffness. MiRNAs are small RNA molecules, ~22 nucleotides long that can negatively control their target gene expression posttranscriptionally. Pathways that affect main components of stiffness such as fibrosis and calcification seem to be influenced by up or downregulation of specific miRNAs. Identification of this aberrant production of miRNAs can help identify epigenetic changes that can be therapeutic targets for prevention and treatment of vascular diseases. The present review summarizes the specific role of the so far discovered miRNAs that are involved in pathways of arterial stiffness.
Collapse
Affiliation(s)
- Sideris Nanoudis
- Hypertension Excellence Center, 1st Department of Internal Medicine, AHEPA University Hospital, Thessaloniki, Greece
| | - Maria Pikilidou
- Hypertension Excellence Center, 1st Department of Internal Medicine, AHEPA University Hospital, Thessaloniki, Greece
| | - Maria Yavropoulou
- Division of Endocrinology and Metabolism, AHEPA University Hospital, Thessaloniki, Greece
| | - Pantelis Zebekakis
- Hypertension Excellence Center, 1st Department of Internal Medicine, AHEPA University Hospital, Thessaloniki, Greece
| |
Collapse
|
32
|
Lou J, Wang Y, Zhang Z, Qiu W. Activation of MMPs in Macrophages by Mycobacterium tuberculosis via the miR-223-BMAL1 Signaling Pathway. J Cell Biochem 2017; 118:4804-4812. [PMID: 28543681 DOI: 10.1002/jcb.26150] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/18/2017] [Indexed: 12/12/2022]
Abstract
An interaction between Mycobacterium tuberculosis and macrophages constitutes an essential step in tuberculosis development, as macrophages exert both positive and negative effects on M. tuberculosis-triggered organ lesions. In this study, we focused on the regulation of the expression of matrix metalloproteinases (MMPs), which is responsible for lung matrix degradation and bacteria dissection, in macrophages following M. tuberculosis infection. Female BALB/c mice were intravenously injected with the M. tuberculosis strain H37Rv at 0 h zeitgeber time (ZT0) or 12 h zeitgeber time (ZT12). The expression and activity of MMP-1, -2, -3, and -9 in lungs and spleens were then evaluated. In vitro, peritoneal macrophages were harvested at ZT0 or at ZT12 and infected with 10 MOI M. tuberculosis. The expression of MMPs, microRNA-223 and BMAL1 was analyzed by qRT-PCR and/or Western blot. The binding of BMAL1 3'-UTR by miR-223 was confirmed by luciferase activity assay. Additionally, wild-type BMAL1 or NLSmut BMAL1 plasmids were transfected to evaluate the effect of BMAL1 on MMPs. The results showed a differential expression of MMPs in mice tissues infected at different times. M. tuberculosis infection caused enhanced MMP-1, -9, and miR-223 expression, with inhibited BMAL1 expression. MiR-223 modulated BMAL1 expression via the direct binding of BMAL1 3'-UTR. Furthermore, wild-type BMAL1 other than NLSmut BMAL1 attenuated MMPs expression in M. tuberculosis-infected macrophages. Overall, this study demonstrated a potential involvement of circadian rhythm in MMP activation by M. tuberculosis in macrophages. J. Cell. Biochem. 118: 4804-4812, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jun Lou
- Department of Clinical Laboratory, Zhumadian Central Hospital, Zhumadian, 463000, P.R. China
| | - Yongli Wang
- Neonatal Intensive Care Unit, Zhumadian Central Hospital, Zhumadian, 463000, P.R. China
| | - Zhimin Zhang
- Department of Clinical Laboratory, Zhumadian Central Hospital, Zhumadian, 463000, P.R. China
| | - Weiqiang Qiu
- Department of Clinical Laboratory, Zhumadian Central Hospital, Zhumadian, 463000, P.R. China
| |
Collapse
|
33
|
Leloup A, De Moudt S, Van Hove C, Fransen P. Cyclic Stretch Alters Vascular Reactivity of Mouse Aortic Segments. Front Physiol 2017; 8:858. [PMID: 29163203 PMCID: PMC5674939 DOI: 10.3389/fphys.2017.00858] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/13/2017] [Indexed: 11/13/2022] Open
Abstract
Large, elastic arteries buffer the pressure wave originating in the left ventricle and are constantly exposed to higher amplitudes of cyclic stretch (10%) than muscular arteries (2%). As a crucial factor for endothelial and smooth muscle cell function, cyclic stretch has, however, never been studied in ex vivo aortic segments of mice. To investigate the effects of cyclic stretch on vaso-reactivity of mouse aortic segments, we used the Rodent Oscillatory Tension Set-up to study Arterial Compliance (ROTSAC). The aortic segments were clamped at frequencies of 6–600 bpm between two variable preloads, thereby mimicking dilation as upon left ventricular systole and recoiling as during diastole. The preloads corresponding to different transmural pressures were chosen to correspond to a low, normal or high amplitude of cyclic stretch. At different time intervals, cyclic stretch was interrupted, the segments were afterloaded and isometric contractions by α1-adrenergic stimulation with 2 μM phenylephrine in the absence and presence of 300 μM L-NAME (eNOS inhibitor) and/or 35 μM diltiazem (blocker of voltage-gated Ca2+ channels) were measured. As compared with static or cyclic stretch at low amplitude (<10 mN) or low frequency (0.1 Hz), cyclic stretch at physiological amplitude (>10 mN) and frequency (1–10 Hz) caused better ex vivo conservation of basal NO release with time after mounting. The relaxation of PE-precontracted segments by addition of ACh to stimulate NO release was unaffected by cyclic stretch. In the absence of basal NO release (hence, presence of L-NAME), physiological in comparison with aberrant cyclic stretch decreased the baseline tension, attenuated the phasic contraction by phenylephrine in the absence of extracellular Ca2+ and shifted the smaller tonic contraction more from a voltage-gated Ca2+ channel-mediated to a non-selective cation channel-mediated. Data highlight the need of sufficient mechanical activation of endothelial and vascular smooth muscle cells to maintain basal NO release and low intracellular Ca2+ in the smooth muscle cells in large arteries. Both phenomena may play a vital role in maintaining the high compliance of large arteries.
Collapse
Affiliation(s)
- Arthur Leloup
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Sofie De Moudt
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Cor Van Hove
- Laboratory of Pharmacology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Paul Fransen
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
34
|
Fletcher EK, Morgan J, Kennaway DR, Bienvenu LA, Rickard AJ, Delbridge LMD, Fuller PJ, Clyne CD, Young MJ. Deoxycorticosterone/Salt-Mediated Cardiac Inflammation and Fibrosis Are Dependent on Functional CLOCK Signaling in Male Mice. Endocrinology 2017; 158:2906-2917. [PMID: 28911177 DOI: 10.1210/en.2016-1911] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 07/13/2017] [Indexed: 12/19/2022]
Abstract
Activation of the mineralocorticoid receptor (MR) promotes inflammation, fibrosis, and hypertension. Clinical and experimental studies show that MR antagonists have significant therapeutic benefit for all-cause heart failure; however, blockade of renal MRs limits their widespread use. Identification of key downstream signaling mechanisms for the MR in the cardiovascular system may enable development of targeted MR antagonists with selectivity for pathological MR signaling and lower impact on physiological renal electrolyte handling. One candidate pathway is the circadian clock, the dysregulation of which is associated with cardiovascular diseases. We have previously shown that the circadian gene Per2 is dysregulated in hearts with selective deletion of cardiomyocyte MR. We therefore investigated MR-mediated cardiac inflammation and fibrosis in mice that lack normal regulation and oscillation of the circadian clock in peripheral tissues, that is, CLOCKΔ19 mutant mice. The characteristic cardiac inflammatory/fibrotic response to a deoxycorticosterone (DOC)/salt for 8 weeks was significantly blunted in CLOCKΔ19 mice when compared with wild-type mice, despite a modest increase at "baseline" for fibrosis and macrophage number in CLOCKΔ19 mice. In contrast, cardiac hypertrophy in response to DOC/salt was significantly greater in CLOCKΔ19 vs wild-type mice. Markers for renal inflammation and fibrosis were similarly attenuated in the CLOCKΔ19 mice given DOC/salt. Moreover, increased CLOCK expression in H9c2 cardiac cells enhanced MR-mediated transactivation of Per1, suggesting cooperative signaling between these transcription factors. This study demonstrates that the full development of MR-mediated cardiac inflammation and fibrosis is dependent on intact signaling by the circadian protein CLOCK.
Collapse
Affiliation(s)
- Elizabeth K Fletcher
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
- Department of Physiology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - James Morgan
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - David R Kennaway
- School of Medicine, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Laura A Bienvenu
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
- Department of Physiology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Amanda J Rickard
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - Lea M D Delbridge
- Department of Physiology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Peter J Fuller
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
- Department of Medicine, Monash University, Clayton, Victoria 3168, Australia
| | - Colin D Clyne
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - Morag J Young
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
- Department of Medicine, Monash University, Clayton, Victoria 3168, Australia
- Department of Physiology, Monash University, Clayton, Victoria 3168, Australia
| |
Collapse
|
35
|
Abstract
The aorta is a blood vessel that provides a low-resistance path for blood flow directed from the heart to peripheral organs and tissues. However, the aorta has another central hemodynamic function, whereby the elastic nature of the aortic wall provides a significant biomechanical buffering capacity complementing the pulsatile cardiac blood flow, and this is often referred to as Windkessel function. Stiffening of the arterial wall leads to fundamental alterations in central hemodynamics, with widespread detrimental implications for organ function. In this Recent Highlights article, we describe recent contributions in ATVB that have highlighted the novel mechanisms and consequences of arterial stiffness and the clinical conditions in which arterial stiffness occurs, with a focus on advancements in the field.
Collapse
Affiliation(s)
- Alicia N. Lyle
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA, United States of America
| | - Uwe Raaz
- Molecular and Translational Vascular Medicine, Department of Cardiology and Pneumology, Heart Center at the University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
36
|
Li B, Jiao Y, Fu C, Xie B, Ma G, Teng G, Yao Y. Contralateral artery enlargement predicts carotid plaque progression based on machine learning algorithm models in apoE -/- mice. Biomed Eng Online 2016; 15:146. [PMID: 28155719 PMCID: PMC5259854 DOI: 10.1186/s12938-016-0265-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND This study specifically focused on anatomical MRI characterization of the low shear stress-induced atherosclerotic plaque in mice. We used machine learning algorithms to analyze multiple correlation factors of plaque to generate predictive models and to find the predictive factor for vulnerable plaque. METHODS Branches of the left carotid artery in apoE-/- and C57BL/6J mice were ligated to produce the partial left carotid artery model. Before surgery, and 7, 14, and 28 days after surgery, in vivo serial MRI measurements of carotid artery diameter were obtained. Meanwhile, proximal blood flow was evaluated. After image acquisition and animal sacrifice, carotid arteries were harvested for histological analysis. Support vector machine (SVM) and decision tree (DT) were used to select features and generate predictive models of vulnerable plaque progression. RESULT Seven days after surgery, neointima formation was visualized on micro-MRI in both apoE-/- and C57BL/6J mice. Ultrasonography showed that blood flow had significantly decreased compared to that in the contralateral artery. Partial ligation of the carotid artery for 4 weeks in apoE-/- mice induced vulnerable plaque; however, in C57BL/6J mice this same technique performed for 4 weeks induced arterial stenosis. Contralateral carotid artery diameter at 7 days after surgery was the most reliable predictive factor in plaque progression. We achieved over 87.5% accuracy, 80% sensitivity, and 95% specificity with SVM. The accuracy, sensitivity, and specificity for the DT classifier were 90, 90, and 90%, respectively. CONCLUSIONS This study is the first to demonstrate that SVM and DT methods could be suitable models for identifying vulnerable plaque progression in mice. And contralateral artery enlargement can predict the vulnerable plaque in carotid artery at the very early stage. It may be a valuable tool which helps to optimize the clinical work flow process by providing more decision in selecting patients for treatment.
Collapse
Affiliation(s)
- Bing Li
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao, Nanjing, 210009, Jiangsu, China
| | - Yun Jiao
- Jiangsu Key Lab of Molecular and Function Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Cong Fu
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao, Nanjing, 210009, Jiangsu, China
| | - Bo Xie
- Jiangsu Key Lab of Molecular and Function Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao, Nanjing, 210009, Jiangsu, China
| | - Gaojun Teng
- Jiangsu Key Lab of Molecular and Function Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Yuyu Yao
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao, Nanjing, 210009, Jiangsu, China.
| |
Collapse
|
37
|
Shang X, Pati P, Anea CB, Fulton DJ, Rudic RD. Differential Regulation of BMAL1, CLOCK, and Endothelial Signaling in the Aortic Arch and Ligated Common Carotid Artery. J Vasc Res 2016; 53:269-278. [PMID: 27923220 PMCID: PMC5765856 DOI: 10.1159/000452410] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 10/08/2016] [Indexed: 12/13/2022] Open
Abstract
The circadian clock is rhythmically expressed in blood vessels, but the interaction between the circadian clock and disturbed blood flow remains unclear. We examined the relationships between BMAL1 and CLOCK and 2 regulators of endothelial function, AKT1 and endothelial nitric oxide synthase (eNOS), in vascular regions of altered blood flow. We found that the aortic arch from WT mice exhibited reduced sensitivity to acetylcholine (Ach)-mediated relaxation relative to the thoracic aorta. In Clock-mutant (mut) mice the aorta exhibited a reduced sensitivity to Ach. In WT mice, the phosphorylated forms of eNOS and AKT were decreased in the aortic arch, while BMAL1 and CLOCK expression followed a similar pattern of reduction in the arch. In conditions of surgically induced flow reduction, phosphorylated-eNOS (serine 1177) increased, as did p-AKT in the ipsilateral left common carotid artery (LC) of WT mice. Similarly, BMAL1 and CLOCK exhibited increased expression after 5 days in the remodeled LC. eNOS expression was increased at 8 p.m. versus 8 a.m. in WT mice, and this pattern was abolished in mut and Bmal1-KO mice. These data suggest that the circadian clock may be a biomechanical and temporal sensor that acts to coordinate timing, flow dynamics, and endothelial function.
Collapse
MESH Headings
- ARNTL Transcription Factors/deficiency
- ARNTL Transcription Factors/genetics
- ARNTL Transcription Factors/metabolism
- Animals
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/metabolism
- CLOCK Proteins/genetics
- CLOCK Proteins/metabolism
- Carotid Artery Diseases/genetics
- Carotid Artery Diseases/metabolism
- Carotid Artery Diseases/physiopathology
- Carotid Artery, External/metabolism
- Carotid Artery, External/physiopathology
- Carotid Artery, External/surgery
- Circadian Rhythm
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Gene Expression Regulation
- Genotype
- Ligation
- Male
- Mechanotransduction, Cellular
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Mutant Strains
- Mutation
- Nitric Oxide Synthase Type III/metabolism
- Phenotype
- Phosphorylation
- Proto-Oncogene Proteins c-akt/metabolism
- Regional Blood Flow
- Stress, Mechanical
- Time Factors
- Vasodilation
- Vasodilator Agents/pharmacology
Collapse
Affiliation(s)
- Xia Shang
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, PR China
| | - Paramita Pati
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Ciprian B. Anea
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - David J.R. Fulton
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - R. Daniel Rudic
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| |
Collapse
|
38
|
Nernpermpisooth N, Qiu S, Mintz JD, Suvitayavat W, Thirawarapan S, Rudic DR, Fulton DJ, Stepp DW. Obesity alters the peripheral circadian clock in the aorta and microcirculation. Microcirculation 2016; 22:257-66. [PMID: 25660131 DOI: 10.1111/micc.12192] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 02/03/2015] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Perturbation of daily rhythm increases cardiovascular risk. The aim of this study was to determine whether obesity alters circadian gene expression and microvascular function in lean mice and obese (db/db) mice. METHODS Mice were subjected to normal LD or DD to alter circadian rhythm. Metabolic parameters and microvascular vasoreactivity were evaluated. Array studies were conducted in the am and pm cycles to assess the rhythmicity of the entire genomics. Rhythmic expression of specific clock genes (Bmal1, Clock, Npas2, Per1, Per2, and Cry1), clock output genes (dbp), and vascular relaxation-related genes (eNOS, GTPCH1) were assessed. RESULTS Obesity was associated with metabolic dysfunction and impaired endothelial dilation in the microvasculature. Circadian rhythm of gene expression was suppressed 80% in both macro- and microcirculations of obese mice. Circadian disruption with DD increased fasting serum glucose and HbA1c in obese but not lean mice. Endothelium-dependent dilation was attenuated in obese mice and in lean mice subjected to DD. Rhythmic expression of per1 and dbp was depressed in obesity. Expression of eNOS expression was suppressed and GTPCH1 lost rhythmic expression both in obesity and by constant darkness. CONCLUSION These results suggest that obesity reduces circadian gene expression in concert with impaired endothelial function. The causal relationship remains to be determined.
Collapse
|
39
|
Pati P, Fulton DJR, Bagi Z, Chen F, Wang Y, Kitchens J, Cassis LA, Stepp DW, Rudic RD. Low-Salt Diet and Circadian Dysfunction Synergize to Induce Angiotensin II-Dependent Hypertension in Mice. Hypertension 2016; 67:661-8. [PMID: 26781276 DOI: 10.1161/hypertensionaha.115.06194] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 12/16/2015] [Indexed: 01/03/2023]
Abstract
Blood pressure exhibits a robust circadian rhythm in health. In hypertension, sleep apnea, and even shift work, this balanced rhythm is perturbed via elevations in night-time blood pressure, inflicting silent damage to the vasculature and body organs. Herein, we examined the influence of circadian dysfunction during experimental hypertension in mice. Using radiotelemetry to measure ambulatory blood pressure and activity, the effects of angiotensin II administration were studied in wild-type (WT) and period isoform knockout (KO) mice (Per2-KO, Per2, 3-KO, and Per1, 2, 3-KO/Per triple KO [TKO] mice). On a normal diet, administration of angiotensin II caused nondipping blood pressure and exacerbated vascular hypertrophy in the Period isoform KO mice relative to WT mice. To study the endogenous effects of angiotensin II stimulation, we then administered a low-salt diet to the mice, which does stimulate endogenous angiotensin II in addition to lowering blood pressure. A low-salt diet decreased blood pressure in wild-type mice. In contrast, Period isoform KO mice lost their circadian rhythm in blood pressure on a low-salt diet, because of an increase in resting blood pressure, which was restorable to rhythmicity by the angiotensin receptor blocker losartan. Chronic administration of low salt caused vascular hypertrophy in Period isoform KO mice, which also exhibited increased renin levels and altered angiotensin 1 receptor expression. These data suggest that circadian clock genes may act to inhibit or control renin/angiotensin signaling. Moreover, circadian disorders such as sleep apnea and shift work may alter the homeostatic responses to sodium restriction to potentially influence nocturnal hypertension.
Collapse
Affiliation(s)
- Paramita Pati
- From the Departments of Pharmacology and Toxicology (P.P., D.J.R.F., J.K., R.D.R.), Medicine (Z.B.), and Physiology (D.W.S.), Vascular Biology Center (D.J.R.F., Z.B., F.C., Y.W., D.W.S.), Medical College of Georgia at Augusta University; and Department of Molecular and Biomedical Pharmacology, University of Kentucky, Lexington (L.A.C.)
| | - David J R Fulton
- From the Departments of Pharmacology and Toxicology (P.P., D.J.R.F., J.K., R.D.R.), Medicine (Z.B.), and Physiology (D.W.S.), Vascular Biology Center (D.J.R.F., Z.B., F.C., Y.W., D.W.S.), Medical College of Georgia at Augusta University; and Department of Molecular and Biomedical Pharmacology, University of Kentucky, Lexington (L.A.C.)
| | - Zsolt Bagi
- From the Departments of Pharmacology and Toxicology (P.P., D.J.R.F., J.K., R.D.R.), Medicine (Z.B.), and Physiology (D.W.S.), Vascular Biology Center (D.J.R.F., Z.B., F.C., Y.W., D.W.S.), Medical College of Georgia at Augusta University; and Department of Molecular and Biomedical Pharmacology, University of Kentucky, Lexington (L.A.C.)
| | - Feng Chen
- From the Departments of Pharmacology and Toxicology (P.P., D.J.R.F., J.K., R.D.R.), Medicine (Z.B.), and Physiology (D.W.S.), Vascular Biology Center (D.J.R.F., Z.B., F.C., Y.W., D.W.S.), Medical College of Georgia at Augusta University; and Department of Molecular and Biomedical Pharmacology, University of Kentucky, Lexington (L.A.C.)
| | - Yusi Wang
- From the Departments of Pharmacology and Toxicology (P.P., D.J.R.F., J.K., R.D.R.), Medicine (Z.B.), and Physiology (D.W.S.), Vascular Biology Center (D.J.R.F., Z.B., F.C., Y.W., D.W.S.), Medical College of Georgia at Augusta University; and Department of Molecular and Biomedical Pharmacology, University of Kentucky, Lexington (L.A.C.)
| | - Julia Kitchens
- From the Departments of Pharmacology and Toxicology (P.P., D.J.R.F., J.K., R.D.R.), Medicine (Z.B.), and Physiology (D.W.S.), Vascular Biology Center (D.J.R.F., Z.B., F.C., Y.W., D.W.S.), Medical College of Georgia at Augusta University; and Department of Molecular and Biomedical Pharmacology, University of Kentucky, Lexington (L.A.C.)
| | - Lisa A Cassis
- From the Departments of Pharmacology and Toxicology (P.P., D.J.R.F., J.K., R.D.R.), Medicine (Z.B.), and Physiology (D.W.S.), Vascular Biology Center (D.J.R.F., Z.B., F.C., Y.W., D.W.S.), Medical College of Georgia at Augusta University; and Department of Molecular and Biomedical Pharmacology, University of Kentucky, Lexington (L.A.C.)
| | - David W Stepp
- From the Departments of Pharmacology and Toxicology (P.P., D.J.R.F., J.K., R.D.R.), Medicine (Z.B.), and Physiology (D.W.S.), Vascular Biology Center (D.J.R.F., Z.B., F.C., Y.W., D.W.S.), Medical College of Georgia at Augusta University; and Department of Molecular and Biomedical Pharmacology, University of Kentucky, Lexington (L.A.C.)
| | - R Daniel Rudic
- From the Departments of Pharmacology and Toxicology (P.P., D.J.R.F., J.K., R.D.R.), Medicine (Z.B.), and Physiology (D.W.S.), Vascular Biology Center (D.J.R.F., Z.B., F.C., Y.W., D.W.S.), Medical College of Georgia at Augusta University; and Department of Molecular and Biomedical Pharmacology, University of Kentucky, Lexington (L.A.C.).
| |
Collapse
|
40
|
Takeda N, Maemura K. The role of clock genes and circadian rhythm in the development of cardiovascular diseases. Cell Mol Life Sci 2015; 72:3225-34. [PMID: 25972277 PMCID: PMC11113935 DOI: 10.1007/s00018-015-1923-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2015] [Revised: 05/04/2015] [Accepted: 05/04/2015] [Indexed: 10/23/2022]
Abstract
The time of onset of cardiovascular disorders such as myocardial infarctions or ventricular arrhythmias exhibits a circadian rhythm. Diurnal variations in autonomic nervous activity, plasma cortisol level or renin-angiotensin activity underlie the pathogenesis of cardiovascular diseases. Transcriptional-translational feedback loop of the clock genes constitute a molecular clock system. In addition to the central clock in the suprachiasmatic nucleus, clock genes are also expressed in a circadian fashion in each organ to make up the peripheral clock. The peripheral clock seems to be beneficial for anticipating external stimuli and thus contributes to the maintenance of organ homeostasis. Loss of synchronization between the central and peripheral clocks also augments disease progression. Moreover, accumulating evidence shows that clock genes affect inflammatory and intracellular metabolic signaling. Elucidating the roles of the molecular clock in cardiovascular pathology through the identification of clock controlled genes will help to establish a novel therapeutic approach for cardiovascular disorders.
Collapse
Affiliation(s)
- Norihiko Takeda
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655 Japan
| | - Koji Maemura
- Department of Cardiovascular Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| |
Collapse
|
41
|
Liao YC, Wang YS, Hsi E, Chang MH, You YZ, Juo SHH. MicroRNA-765 influences arterial stiffness through modulating apelin expression. Mol Cell Endocrinol 2015; 411:11-9. [PMID: 25882991 DOI: 10.1016/j.mce.2015.04.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 03/21/2015] [Accepted: 04/07/2015] [Indexed: 12/17/2022]
Abstract
The present study aims to discover single nucleotide polymorphisms (SNPs) at the apelin gene (APLN) in relation to arterial stiffness, and to explore its molecular mechanisms. A two-step genetic association study was conducted using 799 and 937 subjects in the screening and validation data, respectively. Four tagging SNPs of APLN were tested. SNP rs3115757 was significantly associated with stiffness parameters (β, Ep and PWV) in women, but not in men. The function of rs3115757 was tagged by rs3115758 which is located in miR-765 binding site in the 3' untranslated region of APLN. The reporter assay confirmed that different alleles of rs3115758 interfered with miR-765 binding and then modified APLN expression. Over-expression of miR-765 in endothelial cells decreased mRNA and protein levels of APLN, which further inhibited the phosphorylation of eNOS and ERK/Akt/AMPK signaling. Collectively, our data showed that rs3115758 accounts for the susceptibility of arterial stiffening through miR-765-induced APLN repression.
Collapse
Affiliation(s)
- Yi-Chu Liao
- Department of Neurology, Taipei Veterans General Hospital, No.201, Sec. 2, Shipai Rd., Taipei 112, Taiwan; Department of Neurology, National Yang-Ming University School of Medicine, No.155, Sec.2, Linong Street, Taipei 112, Taiwan
| | - Yung-Song Wang
- Institute of Fisheries Science, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan
| | - Edward Hsi
- Department of Genome Medicine, College of Medicine, Kaohsiung Medical University, 100, Shih-Chuan 1st Road, Kaohsiung 807, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, No.100, Tzyou 1st Road, Kaohsiung 807, Taiwan
| | - Ming-Hung Chang
- Division of Neuromuscular Disease, Neurological Institute, Taichung Veterans General Hospital, 1650 Taiwan Boulevard Sect. 4, Taichung 407, Taiwan
| | - Yun-Zhen You
- Division of Neuromuscular Disease, Neurological Institute, Taichung Veterans General Hospital, 1650 Taiwan Boulevard Sect. 4, Taichung 407, Taiwan
| | - Suh-Hang Hank Juo
- Department of Genome Medicine, College of Medicine, Kaohsiung Medical University, 100, Shih-Chuan 1st Road, Kaohsiung 807, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, No.100, Tzyou 1st Road, Kaohsiung 807, Taiwan.
| |
Collapse
|
42
|
Gao J, Yang C, Ding H, Zhang Y, Xue J, Cai Y. 2,2,2-Trichloroethanol lengthens the circadian period of Bmal1-driven circadian bioluminescence rhythms in U2OS cells. Biochem Biophys Res Commun 2015; 462:239-44. [PMID: 25956065 DOI: 10.1016/j.bbrc.2015.04.123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 04/26/2015] [Indexed: 12/27/2022]
Abstract
2,2,2-Trichloroethanol (TCOH) is responsible for the pharmacological actions of chloral hydrate (CH), and is a major metabolite of trichloroethylene. Human exposure to TCOH is known to be increasing. Recently, it was reported that TCOH causes a significant phase delay of Per2 expression in mouse liver when injected daily over the course of several days. However, it is not clear whether TCOH directly modulates the molecular clock. In the present study we used a cell-based assay system to test this possibility. We found that the daily oscillation period of Bmal1 was lengthened to 3 h following treatment with 1.5 mM TCOH, and increased to 5 h with 3 mM TCOH treatment. However, low concentrations of TCOH had no noticeable effects. The effect of TCOH on Per2 oscillation was marginal. Interestingly, serum from rats anesthetized with CH also modulated Bmal1 period, suggesting that exposure to anesthesia should be taken into consideration for circadian rhythm studies. In summary, our study reveals a direct regulation of TCOH on molecular clock.
Collapse
Affiliation(s)
- Jie Gao
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Parkinson's Disease Center of Beijing Institute for Brain Disorders, Beijing 100053, PR China
| | - Caixia Yang
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Parkinson's Disease Center of Beijing Institute for Brain Disorders, Beijing 100053, PR China
| | - Hui Ding
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Parkinson's Disease Center of Beijing Institute for Brain Disorders, Beijing 100053, PR China
| | - Yanli Zhang
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Parkinson's Disease Center of Beijing Institute for Brain Disorders, Beijing 100053, PR China
| | - Jinhua Xue
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Parkinson's Disease Center of Beijing Institute for Brain Disorders, Beijing 100053, PR China
| | - Yanning Cai
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Parkinson's Disease Center of Beijing Institute for Brain Disorders, Beijing 100053, PR China.
| |
Collapse
|
43
|
Bavachalcone Enhances RORα Expression, Controls Bmal1 Circadian Transcription, and Depresses Cellular Senescence in Human Endothelial Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015. [PMID: 26199639 PMCID: PMC4493309 DOI: 10.1155/2015/920431] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The circadian clock regulates many aspects of (patho)physiology in the central nervous system and in the peripheral tissues. RAR-related orphan receptor α (RORα), an orphan nuclear receptor, is involved in circadian rhythm regulation, including regulation of cardiovascular function. Bavachalcone, a prenylchalcone, is a major bioactive chalcone isolated from Psoralea corylifolia. This natural ingredient activated RORα1 luciferase reporter activity on drug screening. In addition, bavachalcone induced RORα1 expression in mRNA and protein levels in a dose-dependent manner and enhanced the circadian amplitude of Bmal1 mRNA expression after serum shock. Moreover, bavachalcone suppressed senescence in human endothelial cells and mRNA expression of p16(ink4a) (a marker of replicative senescence) and IL-1α (a proinflammatory cytokine of the senescence-associated secretory phenotype). These inhibitory effects were partially reversed by the RORα inhibitor VPR-66. Our results demonstrate that bavachalcone, as a natural medicine ingredient, has a pharmacological function in regulating RORα1.
Collapse
|
44
|
Chen L, Yang G. Recent advances in circadian rhythms in cardiovascular system. Front Pharmacol 2015; 6:71. [PMID: 25883568 PMCID: PMC4381645 DOI: 10.3389/fphar.2015.00071] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 03/16/2015] [Indexed: 12/20/2022] Open
Abstract
Growing evidence shows that intrinsic circadian clocks are tightly related to cardiovascular functions. The diurnal changes in blood pressure and heart rate are well known circadian rhythms. Endothelial function, platelet aggregation and thrombus formation exhibit circadian changes as well. The onset of many cardiovascular diseases (CVDs) or events, such as myocardial infarction, stroke, arrhythmia, and sudden cardiac death, also exhibits temporal trends. Furthermore, there is strong evidence from animal models and epidemiological studies showing that disruption of circadian rhythms is a significant risk factor for many CVDs, and the intervention of CVDs may have a time dependent effect. In this mini review, we summarized recent advances in our understanding of the relationship between circadian rhythm and cardiovascular physiology and diseases including blood pressure regulation and myocardial infarction.
Collapse
Affiliation(s)
- Lihong Chen
- The Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania Philadelphia, PA, USA ; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania Philadelphia, PA, USA
| | - Guangrui Yang
- The Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania Philadelphia, PA, USA ; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania Philadelphia, PA, USA
| |
Collapse
|
45
|
Pearce WJ. In cerebrovascular circadian rhythms, EETs keep the beat. Focus on "Rhythmic expression of cytochrome P450 epoxygenases CYP4x1 and CYP2c11 in the rat brain and vasculature". Am J Physiol Cell Physiol 2014; 307:C986-8. [PMID: 25273881 DOI: 10.1152/ajpcell.00327.2014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- William J Pearce
- Loma Linda University School of Medicine, Loma Linda, California
| |
Collapse
|
46
|
Hepatic overexpression of the prodomain of furin lessens progression of atherosclerosis and reduces vascular remodeling in response to injury. Atherosclerosis 2014; 236:121-30. [PMID: 25026302 DOI: 10.1016/j.atherosclerosis.2014.06.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Revised: 06/16/2014] [Accepted: 06/18/2014] [Indexed: 11/24/2022]
Abstract
OBJECTIVE Atherosclerosis is a complex disease, involving elevated LDL-c, lipid accumulation in the blood vessel wall, foam cell formation and vascular dysfunction. Lowering plasma LDL-c is the cornerstone of current management of cardiovascular disease. However, new approaches which reduce plasma LDL-c and lessen the pathological vascular remodeling occurring in the disease should also have therapeutic value. Previously, we found that overexpression of profurin, the 83-amino acid prodomain of the proprotein convertase furin, lowered plasma HDL levels in wild-type mice. The question that remained was whether it had effects on apolipoprotein B (ApoB)-containing lipoproteins. METHODS Adenovirus mediated overexpression of hepatic profurin in Ldlr(-/-)mice and wild-type mice were used to evaluate effects of profurin on ApoB-containing lipoproteins, atherosclerosis and vascular remodeling. RESULTS Hepatic profurin overexpression resulted in a significant reduction in atherosclerotic lesion development in Ldlr(-/-)mice and a robust reduction in plasma LDL-c. Metabolic studies revealed lower secretion of ApoB and triglycerides in VLDL particles. Mechanistic studies showed that in the presence of profurin, hepatic ApoB, mainly ApoB100, was degraded by proteasomes. There was no effect on ApoB mRNA expression. Importantly, short-term hepatic profurin overexpression did not result in hepatic lipid accumulation. Blood vessel wall thickening caused by either wire-induced femoral artery injury or common carotid artery ligation was reduced. Profurin expression inhibited proliferation and migration in vascular smooth muscle cells in vitro. CONCLUSION These results indicate that a profurin-based therapy has the potential to treat atherosclerosis by improving metabolic lipid profiles and reducing both atherosclerotic lesion development and pathological vascular remodeling.
Collapse
|
47
|
Leloup AJ, Fransen P, Van Hove CE, Demolder M, De Keulenaer GW, Schrijvers DM. Applanation Tonometry in Mice. Hypertension 2014; 64:195-200. [DOI: 10.1161/hypertensionaha.114.03312] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Arthur J.A. Leloup
- From the Laboratories of Physiopharmacology Department of Pharmaceutical Sciences (A.J.A.L., P.F., M.D., G.W.D.K., D.M.S.) and Pharmacology Faculty of Medicine (C.E.V.H.), University of Antwerp, Antwerp, Belgium
| | - Paul Fransen
- From the Laboratories of Physiopharmacology Department of Pharmaceutical Sciences (A.J.A.L., P.F., M.D., G.W.D.K., D.M.S.) and Pharmacology Faculty of Medicine (C.E.V.H.), University of Antwerp, Antwerp, Belgium
| | - Cor E. Van Hove
- From the Laboratories of Physiopharmacology Department of Pharmaceutical Sciences (A.J.A.L., P.F., M.D., G.W.D.K., D.M.S.) and Pharmacology Faculty of Medicine (C.E.V.H.), University of Antwerp, Antwerp, Belgium
| | - Marc Demolder
- From the Laboratories of Physiopharmacology Department of Pharmaceutical Sciences (A.J.A.L., P.F., M.D., G.W.D.K., D.M.S.) and Pharmacology Faculty of Medicine (C.E.V.H.), University of Antwerp, Antwerp, Belgium
| | - Gilles W. De Keulenaer
- From the Laboratories of Physiopharmacology Department of Pharmaceutical Sciences (A.J.A.L., P.F., M.D., G.W.D.K., D.M.S.) and Pharmacology Faculty of Medicine (C.E.V.H.), University of Antwerp, Antwerp, Belgium
| | - Dorien M. Schrijvers
- From the Laboratories of Physiopharmacology Department of Pharmaceutical Sciences (A.J.A.L., P.F., M.D., G.W.D.K., D.M.S.) and Pharmacology Faculty of Medicine (C.E.V.H.), University of Antwerp, Antwerp, Belgium
| |
Collapse
|
48
|
Jensen LD, Gyllenhaal C, Block K. Circadian angiogenesis. Biomol Concepts 2014; 5:245-56. [DOI: 10.1515/bmc-2014-0009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Accepted: 05/08/2014] [Indexed: 12/25/2022] Open
Abstract
AbstractDaily rhythms of light/darkness, activity/rest and feeding/fasting are important in human physiology and their disruption (for example by frequent changes between day and night shifts) increases the risk of disease. Many of the diseases found to be associated with such disrupted circadian lifestyles, including cancer, cardiovascular diseases, metabolic disorders and neurological diseases, depend on pathological de-regulation of angiogenesis, suggesting that disrupting the circadian clock will impair the physiological regulation of angiogenesis leading to development and progression of these diseases. Today there is little known regarding circadian regulation of pathological angiogenesis but there is some evidence that supports both direct and indirect regulation of angiogenic factors by the cellular circadian clock machinery, as well as by circulating circadian factors, important for coordinating circadian rhythms in the organism. Through highlighting recent advances both in pre-clinical and clinical research on various diseases including cancer, cardiovascular disorders and obesity, we will here present an overview of the available knowledge on the importance of circadian regulation of angiogenesis and discuss how the circadian clock may provide alternative targets for pro- or anti-angiogenic therapy in the future.
Collapse
Affiliation(s)
| | | | - Keith Block
- 3The Block Center for Integrative Cancer Treatment, 60077 Skokie, IL, USA
| |
Collapse
|
49
|
Chen S, Ding Y, Zhang Z, Wang H, Liu C. Hyperlipidaemia impairs the circadian clock and physiological homeostasis of vascular smooth muscle cells via the suppression of Smarcd1. J Pathol 2014; 233:159-69. [PMID: 24615205 DOI: 10.1002/path.4338] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 01/25/2014] [Accepted: 02/13/2014] [Indexed: 11/06/2022]
Abstract
Many mammalian physiological processes show diurnal oscillation and are controlled by a circadian clock. Disruption of the circadian clock has been implicated in the pathogenesis of cardiovascular disorders, but the mechanism through which clock and vessel function are integrated is unclear. Here we show that the rhythmicity of key clock genes and Smarcd1, a member of the SWI/SNF chromatin remodelling complex family, is suppressed in the layer of vascular smooth muscle cells (VSMCs) of the thoracic aorta of hyperlipidaemic rats fed a high-fat diet (HFD). Smarcd1 stimulates the transcription of clock genes, notably bmal1, through co-activation of the nuclear orphan receptor RORα in VSMCs. The co-activation of Smarcd1 and RORα is dependent on the mediation of PGC-1α, a transcriptional co-activator. Pathophysiologically, Smarcd1 inhibits VSMC proliferation and migration by blocking cell cycle re-entry and via the activation of kinase signalling pathways. Our results demonstrate that Smarcd1 is a critical node integrating the circadian clock and VSMC physiological homeostasis.
Collapse
Affiliation(s)
- Siyu Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology and College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu, People's Republic of China
| | | | | | | | | |
Collapse
|
50
|
Anea CB, Zhang M, Chen F, Ali MI, Hart CMM, Stepp DW, Kovalenkov YO, Merloiu AM, Pati P, Fulton D, Rudic RD. Circadian clock control of Nox4 and reactive oxygen species in the vasculature. PLoS One 2013; 8:e78626. [PMID: 24205282 PMCID: PMC3808297 DOI: 10.1371/journal.pone.0078626] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 09/14/2013] [Indexed: 12/30/2022] Open
Abstract
Recent studies have shown that circadian clock disruption is associated with pathological remodeling in the arterial structure and vascular stiffness. Moreover, chronic circadian disruption is associated with dysfunction in endothelial responses and signaling. Reactive oxygen species have emerged as key regulators in vascular pathology. Previously, we have demonstrated that circadian clock dysfunction exacerbates superoxide production through eNOS uncoupling. To date, the impact of circadian clock mutation on vascular NADPH oxidase expression and function is not known. The goal in the current study was to determine if the circadian clock controls vascular Nox4 expression and hydrogen peroxide formation in arteries, particularly in endothelial and vascular smooth muscle cells. In aorta, there was an increase in hydrogen peroxide and Nox4 expression in mice with a dysfunctional circadian rhythm (Bmal1-KO mice). In addition, the Nox4 gene promoter is activated by the core circadian transcription factors. Lastly, in synchronized cultured human endothelial cells, Nox4 gene expression exhibited rhythmic oscillations. These data reveal that the circadian clock plays an important role in the control of Nox4 and disruption of the clock leads to subsequent production of reaction oxygen species.
Collapse
Affiliation(s)
- Ciprian B. Anea
- Department of Pharmacology & Toxicology, Georgia Regents University, Augusta, Georgia, United States of America
| | - Maoxiang Zhang
- Department of Pharmacology & Toxicology, Georgia Regents University, Augusta, Georgia, United States of America
| | - Feng Chen
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia, United States of America
| | - M. Irfan Ali
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia, United States of America
| | - C. Michael M. Hart
- Department of Medicine, Atlanta Veterans Affairs and Emory University Medical Center, Atlanta, Georgia, United States of America
| | - David W. Stepp
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia, United States of America
- Department of Physiology, Georgia Regents University, Augusta, Georgia, United States of America
| | - Yevgeniy O. Kovalenkov
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia, United States of America
| | - Ana-Maria Merloiu
- Department of Pharmacology & Toxicology, Georgia Regents University, Augusta, Georgia, United States of America
| | - Paramita Pati
- Department of Pharmacology & Toxicology, Georgia Regents University, Augusta, Georgia, United States of America
| | - David Fulton
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia, United States of America
| | - R. Daniel Rudic
- Department of Pharmacology & Toxicology, Georgia Regents University, Augusta, Georgia, United States of America
- * E-mail:
| |
Collapse
|