1
|
Rezazadeh S, Salami SR, Hosseini M, Oster H, Saebipour MR, Hassanzadeh-Taheri MM, Shoorei H. Investigating the resilience of kidneys in rats exposed to chronic partial sleep deprivation and circadian rhythm disruption as disruptive interventions. Neurobiol Sleep Circadian Rhythms 2025; 18:100109. [PMID: 39720585 PMCID: PMC11664416 DOI: 10.1016/j.nbscr.2024.100109] [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: 09/17/2024] [Revised: 11/29/2024] [Accepted: 11/29/2024] [Indexed: 12/26/2024] Open
Abstract
Sleep is a vital biological function that significantly influences overall health. While sleep deprivation (SD) and circadian rhythm disruption are known to negatively impact various organs, their specific effects on kidney function remain understudied. This study aimed to investigate the impact of chronic partial sleep deprivation and circadian rhythm disruption on renal function in rats, providing insights into the relationship between sleep disturbances and kidney health. A total of 40 male Wistar rats were divided into five groups: a control group, a group with circadian rhythm disruption (CIR), a group with sleep deprivation during the light phase (SD-AM), a group with sleep deprivation during the dark phase (SD-PM), and a group with combined sleep deprivation and circadian rhythm disruption (SD-CIR). Sleep deprivation was induced using a specialized machine, depriving rats of sleep for 4 h daily, while circadian rhythm disruption was achieved through a 3.5-h light/dark cycle. After four weeks, kidney tissues and blood samples were collected for histological and biochemical analyses. The results showed that all experimental groups exhibited reduced water intake, with the CIR and SD-CIR groups also showing significantly lower food intake and reduced weight gain compared to controls. Oxidative stress markers revealed increased serum malondialdehyde (MDA) levels in the SD-PM and SD-CIR groups. Despite these metabolic and oxidative changes, histological examination of the kidneys revealed no significant alterations in renal structure or function across the groups. This study highlights the negative effects of chronic partial sleep deprivation and circadian rhythm disruption on feeding behavior, weight gain, and oxidative stress in rats. However, these interventions did not significantly alter renal structure or function. Further research is needed to explore the physiological mechanisms underlying these findings and the potential long-term effects of sleep disturbances on kidney health.
Collapse
Affiliation(s)
- Shirin Rezazadeh
- Department of Anatomical Sciences, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Saeed Rastgoo Salami
- Department of Anatomical Sciences, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Mehran Hosseini
- Cellular and Molecular Research Center, Department of Anatomical Sciences, Birjand University of Medical Sciences, Birjand, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Henrik Oster
- Institute of Neurobiology, Center of Brain, Behavior & Metabolism, University of Lübeck, Lübeck, Germany
| | - Mohammad Reza Saebipour
- Department of Anatomical Sciences, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | | | - Hamed Shoorei
- Cellular and Molecular Research Center, Department of Anatomical Sciences, Birjand University of Medical Sciences, Birjand, Iran
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
2
|
Myung J, Vitet H, Truong VH, Ananthasubramaniam B. The role of the multiplicity of circadian clocks in mammalian systems. Sleep Med 2025; 131:106518. [PMID: 40222295 DOI: 10.1016/j.sleep.2025.106518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/22/2025] [Accepted: 04/09/2025] [Indexed: 04/15/2025]
Abstract
Circadian clocks regulate rhythmic biological processes in nearly every tissue, aligning physiology and behavior with the 24-h light-dark cycle. While the central circadian clock in the suprachiasmatic nucleus (SCN) has been extensively studied, emerging evidence indicates that virtually every cell in the body possesses its own locally autonomous circadian clock. This raises a fundamental question: why do multicellular organisms utilize multiple circadian clocks instead of a single master clock broadcasting time cues? Here, we examine how distributed local clocks differ from phase-resettable cycles and ensure robust temporal scheduling of physiological processes. We discuss how internal entrainment among local clocks governs self-sustained, yet flexible, circadian organization of tissue-specific responses to environmental changes. We also examine how the organization of clocks contributes to seasonal homeostasis, and the implications for disease when coordination among these clocks is disrupted.
Collapse
Affiliation(s)
- Jihwan Myung
- Graduate Institute of Mind, Brain and Consciousness (GIMBC), Taipei Medical University, New Taipei City 235, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Hélène Vitet
- Graduate Institute of Mind, Brain and Consciousness (GIMBC), Taipei Medical University, New Taipei City 235, Taiwan
| | - Vuong Hung Truong
- Graduate Institute of Mind, Brain and Consciousness (GIMBC), Taipei Medical University, New Taipei City 235, Taiwan
| | | |
Collapse
|
3
|
Le DC, Ngo MHT, Kuo YC, Chen SH, Lin CY, Ling TY, Pham QTT, Au HK, Myung J, Huang YH. Secretome from estrogen-responding human placenta-derived mesenchymal stem cells rescues ovarian function and circadian rhythm in mice with cyclophosphamide-induced primary ovarian insufficiency. J Biomed Sci 2024; 31:95. [PMID: 39390588 PMCID: PMC11468397 DOI: 10.1186/s12929-024-01085-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 09/11/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Primary ovarian insufficiency (POI) is an early decline in ovarian function that leads to ovarian failure. Conventional treatments for POI are inadequate, and treatments based on mesenchymal stem cells (MSCs) have emerged as an option. However, the lack of consideration of the estrogen niche in ovarian tissue significantly reduces the therapeutic efficacy, with an unclear mechanism in the MSCs in POI treatment. Furthermore, the disruption of circadian rhythm associated with POI has not been previously addressed. METHODS Conditioned medium (CM) and estradiol-conditioned medium (E2-CM) were generated from estrogen receptor positive MSCs (ER+pcMSCs). Chemotherapy-induced POI models were established using C57BL/6 mice (in vivo) and KGN cells (in vitro) treated with cyclophosphamide (CTX) or 4-hydroperoxycyclophosphamide (4-OOH-CP). Gene/protein expressions were detected using RT-qPCR, Western blotting, and immunohistochemistry assays. Locomotor activity was monitored for behavioral circadian rhythmicity. Cytokine arrays and miRNA analysis were conducted to analyze potential factors within CM/E2-CM. RESULTS The secretome of ER+pcMSCs (CM and E2-CM) significantly reduced the CTX-induced defects in ovarian folliculogenesis and circadian rhythm. CM/E2-CM also reduced granulosa cell apoptosis and rescued angiogenesis in POI ovarian tissues. E2-CM had a more favorable effect than the CM. Notably, ER+pcMSC secretome restored CTX-induced circadian rhythm defects, including the gene expressions associated with the ovarian circadian clock (e.g., Rora, E4bp4, Rev-erbα, Per2 and Dbp) and locomotor activity. Additionally, the cytokine array analysis revealed a significant increase in cytokines and growth factors associated with immunomodulation and angiogenesis, including angiogenin. Neutralizing the angiogenin in CM/E2-CM significantly reduced its ability to promote HUVEC tube formation in vitro. Exosomal miRNA analysis revealed the miRNAs involved in targeting the genes associated with POI rescue (PTEN and PDCD4), apoptosis (caspase-3, BIM), estrogen synthesis (CYP19A1), ovarian clock regulation (E4BP4, REV-ERBα) and fibrosis (COL1A1). CONCLUSION This study is the first to demonstrate that, in considering the estrogen niche in ovarian tissue, an estrogen-priming ER+pcMSC secretome achieved ovarian regeneration and restored the circadian rhythm in a CTX-induced POI mouse model. The potential factors involved include angiogenin and exosomal miRNAs in the ER+pcMSC secretome. These findings offer insights into potential stem cell therapies for chemotherapy-induced POI and circadian rhythm disruption.
Collapse
Affiliation(s)
- Duy-Cuong Le
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- Laboratory, Vinmec International Hospital, Minh Khai Street, Hai Ba Trung, Hanoi, Vietnam
| | - Mai-Huong T Ngo
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Yung-Che Kuo
- TMU Research Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Shu-Hwa Chen
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Chung-Yen Lin
- Institute of Information Science, Academia Sinica, Taipei, 11529, Taiwan
- Institute of Fishery Sciences, College of Life Science, National Taiwan University, Taipei, 10617, Taiwan
- Genome and Systems Biology Degree Program, National Taiwan University, Taipei, 10617, Taiwan
| | - Thai-Yen Ling
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Quoc Thao Trang Pham
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Heng-Kien Au
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
- Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.
- Department of Obstetrics and Gynecology, Taipei Medical University Hospital, Taipei, 11042, Taiwan.
- Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei, 11042, Taiwan.
| | - Jihwan Myung
- Graduate Institute of Mind, Brain and Consciousness, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.
- Brain and Consciousness Research Centre (BCRC), TMU-Shuang Ho Hospital, New Taipei City, 23561, Taiwan.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Wuxing Street, Taipei, 11031, Taiwan.
| | - Yen-Hua Huang
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.
- TMU Research Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.
- Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei, 11042, Taiwan.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Wuxing Street, Taipei, 11031, Taiwan.
| |
Collapse
|
4
|
Xiong Y, Zhong Q, Zhang Y, Liu Z, Wang X. The association between circadian syndrome and chronic kidney disease in an aging population: a 4-year follow-up study. Front Endocrinol (Lausanne) 2024; 15:1338110. [PMID: 38737554 PMCID: PMC11082579 DOI: 10.3389/fendo.2024.1338110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 03/21/2024] [Indexed: 05/14/2024] Open
Abstract
Introduction Circadian syndrome (CircS) is proposed as a novel risk cluster based on reduced sleep duration, abdominal obesity, depression, hypertension, dyslipidemia and hyperglycemia. However, the association between CircS and chronic kidney disease (CKD) remains unclear. To investigate the cross-sectional and longitudinal association between CircS and CKD, this study was performed. Methods A national prospective cohort (China Health and Retirement Longitudinal Study, CHARLS) was used in this study. To define CKD, the estimated glomerular filtration rate (eGFR) was calculated based on the 2012 CKD-EPI creatinine-cystatin C equation. Participants with eGFR <60 mL.min-1/1.73/m2 were diagnosed with CKD. Multivariate binary logistic regression was used to assess the cross-sectional association between CircS and CKD. Subgroup and interactive analyses were performed to determine the interactive effects of covariates. In the sensitivity analysis, the obese population was excluded and another method for calculating the eGFR was used to verify the robustness of previous findings. In addition, participants without CKD at baseline were followed up for four years to investigate the longitudinal relationship between CircS and CKD. Results A total of 6355 participants were included in this study. In the full model, CircS was positively associated with CKD (OR = 1.28, 95% CI = 1.04-1.59, P < 0.05). As per one increase of CircS components, there was a 1.11-fold (95% CI = 1.04-1.18, P < 0.05) risk of prevalent CKD in the full model. A significant interactive effect of hyperuricemia in the CircS-CKD association (P for interaction < 0.01) was observed. Sensitivity analyses excluding the obese population and using the 2009 CKD-EPI creatinine equation to diagnose CKD supported the positive correlation between CircS and CKD. In the 2011-2015 follow-up cohort, the CircS group had a 2.18-fold risk of incident CKD (95% CI = 1.33-3.58, P < 0.01) in the full model. The OR was 1.29 (95% CI = 1.10-1.51, P < 0.001) with per one increase of CircS components. Conclusion CircS is a risk factor for CKD and may serve as a predictor of CKD for early identification and intervention.
Collapse
Affiliation(s)
- Yang Xiong
- Department of Urology and Andrology Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qian Zhong
- Department of Endocrinology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yangchang Zhang
- Department of Public Health, Capital Medical University, Beijing, China
| | - Zhihong Liu
- Department of Urology and Andrology Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xianding Wang
- Department of Urology and Andrology Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Kidney Transplant Center, Transplant Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
5
|
Truong VH, Myung J. LocoBox: Modular Hardware and Open-Source Software for Circadian Entrainment and Behavioral Monitoring in Home Cages. SENSORS (BASEL, SWITZERLAND) 2023; 23:9469. [PMID: 38067841 PMCID: PMC10708669 DOI: 10.3390/s23239469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/05/2023] [Accepted: 11/26/2023] [Indexed: 12/18/2023]
Abstract
Day-night locomotor activities are the most readily observed outputs of the circadian (~24-h period) clock in many animals. Temporal patterns of the light-dark schedule serve as input to the clock. While circadian activity patterns under various lighting conditions have been observed and documented, the full extent of circadian locomotor activities by genotype and entrainment remains uncharacterized. To facilitate large-scale, parallel cataloging of circadian input-output patterns, we created the LocoBox, an easy-to-construct and easy-to-operate system that can control environmental light with flexible entrainment scenarios combined with the T-cycle and measure locomotor activities in individual home cages. The LocoBox is made using economical, common components, and normal breeding cages can be used for long-term recording. We provide details of the components and blueprints, along with software programs for Arduino and a Python-based graphical user interface (GUI), so that the system can be easily replicated in other laboratories.
Collapse
Affiliation(s)
- Vuong Hung Truong
- Graduate Institute of Mind, Brain and Consciousness (GIMBC), Taipei Medical University, New Taipei City 235, Taiwan
- Brain and Consciousness Research Centre (BCRC), TMU-Shuang Ho Hospital, New Taipei City 235, Taiwan
| | - Jihwan Myung
- Graduate Institute of Mind, Brain and Consciousness (GIMBC), Taipei Medical University, New Taipei City 235, Taiwan
- Brain and Consciousness Research Centre (BCRC), TMU-Shuang Ho Hospital, New Taipei City 235, Taiwan
- Graduate Institute of Medical Sciences (GIMS), Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
6
|
Myung J, Hong S, Schmal C, Vitet H, Wu MY. Weak synchronization can alter circadian period length: implications for aging and disease conditions. Front Neurosci 2023; 17:1242800. [PMID: 37829718 PMCID: PMC10564985 DOI: 10.3389/fnins.2023.1242800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/29/2023] [Indexed: 10/14/2023] Open
Abstract
The synchronization of multiple oscillators serves as the central mechanism for maintaining stable circadian rhythms in physiology and behavior. Aging and disease can disrupt synchronization, leading to changes in the periodicity of circadian activities. While our understanding of the circadian clock under synchronization has advanced significantly, less is known about its behavior outside synchronization, which can also fall within a predictable domain. These states not only impact the stability of the rhythms but also modulate the period length. In C57BL/6 mice, aging, diseases, and removal of peripheral circadian oscillators often result in lengthened behavioral circadian periods. Here, we show that these changes can be explained by a surprisingly simple mathematical relationship: the frequency is the reciprocal of the period, and its distribution becomes skewed when the period distribution is symmetric. The synchronized frequency of a population in the skewed distribution and the macroscopic frequency of combined oscillators differ, accounting for some of the atypical circadian period outputs observed in networks without synchronization. Building on this finding, we investigate the dynamics of circadian outputs in the context of aging and disease, where synchronization is weakened.
Collapse
Affiliation(s)
- Jihwan Myung
- Graduate Institute of Mind, Brain and Consciousness (GIMBC), Taipei Medical University, Taipei City, Taiwan
- Brain and Consciousness Research Centre (BCRC), TMU-Shuang Ho Hospital, New Taipei City, Taiwan
- Computational Neuroscience Unit, Okinawa Institute of Science and Technology, Okinawa, Japan
| | - Sungho Hong
- Computational Neuroscience Unit, Okinawa Institute of Science and Technology, Okinawa, Japan
| | - Christoph Schmal
- Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Hélène Vitet
- Graduate Institute of Mind, Brain and Consciousness (GIMBC), Taipei Medical University, Taipei City, Taiwan
- Brain and Consciousness Research Centre (BCRC), TMU-Shuang Ho Hospital, New Taipei City, Taiwan
- Department of Pediatrics, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Mei-Yi Wu
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
- TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
7
|
Sharma RK, Kamble SH, Krishnan S, Gomes J, To B, Li S, Liu IC, Gumz ML, Mohandas R. Involvement of lysyl oxidase in the pathogenesis of arterial stiffness in chronic kidney disease. Am J Physiol Renal Physiol 2023; 324:F364-F373. [PMID: 36825626 PMCID: PMC10069822 DOI: 10.1152/ajprenal.00239.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 02/01/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Patients with chronic kidney disease (CKD) are at increased risk for adverse cardiovascular events. CKD is associated with increases in arterial stiffness, whereas improvements in arterial stiffness correlate with better survival. However, arterial stiffness is increased early in CKD, suggesting that there might be additional factors, unique to kidney disease, that increase arterial stiffness. Lysyl oxidase (LOX) is a key mediator of collagen cross linking and matrix remodeling. LOX is predominantly expressed in the cardiovascular system, and its upregulation has been associated with increased tissue stiffening and extracellular matrix remodeling. Thus, this study was designed to evaluate the role of increased LOX activity in inducing aortic stiffness in CKD and whether β-aminopropionitrile (BAPN), a LOX inhibitor, could prevent aortic stiffness by reducing collagen cross linking. Eight-week-old male C57BL/6 mice were subjected to 5/6 nephrectomy (Nx) or sham surgery. Two weeks after surgery, mice were randomized to BAPN (300 mg/kg/day in water) or vehicle treatment for 4 wk. Aortic stiffness was assessed by pulse wave velocity (PWV) using Doppler ultrasound. Aortic levels of LOX were assessed by ELISA, and cross-linked total collagen levels were analyzed by mass spectrometry and Sircol assay. Nx mice showed increased PWV and aortic wall remodeling compared with control mice. Collagen cross linking was increased in parallel with the increases in total collagen in the aorta of Nx mice. In contrast, Nx mice that received BAPN treatment showed decreased cross-linked collagens and PWV compared with that received vehicle treatment. Our results indicated that LOX might be an early and key mediator of aortic stiffness in CKD.NEW & NOTEWORTHY Arterial stiffness in CKD is associated with adverse cardiovascular outcomes. However, the mechanisms underlying increased aortic stiffness in CKD are unclear. Herein, we demonstrated that 1) increased aortic stiffness in CKD is independent of hypertension and calcification and 2) LOX-mediated changes in extracellular matrix are at least in part responsible for increased aortic stiffness in CKD. Prevention of excess LOX may have therapeutic potential in alleviating increased aortic stiffness and improving cardiovascular disease in CKD.
Collapse
Affiliation(s)
- Ravindra K Sharma
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida College of Medicine, Gainesville, Florida, United States
| | - Shyam H Kamble
- Department of Pharmacology, University of Florida College of Medicine, Gainesville, Florida, United States
| | - Suraj Krishnan
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida College of Medicine, Gainesville, Florida, United States
| | - Joshua Gomes
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida College of Medicine, Gainesville, Florida, United States
| | - Brandon To
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida College of Medicine, Gainesville, Florida, United States
| | - Shiyu Li
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida College of Medicine, Gainesville, Florida, United States
| | - I-Chia Liu
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida College of Medicine, Gainesville, Florida, United States
| | - Michelle L Gumz
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida College of Medicine, Gainesville, Florida, United States
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, Florida, United States
| | - Rajesh Mohandas
- Division of Nephrology and Hypertension, Louisiana State University Health Sciences Center School of Medicine, New Orleans, Louisiana, United States
| |
Collapse
|
8
|
Luo YM, Yang SD, Wen MY, Wang B, Liu JH, Li ST, Li YY, Cheng H, Zhao LL, Li SM, Jiang JJ. Insights into the mechanisms of triptolide nephrotoxicity through network pharmacology-based analysis and RNA-seq. FRONTIERS IN PLANT SCIENCE 2023; 14:1144583. [PMID: 36959927 PMCID: PMC10027700 DOI: 10.3389/fpls.2023.1144583] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 02/17/2023] [Indexed: 06/18/2023]
Abstract
INTRODUCTION Triptolide (TPL) is a promising plant-derived compound for clinical therapy of multiple human diseases; however, its application was limited considering its toxicity. METHODS To explore the underlying molecular mechanism of TPL nephrotoxicity, a network pharmacology based approach was utilized to predict candidate targets related with TPL toxicity, followed by deep RNA-seq analysis to characterize the features of three transcriptional elements include protein coding genes (PCGs), long noncoding RNAs (lncRNAs) and circular RNAs (circRNAs) as well as their associations with nephrotoxicity in rats with TPL treatment. RESULTS & DISCUSSION Although the deeper mechanisms of TPL nephrotoxcity remain further exploration, our results suggested that c-Jun is a potential target of TPL and Per1 related circadian rhythm signaling is involved in TPL induced renal toxicity.
Collapse
Affiliation(s)
- Yue-Ming Luo
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Shu-Dong Yang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Miao-Yu Wen
- Department of Geriatrics, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Bing Wang
- Department of Nephrology, Shenzhen Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jia-Hui Liu
- Department of Nephrology, Shenzhen Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Si-Ting Li
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yu-Yan Li
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Hong Cheng
- Department of Geriatrics, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Li-Li Zhao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
- Graduate school of Clinical Medicine, Anhui Medical University, Hefei, China
| | - Shun-Min Li
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Jian-Jun Jiang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| |
Collapse
|
9
|
Potential Role of the Circadian Clock in the Regulation of Cancer Stem Cells and Cancer Therapy. Int J Mol Sci 2022; 23:ijms232214181. [PMID: 36430659 PMCID: PMC9698777 DOI: 10.3390/ijms232214181] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022] Open
Abstract
Circadian rhythms, including sleep/wake cycles as well as hormonal, immune, metabolic, and cell proliferation rhythms, are fundamental biological processes driven by a cellular time-keeping system called the circadian clock. Disruptions in these rhythms due to genetic alterations or irregular lifestyles cause fundamental changes in physiology, from metabolism to cellular proliferation and differentiation, resulting in pathological consequences including cancer. Cancer cells are not uniform and static but exist as different subtypes with phenotypic and functional differences in the tumor microenvironment. At the top of the heterogeneous tumor cell hierarchy, cancer stem cells (CSCs), a self-renewing and multi-potent cancer cell type, are most responsible for tumor recurrence and metastasis, chemoresistance, and mortality. Phenotypically, CSCs are associated with the epithelial-mesenchymal transition (EMT), which confers cancer cells with increased motility and invasion ability that is characteristic of malignant and drug-resistant stem cells. Recently, emerging studies of different cancer types, such as glioblastoma, leukemia, prostate cancer, and breast cancer, suggest that the circadian clock plays an important role in the maintenance of CSC/EMT characteristics. In this review, we describe recent discoveries regarding how tumor intrinsic and extrinsic circadian clock-regulating factors affect CSC evolution, highlighting the possibility of developing novel chronotherapeutic strategies that could be used against CSCs to fight cancer.
Collapse
|
10
|
Costello HM, Johnston JG, Juffre A, Crislip GR, Gumz ML. Circadian clocks of the kidney: function, mechanism, and regulation. Physiol Rev 2022; 102:1669-1701. [PMID: 35575250 PMCID: PMC9273266 DOI: 10.1152/physrev.00045.2021] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 05/03/2022] [Accepted: 05/07/2022] [Indexed: 11/22/2022] Open
Abstract
An intrinsic cellular circadian clock is located in nearly every cell of the body. The peripheral circadian clocks within the cells of the kidney contribute to the regulation of a variety of renal processes. In this review, we summarize what is currently known regarding the function, mechanism, and regulation of kidney clocks. Additionally, the effect of extrarenal physiological processes, such as endocrine and neuronal signals, on kidney function is also reviewed. Circadian rhythms in renal function are an integral part of kidney physiology, underscoring the importance of considering time of day as a key biological variable. The field of circadian renal physiology is of tremendous relevance, but with limited physiological and mechanistic information on the kidney clocks this is an area in need of extensive investigation.
Collapse
Affiliation(s)
- Hannah M Costello
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
| | - Jermaine G Johnston
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
- North Florida/South Georgia Malcom Randall Department of Veterans Affairs Medical Center, Gainesville, Florida
| | - Alexandria Juffre
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida
| | - G Ryan Crislip
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
| | - Michelle L Gumz
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
- North Florida/South Georgia Malcom Randall Department of Veterans Affairs Medical Center, Gainesville, Florida
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, Florida
| |
Collapse
|
11
|
Fang Y, Son S, Yang J, Oh S, Jo SK, Cho W, Kim MG. Perturbation of Circadian Rhythm Is Associated with Increased Prevalence of Chronic Kidney Disease: Results of the Korean Nationwide Population-Based Survey. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:5732. [PMID: 35565131 PMCID: PMC9102791 DOI: 10.3390/ijerph19095732] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/02/2022] [Accepted: 05/06/2022] [Indexed: 02/01/2023]
Abstract
Disturbances in circadian rhythms cause several health problems, such as psychosis, metabolic syndrome, and cancer; however, their effect on kidney disease remains unclear. This study aimed to evaluate the association between chronic kidney disease (CKD) and sleep disturbance in a Korean adult population. A total of 17,408 participants who completed the National Health and Nutrition Examination Survey from 2016 to 2018 were assessed for their sleep patterns and renal function. CKD was defined as an estimated glomerular filtration rate ≤ 60 mL/min/1.73 m² or a positive dipstick urinalysis. Sleep onset time and sleep duration showed significant differences between the control and CKD groups (p < 0.001). After adjusting for the covariates, sleep onset time rather than sleep duration was independently associated with incidence of CKD, and this association was more significant in people who were older, in women, and in those with low body mass index and no comorbidities. When comparing the prevalence of newly diagnosed CKD according to sleep onset time in a population with no CKD risk factors or no history of CKD, the early bedtime group showed an independent association with incidence of new CKD (odds ratio (OR), 1.535; 95% confidence interval (CI), 1.011−2.330) even after adjusting for covariates. Impaired circadian rhythm along with sleep disturbance could be associated with CKD development; therefore, sleep disturbance might be an important therapeutic target for CKD.
Collapse
Affiliation(s)
- Yina Fang
- Department of Internal Medicine, Korea University Anam Hospital, Seoul 02841, Korea; (Y.F.); (J.Y.); (S.O.); (S.-K.J.); (W.C.)
| | - Serhim Son
- Department of Biostatistics, Korea University College of Medicine, Seoul 02842, Korea;
| | - Jihyun Yang
- Department of Internal Medicine, Korea University Anam Hospital, Seoul 02841, Korea; (Y.F.); (J.Y.); (S.O.); (S.-K.J.); (W.C.)
| | - Sewon Oh
- Department of Internal Medicine, Korea University Anam Hospital, Seoul 02841, Korea; (Y.F.); (J.Y.); (S.O.); (S.-K.J.); (W.C.)
| | - Sang-Kyung Jo
- Department of Internal Medicine, Korea University Anam Hospital, Seoul 02841, Korea; (Y.F.); (J.Y.); (S.O.); (S.-K.J.); (W.C.)
| | - Wonyong Cho
- Department of Internal Medicine, Korea University Anam Hospital, Seoul 02841, Korea; (Y.F.); (J.Y.); (S.O.); (S.-K.J.); (W.C.)
| | - Myung-Gyu Kim
- Department of Internal Medicine, Korea University Anam Hospital, Seoul 02841, Korea; (Y.F.); (J.Y.); (S.O.); (S.-K.J.); (W.C.)
| |
Collapse
|
12
|
Fukumoto K, Ito K, Saer B, Taylor G, Ye S, Yamano M, Toriba Y, Hayes A, Okamura H, Fustin JM. Excess S-adenosylmethionine inhibits methylation via catabolism to adenine. Commun Biol 2022; 5:313. [PMID: 35383287 PMCID: PMC8983724 DOI: 10.1038/s42003-022-03280-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 03/09/2022] [Indexed: 11/23/2022] Open
Abstract
The global dietary supplement market is valued at over USD 100 billion. One popular dietary supplement, S-adenosylmethionine, is marketed to improve joints, liver health and emotional well-being in the US since 1999, and has been a prescription drug in Europe to treat depression and arthritis since 1975, but recent studies questioned its efficacy. In our body, S-adenosylmethionine is critical for the methylation of nucleic acids, proteins and many other targets. The marketing of SAM implies that more S-adenosylmethionine is better since it would stimulate methylations and improve health. Previously, we have shown that methylation reactions regulate biological rhythms in many organisms. Here, using biological rhythms to assess the effects of exogenous S-adenosylmethionine, we reveal that excess S-adenosylmethionine disrupts rhythms and, rather than promoting methylation, is catabolized to adenine and methylthioadenosine, toxic methylation inhibitors. These findings further our understanding of methyl metabolism and question the safety of S-adenosylmethionine as a supplement. S-adenosylmethionine (SAM) is a widely available dietary supplement. Exogenous SAM is catabolized to adenine, an inhibitor of adenosylhomocysteinase, leading to widespread methylation inhibition and disruption of circadian rhythms in vitro and in mice.
Collapse
Affiliation(s)
- Kazuki Fukumoto
- Kyoto University, Graduate School of Pharmaceutical Sciences, Department of Molecular Metabology, Kyoto, Japan.,Kokando Pharmaceutical Co., Ltd, Tokyo, Japan
| | - Kakeru Ito
- Kyoto University, Graduate School of Pharmaceutical Sciences, Department of Molecular Metabology, Kyoto, Japan
| | - Benjamin Saer
- The University of Manchester, Centre for Biological Timing, Manchester, UK
| | - George Taylor
- The University of Manchester, BioMS Core Facility, Manchester, UK
| | - Shiqi Ye
- Kyoto University, Graduate School of Pharmaceutical Sciences, Department of Molecular Metabology, Kyoto, Japan.,Cancer Epigenetics Laboratory, Francis Crick Institute, Cambridge, UK
| | - Mayu Yamano
- Kyoto University, Graduate School of Pharmaceutical Sciences, Department of Molecular Metabology, Kyoto, Japan
| | - Yuki Toriba
- Kyoto University, Graduate School of Pharmaceutical Sciences, Department of Molecular Metabology, Kyoto, Japan.,Master's Programme in Molecular Biology, Faculty of Science and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Andrew Hayes
- The University of Manchester, Genomics Technologies Core Facility, Manchester, UK
| | - Hitoshi Okamura
- Kyoto University, Graduate School of Medicine, Division of Physiology and Neurobiology, Kyoto, Japan.
| | - Jean-Michel Fustin
- Kyoto University, Graduate School of Pharmaceutical Sciences, Department of Molecular Metabology, Kyoto, Japan. .,The University of Manchester, Centre for Biological Timing, Manchester, UK.
| |
Collapse
|
13
|
Rhoads MK, Speed JS, Roth KJ, Zhang D, Jin C, Gamble KL, Pollock DM. Short-term daytime restricted feeding in rats with high salt impairs diurnal variation of Na + excretion. Am J Physiol Renal Physiol 2022; 322:F335-F343. [PMID: 35100821 PMCID: PMC8896996 DOI: 10.1152/ajprenal.00287.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 01/06/2023] Open
Abstract
Night shift work increases risk of cardiovascular disease associated with an irregular eating schedule. Elevating this risk is the high level of salt intake observed in the typical Western diet. Renal Na+ excretion has a distinct diurnal pattern, independent of time of intake, yet the interactions between the time of intake and the amount of salt ingested are not clear. The hypothesis of the present study was that limiting food intake to the typically inactive period in addition to high-salt (HS) feeding will disrupt the diurnal rhythm of renal Na+ excretion. Male Sprague-Dawley rats were placed on either normal-salt (NS; 0.49% NaCl) or HS (4% NaCl) diets. Rats were housed in metabolic cages and allowed food ad libitum and then subjected to inactive period time-restricted feeding (iTRF) for 5 days. As expected, rats fed NS and allowed food ad libitum had a diurnal pattern of Na+ excretion. The diurnal pattern of Na+ excretion was not significantly different after 5 days of iTRF compared with ad libitum rats. In response to HS, the diurnal pattern of Na+ excretion was similar to NS-fed rats. However, this pattern was attenuated after 5 days of HS iTRF. The diurnal excretion pattern of urinary aldosterone was abolished in both NS iTRF and HS iTRF rats. These data support the hypothesis that HS intake combined with iTRF impairs circadian mechanisms associated with renal Na+ excretion.NEW & NOTEWORTHY Timing of food intake normally has little effect on the diurnal pattern of Na+ and water excretion. However, rats on a high-salt diet were unable to maintain this pattern, yet K+ excretion was more readily adjusted to match timing of intake. These data support the hypothesis that Na+ and water homeostasis are impacted by timing of high-salt diets.
Collapse
Affiliation(s)
- Megan K Rhoads
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Joshua S Speed
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Kaehler J Roth
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Dingguo Zhang
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Chunhua Jin
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Karen L Gamble
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - David M Pollock
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
14
|
Multi-Modal Regulation of Circadian Physiology by Interactive Features of Biological Clocks. BIOLOGY 2021; 11:biology11010021. [PMID: 35053019 PMCID: PMC8772734 DOI: 10.3390/biology11010021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/21/2021] [Accepted: 12/23/2021] [Indexed: 12/26/2022]
Abstract
The circadian clock is a fundamental biological timing mechanism that generates nearly 24 h rhythms of physiology and behaviors, including sleep/wake cycles, hormone secretion, and metabolism. Evolutionarily, the endogenous clock is thought to confer living organisms, including humans, with survival benefits by adapting internal rhythms to the day and night cycles of the local environment. Mirroring the evolutionary fitness bestowed by the circadian clock, daily mismatches between the internal body clock and environmental cycles, such as irregular work (e.g., night shift work) and life schedules (e.g., jet lag, mistimed eating), have been recognized to increase the risk of cardiac, metabolic, and neurological diseases. Moreover, increasing numbers of studies with cellular and animal models have detected the presence of functional circadian oscillators at multiple levels, ranging from individual neurons and fibroblasts to brain and peripheral organs. These oscillators are tightly coupled to timely modulate cellular and bodily responses to physiological and metabolic cues. In this review, we will discuss the roles of central and peripheral clocks in physiology and diseases, highlighting the dynamic regulatory interactions between circadian timing systems and multiple metabolic factors.
Collapse
|
15
|
Abstract
Circadian clocks are biological timing mechanisms that generate 24-h rhythms of physiology and behavior, exemplified by cycles of sleep/wake, hormone release, and metabolism. The adaptive value of clocks is evident when internal body clocks and daily environmental cycles are mismatched, such as in the case of shift work and jet lag or even mistimed eating, all of which are associated with physiological disruption and disease. Studies with animal and human models have also unraveled an important role of functional circadian clocks in modulating cellular and organismal responses to physiological cues (ex., food intake, exercise), pathological insults (e.g. virus and parasite infections), and medical interventions (e.g. medication). With growing knowledge of the molecular and cellular mechanisms underlying circadian physiology and pathophysiology, it is becoming possible to target circadian rhythms for disease prevention and treatment. In this review, we discuss recent advances in circadian research and the potential for therapeutic applications that take patient circadian rhythms into account in treating disease.
Collapse
Affiliation(s)
- Yool Lee
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, Washington
| | - Jeffrey M. Field
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Amita Sehgal
- Howard Hughes Medical Institute, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
16
|
Chrobok L, Pradel K, Janik ME, Sanetra AM, Bubka M, Myung J, Ridla Rahim A, Klich JD, Jeczmien-Lazur JS, Palus-Chramiec K, Lewandowski MH. Intrinsic circadian timekeeping properties of the thalamic lateral geniculate nucleus. J Neurosci Res 2021; 99:3306-3324. [PMID: 34758124 DOI: 10.1002/jnr.24973] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 09/07/2021] [Accepted: 09/17/2021] [Indexed: 01/08/2023]
Abstract
Circadian rhythmicity in mammals is sustained by the central brain clock-the suprachiasmatic nucleus of the hypothalamus (SCN), entrained to the ambient light-dark conditions through a dense retinal input. However, recent discoveries of autonomous clock gene expression cast doubt on the supremacy of the SCN and suggest circadian timekeeping mechanisms devolve to local brain clocks. Here, we use a combination of molecular, electrophysiological, and optogenetic tools to evaluate intrinsic clock properties of the main retinorecipient thalamic center-the lateral geniculate nucleus (LGN) in male rats and mice. We identify the dorsolateral geniculate nucleus as a slave oscillator, which exhibits core clock gene expression exclusively in vivo. Additionally, we provide compelling evidence for intrinsic clock gene expression accompanied by circadian variation in neuronal activity in the intergeniculate leaflet and ventrolateral geniculate nucleus (VLG). Finally, our optogenetic experiments propose the VLG as a light-entrainable oscillator, whose phase may be advanced by retinal input at the beginning of the projected night. Altogether, this study for the first time demonstrates autonomous timekeeping mechanisms shaping circadian physiology of the LGN.
Collapse
Affiliation(s)
- Lukasz Chrobok
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Kamil Pradel
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Marcelina Elzbieta Janik
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Anna Magdalena Sanetra
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Monika Bubka
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Jihwan Myung
- Graduate Institute of Mind, Brain, and Consciousness, Taipei Medical University, Taipei, Taiwan.,Brain and Consciousness Research Centre, Taipei Medical University-Shuang Ho Hospital, Ministry of Health and Welfare, New Taipei City, Taiwan
| | - Amalia Ridla Rahim
- Graduate Institute of Mind, Brain, and Consciousness, Taipei Medical University, Taipei, Taiwan.,Brain and Consciousness Research Centre, Taipei Medical University-Shuang Ho Hospital, Ministry of Health and Welfare, New Taipei City, Taiwan
| | - Jasmin Daniela Klich
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Jagoda Stanislawa Jeczmien-Lazur
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Katarzyna Palus-Chramiec
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Marian Henryk Lewandowski
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| |
Collapse
|
17
|
Mishra R, Bethunaickan R, Berthier CC, Yi Z, Strohl JJ, Huerta PT, Zhang W, Davidson A. Reversible dysregulation of renal circadian rhythm in lupus nephritis. Mol Med 2021; 27:99. [PMID: 34488619 PMCID: PMC8419890 DOI: 10.1186/s10020-021-00361-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 08/23/2021] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND We have found disruption of expression of major transcriptional regulators of circadian rhythm in the kidneys of several mouse models of lupus nephritis. Here we define the consequence of this disturbance with respect to circadian gene expression and renal homeostatic function in a mouse model of lupus nephritis. METHODS Molecular profiling of kidneys from 47 young and 41 nephritic female NZB/W F1 mice was performed at 4 hourly intervals over a 24 h period. Disruption of major circadian transcriptional regulators was confirmed by qPCR. Molecular data was normalized and analyzed for rhythmicity using RAIN analysis. Serum aldosterone and glucose and urine sodium and potassium were measured at 4 hourly intervals in pre-nephritic and nephritic mice and blood pressure was measured every 4 h. Analyses were repeated after induction of complete remission of nephritis using combination cyclophosphamide and costimulatory blockade. RESULTS We show a profound alteration of renal circadian rhythms in mice with lupus nephritis affecting multiple renal pathways. Using Cosinor analysis we identified consequent alterations of renal homeostasis and metabolism as well as blood pressure dipper status. This circadian dysregulation was partially reversed by remission induction therapy. CONCLUSIONS Our studies indicate the role of inflammation in causing the circadian disruption and suggest that screening for loss of normal blood pressure dipping should be incorporated into LN management. The data also suggest a potential role for circadian agonists in the treatment of lupus nephritis.
Collapse
Affiliation(s)
- Rakesh Mishra
- Institute for Molecular Medicine, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Ramalingam Bethunaickan
- Institute for Molecular Medicine, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Celine C Berthier
- Department of Internal Medicine, Nephrology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Zhengzi Yi
- Department of Medicine, Mount Sinai Medical Center, One Gustave L. Levy Place, P.O. Box 1243, New York, NY, 10029, USA
| | - Joshua J Strohl
- Institute for Molecular Medicine, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Patricio T Huerta
- Institute for Molecular Medicine, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Weijia Zhang
- Department of Medicine, Mount Sinai Medical Center, One Gustave L. Levy Place, P.O. Box 1243, New York, NY, 10029, USA.
| | - Anne Davidson
- Institute for Molecular Medicine, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA.
| |
Collapse
|
18
|
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
|
19
|
Batla A, Simeoni S, Uchiyama T, deMin L, Baldwin J, Melbourne C, Islam S, Bhatia KP, Pakzad M, Eriksson S, Panicker JN. Exploratory pilot study of exogenous sustained-release melatonin on nocturia in Parkinson's disease. Eur J Neurol 2021; 28:1884-1892. [PMID: 33576095 DOI: 10.1111/ene.14774] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Nocturia is one of the commonest non-motor symptoms in Parkinson's disease (PD). Nocturia has evolved from being understood as a symptom of urological disorders or neurogenic bladder dysfunction to being considered as a form of circadian dysregulation. Exogenous melatonin is known to help circadian function and can be an effective strategy for nocturia in PD. METHODS In this open-label, single-site, exploratory, phase 2 pilot study, adults with PD and nocturia underwent assessments using standardized questionnaires, urodynamics studies and a bladder scan. This was followed by completion of a frequency volume chart (FVC) and 2-week sleep diary. Sustained-release melatonin 2 mg was then administered once-nightly for 6 weeks. A repeat assessment using questionnaires, the FVC and sleep diary was performed whilst on treatment with melatonin. Companion or bed partners filled in sleep questionnaires to assess their sleep during the intervention. RESULTS Twenty patients (12 males; mean age 68.2 [SD = 7.8] years; mean PD duration 8.0 [±5.5] years) with PD reporting nocturia were included. Administration of melatonin was associated with a significant reduction in the primary outcome bother related to nocturia measured using the International Consultation on Incontinence Questionnaire Nocturia (ICIQ-N) (p = 0.01), number of episodes of nocturia per night (p = 0.013) and average urine volume voided at night (p = 0.013). No serious adverse events were reported. No significant improvement was noted in bed partner sleep scores. CONCLUSIONS In this preliminary open-label study, administration of sustained-release melatonin 2 mg was found to be safe for clinical use and was associated with significant improvements in night-time frequency and nocturnal voided volumes in PD patients.
Collapse
Affiliation(s)
- Amit Batla
- Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, London, UK
| | - Sara Simeoni
- Department of Uro-Neurology, The National Hospital for Neurology and Neurosurgery, London, UK.,Department of Brain Repair and Rehabilitation, Faculty of Brain Sciences, University College London Queen Square Institute of Neurology, University College London, London, UK
| | - Tomoyuki Uchiyama
- Department of Uro-Neurology, The National Hospital for Neurology and Neurosurgery, London, UK
| | - Lorenzo deMin
- Department of Uro-Neurology, The National Hospital for Neurology and Neurosurgery, London, UK
| | - Joanne Baldwin
- Department of Uro-Neurology, The National Hospital for Neurology and Neurosurgery, London, UK
| | - Charles Melbourne
- Department of Uro-Neurology, The National Hospital for Neurology and Neurosurgery, London, UK
| | | | - Kailash P Bhatia
- Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, London, UK
| | - Mahreen Pakzad
- Department of Uro-Neurology, The National Hospital for Neurology and Neurosurgery, London, UK
| | | | - Jalesh N Panicker
- Department of Uro-Neurology, The National Hospital for Neurology and Neurosurgery, London, UK.,Department of Brain Repair and Rehabilitation, Faculty of Brain Sciences, University College London Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
20
|
Circadian rhythms of mineral metabolism in chronic kidney disease-mineral bone disorder. Curr Opin Nephrol Hypertens 2021; 29:367-377. [PMID: 32452917 DOI: 10.1097/mnh.0000000000000611] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW The circadian rhythms have a systemic impact on all aspects of physiology. Kidney diseases are associated with extremely high-cardiovascular mortality, related to chronic kidney disease-mineral bone disorder (CKD-MBD), involving bone, parathyroids and vascular calcification. Disruption of circadian rhythms may cause serious health problems, contributing to development of cardiovascular diseases, metabolic syndrome, cancer, organ fibrosis, osteopenia and aging. Evidence of disturbed circadian rhythms in CKD-MBD parameters and organs involved is emerging and will be discussed in this review. RECENT FINDINGS Kidney injury induces unstable behavioral circadian rhythm. Potentially, uremic toxins may affect the master-pacemaker of circadian rhythm in hypothalamus. In CKD disturbances in the circadian rhythms of CKD-MBD plasma-parameters, activin A, fibroblast growth factor 23, parathyroid hormone, phosphate have been demonstrated. A molecular circadian clock is also expressed in peripheral tissues, involved in CKD-MBD; vasculature, parathyroids and bone. Expression of the core circadian clock genes in the different tissues is disrupted in CKD-MBD. SUMMARY Disturbed circadian rhythms is a novel feature of CKD-MBD. There is a need to establish which specific input determines the phase of the local molecular clock and to characterize its regulation and deregulation in tissues involved in CKD-MBD. Finally, it is important to establish what are the implications for treatment including the potential applications for chronotherapy.
Collapse
|
21
|
Kurhaluk N, Tkachenko H, Lukash O. Melatonin modulates oxidative phosphorylation, hepatic and kidney autophagy-caused subclinical endotoxemia and acute ethanol-induced oxidative stress. Chronobiol Int 2020; 37:1709-1724. [DOI: 10.1080/07420528.2020.1830792] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Natalia Kurhaluk
- Department of Zoology and Animal Physiology, Institute of Biology and Earth Sciences, Pomeranian University in Słupsk, Słupsk, Poland
| | - Halyna Tkachenko
- Department of Zoology and Animal Physiology, Institute of Biology and Earth Sciences, Pomeranian University in Słupsk, Słupsk, Poland
| | - Oleksandr Lukash
- Department of Ecology and Nature Protection, T.G. Shevchenko National University “Chernihiv Collegium”, Chernihiv, Ukraine
| |
Collapse
|
22
|
Carriazo S, Ramos AM, Sanz AB, Sanchez-Niño MD, Kanbay M, Ortiz A. Chronodisruption: A Poorly Recognized Feature of CKD. Toxins (Basel) 2020; 12:E151. [PMID: 32121234 PMCID: PMC7150823 DOI: 10.3390/toxins12030151] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 02/17/2020] [Accepted: 02/20/2020] [Indexed: 02/07/2023] Open
Abstract
Multiple physiological variables change over time in a predictable and repetitive manner, guided by molecular clocks that respond to external and internal clues and are coordinated by a central clock. The kidney is the site of one of the most active peripheral clocks. Biological rhythms, of which the best known are circadian rhythms, are required for normal physiology of the kidneys and other organs. Chronodisruption refers to the chronic disruption of circadian rhythms leading to disease. While there is evidence that circadian rhythms may be altered in kidney disease and that altered circadian rhythms may accelerate chronic kidney disease (CKD) progression, there is no comprehensive review on chronodisruption and chronodisruptors in CKD and its manifestations. Indeed, the term chronodisruption has been rarely applied to CKD despite chronodisruptors being potential therapeutic targets in CKD patients. We now discuss evidence for chronodisruption in CKD and the impact of chronodisruption on CKD manifestations, identify potential chronodisruptors, some of them uremic toxins, and their therapeutic implications, and discuss current unanswered questions on this topic.
Collapse
Affiliation(s)
- Sol Carriazo
- IIS-Fundacion Jimenez Diaz, Department of Medicine, Universidad Autonoma de Madrid, Fundacion Renal Iñigo Alvarez de Toledo-IRSIN, 28040 Madrid, Spain; (S.C.); (A.MR.); (A.BS.); (M.D.S.-N.)
- Red de Investigación Renal (REDINREN), 28040 Madrid, Spain
| | - Adrián M Ramos
- IIS-Fundacion Jimenez Diaz, Department of Medicine, Universidad Autonoma de Madrid, Fundacion Renal Iñigo Alvarez de Toledo-IRSIN, 28040 Madrid, Spain; (S.C.); (A.MR.); (A.BS.); (M.D.S.-N.)
- Red de Investigación Renal (REDINREN), 28040 Madrid, Spain
| | - Ana B Sanz
- IIS-Fundacion Jimenez Diaz, Department of Medicine, Universidad Autonoma de Madrid, Fundacion Renal Iñigo Alvarez de Toledo-IRSIN, 28040 Madrid, Spain; (S.C.); (A.MR.); (A.BS.); (M.D.S.-N.)
- Red de Investigación Renal (REDINREN), 28040 Madrid, Spain
| | - Maria Dolores Sanchez-Niño
- IIS-Fundacion Jimenez Diaz, Department of Medicine, Universidad Autonoma de Madrid, Fundacion Renal Iñigo Alvarez de Toledo-IRSIN, 28040 Madrid, Spain; (S.C.); (A.MR.); (A.BS.); (M.D.S.-N.)
- Red de Investigación Renal (REDINREN), 28040 Madrid, Spain
| | - Mehmet Kanbay
- Division of Nephrology, Department of Medicine, Koc University School of Medicine, 34010 Istanbul, Turkey;
| | - Alberto Ortiz
- IIS-Fundacion Jimenez Diaz, Department of Medicine, Universidad Autonoma de Madrid, Fundacion Renal Iñigo Alvarez de Toledo-IRSIN, 28040 Madrid, Spain; (S.C.); (A.MR.); (A.BS.); (M.D.S.-N.)
- Red de Investigación Renal (REDINREN), 28040 Madrid, Spain
| |
Collapse
|
23
|
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
|
24
|
Analysis of the Effects of Day-Time vs. Night-Time Surgery on Renal Transplant Patient Outcomes. J Clin Med 2019; 8:jcm8071051. [PMID: 31323849 PMCID: PMC6678185 DOI: 10.3390/jcm8071051] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 07/11/2019] [Accepted: 07/12/2019] [Indexed: 02/01/2023] Open
Abstract
Sleep deprivation and disruption of the circadian rhythms could impair individual surgical performance and decision making. For this purpose, this study identified potential confounding factors on surgical renal transplant patient outcomes during day and night. Our retrospective cohort study of 215 adult renal cadaver transplant recipients, of which 132 recipients were allocated in the “day-time” group and 83 recipients in the “night-time” group, primarily stratified the patients into two cohorts, depending on the start time. Within a 24 h operational system, “day-time” was considered as being from 8 a.m. to 8 p.m. and “night-time” from 8 p.m. to 8 a.m.. Primary outcomes examined patient and graft survival after three months and one year. Secondary outcomes included the presence of acute rejection (AR) and delayed graft function (DGF), as well as the rate of postoperative complications. In log-rank testing, “day-time” surgery was associated with a significantly higher risk of patient death (p = 0.003), whereas long-term graft survival was unaffected by the operative time of day. The mean cold ischemia time (CIT), which was 12.4 ± 5.3 h in the “night-time” group, was significantly longer compared to 10.7 ± 3.6 for those during the day (p = 0.01). We observed that “night-time” kidney recipients experienced more wound complications. From our single-centre data, we conclude that night-time kidney transplantation does not increase the risk of adverse events or predispose the patient to a worse outcome. Nevertheless, further research is required to explore the effect of fatigue on nocturnal surgical performance.
Collapse
|