Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 May 1.
Published in final edited form as: J Heart Lung Transplant. 2024 Feb 2;43(5):849–857. doi: 10.1016/j.healun.2024.01.017

Circadian Rhythms in Solid Organ Transplantation

Brielle H Patlin 1, Huram Mok 1, Monaj Arra 1, Jeffrey A Haspel 1,*
PMCID: PMC11070314  NIHMSID: NIHMS1964099  PMID: 38310995

Abstract

Circadian rhythms are daily cycles in physiology that can affect medical interventions. This review considers how these rhythms may relate to solid organ transplantation. It begins by summarizing the mechanism for circadian rhythm generation known as the molecular clock, and basic research connecting the clock to biological activities germane to organ acceptance. Next follows a review of clinical evidence relating time of day to adverse transplantation outcomes. The concluding section discusses knowledge gaps and practical areas where applying circadian biology might improve transplantation success.

Keywords: Circadian rhythms, time of day, molecular clock


Circadian rhythms are daily oscillations in biological function that endow organisms with an intrinsic ability to keep time [13]. By enabling organisms to anticipate the day-night cycle regardless of weather conditions, circadian rhythms provide a powerful evolutionary advantage. In any given organism, circadian rhythms occur up and down the biological scale and manifest in clinically important functions like drug metabolism and immunity. There is a growing interest in the concept of “circadian medicine”, where therapies are tailored around patients’ circadian rhythms to improve their effectiveness [2, 4, 5].

This review considers the application of circadian biology to solid organ transplantation, which is little studied to date. It is intended as a primer for practitioners and transplant biologists, beginning with a description of the molecular clock mechanism and concluding with a review of the clinical literature evaluating the time of day as a factor in transplant success.

The molecular clock mechanism.

A biological rhythm is any predictably recurring variation in organism activity over time. Such rhythms are ubiquitous and occur across timescales ranging from milliseconds to decades [6, 7]. By definition, a biological rhythm is “circadian” if it repeats roughly every 24 hours without the need for environmental input to perpetuate the pattern [7, 8]. While not dependent on the environment, circadian rhythms must be responsive to exposures like light onset to align them with the environment, a process called entrainment [8]. The system that generates circadian rhythms is often conceptualized as an “oscillator” or clock that takes cyclical stimuli in the environment as input and imparts 24-hour patterns onto physiology as output (Fig. 1A).

Figure. 1: The circadian system in mammals.

Figure. 1:

Panel A depicts the “oscillator” paradigm of circadian rhythm generation, in which environmental cues set the phase of an intrinsic and continuously running clock. The clock then imparts a daily structure to biological function, from behavior to biomolecules. Panel B is a schematic of the circadian clock mechanism, composed of transcription factors called “clock genes” that regulate their expression in a feedback loop that takes 24 hours to reinitialize. Panel C depicts the two circadian clock compartments at issue during transplant. The recipient’s circadian system (black) consists of the central CNS pacemaker and peripheral clocks in recipient tissues. Clocks in the donor organ (red), may be set to a different circadian clock time (out of phase) than the recipient clocks at the time of transplantation and lack autonomic innervation that helps to synchronize peripheral clocks.

Recognition of circadian rhythms goes back millennia, but an understanding of how they come about only emerged in the 1990s with genetic experiments in model organisms combined with analyses of patients with heritable “circadian” sleep disorders [912]. Using mutant flies whose circadian pattern of activity was either disrupted or altered, it was possible to clone the first “clock” genes (Fig. 1B) [9, 10]. Studies revealed that clock genes are organized into a transcription factor network connected by a series of autoregulatory feedback loops [13]. In mammals, the core element is a heterodimer composed of the proteins BMAL1 and CLOCK that stimulate transcription from promoters and enhancer elements containing “E-box” motifs [14]. Among the hundreds of transactivated genes are the negative regulator genes PER1–3 and CRY1–2, whose protein products form a multimeric complex that moves to the nucleus and inhibits BMAL1/CLOCK. The stability of the PER/CRY complex is modulated by phosphorylation and ubiquitination, thereby speeding up or slowing down its feedback inhibition and allowing the “phase” of the clock to be adjusted [15]. A second group of clock genes called REVERBα/β (or NR1D1/2) are inhibitory transcription factors that form another negative feedback loop by repressing genes containing ROR-responsive elements (RREs), including BMAL1. Positive feedback is supplied by the clock genes RORαγ (RAR Related Orphan Receptor A–C) that stimulate transcription at RREs and directly compete with REVERBα/β. The cumulative result is that clock genes all oscillate in abundance in a roughly 24-hour cycle and— because they are transcription factors— impart a 24-hour bias to gene expression in general.

Virtually all nucleated cells express clock genes and have a functioning molecular clock. In mammals, roughly 50–85% of protein-coding mRNAs show rhythmic abundance organism-wide, with 5–10% exhibiting rhythms in any one organ [16, 17]. Among genes regulated by the clock are master transcription factors important for metabolic, immune, and redox functions, like nuclear factor IL-3–regulated (NFIL3), the PAR domain basic leucine zipper transcription factors (DBP, TEF, and HLF), the peroxisome proliferator–activated receptors (PPARs) [1820], and nuclear factor erythroid 2-related factor 2 (NRF2) [2123]. Beyond gene expression, other biomolecules oscillate as well, including metabolites [24], proteins [25, 26], and covalent modifications like phosphorylation and ubiquitination [2729]. It follows that cellular physiology has a strong circadian component, especially metabolism and cell division [30, 31].

Moving from the microscopic to the macroscopic, molecular clocks within neighboring cells are kept in register with one another through pulsatile extracellular cues like nutrient availability, corticosteroids, and autonomic tone [32]. The result is a general “internal synchrony” of billions of cell-autonomous circadian clocks within organisms, which allow rhythms in organ physiology to emerge. In mammals, neuro-humoral cues are under CNS control through a circadian “central pacemaker” located in the suprachiasmatic nucleus (SCN) of the ventral hypothalamus. The SCN receives direct light input from retinal ganglion cells and projects to hypothalamic areas, producing daily rhythms of pituitary hormone secretions, wakefulness, appetite, and temperature [32, 33]. Thus, circadian rhythms in organisms are the product of a two-tiered system, consisting of a “local” clock within each cell and rhythmic synchronizing cues broadcast by the CNS, which entrain clocks to the day-night cycle.

Understanding the role of the circadian system in transplantation can be especially challenging, as there are two circadian systems to consider: that of the recipient and that of the donated organ (Fig. 1C). Of the many circadian outputs in physiology, three stand out as particularly germane to the immediate transplantation setting. First, immune system activity is highly rhythmic across the day at virtually every level (Table 1). For example, inflammatory cytokine release from macrophages in response to endotoxin or other microbial products varies markedly based on the time of day. Leukocyte trafficking into organs exhibits a circadian rhythm, hence the WBC burden of transplanted organs could theoretically vary with the time of procurement. On the recipient side, the potential for leukocyte infiltration into a new allograft might vary by the phase in the circadian cycle when transplantation occurs. Finally, T cell activation has a circadian rhythm, and hence the time of transplantation may figure into graft tolerance.

Table 1:

Circadian Clock Regulation of Basic Immune Processes.

Immune Activity Cell Types Clock-Dependent Mechanisms References
Leukocyte Development Innate Lymphoid Cells
  • Regulated by clock genes NFIL3, RORa

[8995]
TH17 cells
  • Regulated by clock genes RORa, RORgt, NR1D1

[9698]
Treg cells
  • Accessory clock gene NFIL3 regulates FOXP3

[99]
Neutrophils
  • BMAL regulates CXCL2

[5254]
Leukocyte Trafficking All
  • Rhythms in cell adhesion molecule expression on leukocytes and endothelial cells.

[57]
  • Rhythms in leukocyte chemotactic factors and receptors.

[56, 57, 100104]
Innate Immunity Macrophages
  • Rhythms in secretion of numerous cytokines and chemokines.

[4347, 105107]
  • BMAL1 regulates PD-L1promoting immune senescence.

[42, 48]
  • Rhythms in phagocytosis associated with oscillations in mitochondrial dynamics.

[108111]
NK cells
  • Rhythms in IFNg, Perforin, and granzyme expression.

[112, 113]
Neutrophils
  • Rhythms in NETosis, NADPH oxidase, and phagocytosis.

[53, 55]
Mast cells/Eosinophils
  • Rhythms in IgE and fMLP stimulated chemokine secretion.

[114]
Adaptive Immunity Dendritic cells
  • Rhythms in activation via TLR ligands.

[69, 70]
  • Rhythms in antigen processing which correlate with mitochondrial dynamics.

[115]
  • Rhythms in CD80 costimulatory molecule expression.

[116]
T cells
  • Rhythms in CD8 cell expansion upon antigen challenge.

[117, 118]
B cells
  • Rhythms in antibody titers based on immunization time of day. Correlates with times of higher lymph node cellularity.

[119]
  • Rhythm in IgA secretion from intestinal plasma cells.

[120]
  • Regulation of BCR signaling by CRY1/2. Deletion of these genes produces spontaneous antibodymediated autoimmune disease in mice.

[121]

Secondly, there are circadian rhythms in the response to tissue ischemia. In animal surgical models, the extent of ischemia-reperfusion injury in the heart and kidney varies with time of day in a circadian manner [3438]. This may reflect circadian rhythms in antioxidant capacity, including the direct transcriptional regulation of the master regular NRF2 by the circadian clock [2123]. Additionally, induction of hypoxia-inducible factor 1α (HIF1a) by low oxygen conditions and exercise is enhanced by the clock protein BMAL1 [39, 40]. Neutrophils and macrophages which are key in ischemia reperfusion injury, are under strong circadian regulation. Macrophages exhibit circadian patterns of phagocytosis, chemokine and secretion, as well as cell adhesion molecule expression and trafficking [4148]. This is important because changes in macrophage activation, abundance, and function play a part in determining the severity of the ischemic injury [4951]. Similarly, neutrophils exacerbate post-ischemic inflammation and demonstrate circadian regulation in trafficking patterns, cytokine secretion, NETosis, and phagocytosis [5258].

Finally, there are circadian rhythms in drug metabolism [16, 59]. Multiple P450 enzymes are rhythmically expressed, including CYP3A4 and 2C8 that contribute to the metabolism of immunosuppressive agents like tacrolimus, sirolimus, prednisolone, and mycophenolate motefil [60]. For drugs like prednisone and tacrolimus whose half-lives are shorter than a circadian cycle (about 12 hours in each case), the time of dosing can affect their pharmacodynamics [6163]. Evidence for clinical relevance exists for asthmatic patients, where the time of inhaled steroid dosing alters the minimum clinically effective dose [64].

Evidence that transplantation time of day matters.

In contrast to basic research studies, the clinical literature assessing circadian rhythms in solid organ transplantation is sparse. There are 16 retrospective studies in PUBMED analyzing associations between transplantation surgery time of day and outcomes (Table 2), and 6 examining the time of organ procurement (Table 3). As a group, the studies differ in terms of the organs considered, sample size, outcomes measured, and the approach to binning times of day. Most studies dichotomously compare “day’ versus “night” transplantation, which has limited sensitivity. Nevertheless, most studies conclude that night-time or early morning transplantation correlates with moderately worse outcomes. These include acute complications like graft dysfunction and long-term graft survival. The evidence is strongest for lung transplantation (4/4 studies), which is biologically plausible given the heavy immune composition of this organ: about 25% of cells in the lung are of hematopoietic origin and the density of neutrophils in the lung is 3 times higher than peripheral blood [65, 66].

Table 2: Association between transplantation time of day and outcomes.

Times of day are expressed in military time.

Organ Transplantation Time Bins Compared Sample Size Duration of Followup Outcomes Time Effect (Highest Risk Bin for Adverse Outcomes) Reference
Lung 7:00–18:59
19:00–6:59
10.545 1 Year
  • Postoperative Events

  • Airway Dehiscence

  • Patient Survival


+ (19:00–6:59).
+ 90-day survival
(19:00–6:59)
[122]
Lung 0:00–3:59
4:00–7:59
8:00–11:59
12:00–15:59
16:00–19:59
12:00–23:59
563 3 Days
  • Primary Graft Dysfunction

+ (4:00–7:59) [80]
Lung 5:00–17:59
18:00–4:59
740 5 Years
  • Postoperative Events

  • Overall Survival

  • Chronic Rejection-Free Survival

+ (18:00–4:59)
+ (18:00–4:59)
+ (18:00–4:59)
[123]
Lung 0:00–5:59
6:00–11:59
12:00–17:59
18:00–23:59
32,841 10 years
  • Length of Stay

  • Overall Survival

  • Primary Graft Dysfunction

+ (18:00–0:00)
+ (18:00–0:00)
+ (18:00–0:00)
[124]
Heart 4:00–1159
12:00–19:59
20:00–3:59
235 3 Years
  • Delayed Graft Function

  • Postoperative Events

  • Infection

  • Acute Rejection

  • Patient Survival





− (trend towards worse survival with morning to afternoon)
[125]
Heart 7:00–18:59
19:00–6:59
16,573 1 Year
  • Post-operative Events

  • Patient Survival


[122]
Liver 8:00–14:00
20:00–2:00
147 Not Specified
  • Intraoperative Events

  • Delayed Graft Function

+
+ (20:00–2:00)
[126]
Liver 7:00–18:59
19:00–6:59
8816 3 Years
  • Graft Survival

+/− (19:00–6:59 in unadjusted analysis) [127]
Liver 8:00–17:59
18:00–7:59
350 1 Year
  • Patient Survival

  • Graft Survival

[128]
Kidney 0:00–5:59
6:00–11:59
12:00–17:59
18:00–23:59
726 7 Days
  • Delayed Graft Function

+ (6:00–11:59)
Note: association noted only with organs obtained from brain-dead donors (n=536).
[84]
Kidney 10:00–17:59
18:00–9:59
8962 3 Years
  • Patient Survival

+ (18:00–9:59) [129]
Kidney 8:00–19:59
20:00–7:59
215 1 Year
  • Graft Survival

  • Patient Survival

[130]
Kidney 8:00–19:59
20:00–7:59
260 43 Months (Median)
  • Graft Survival

  • Re-transplantation

+ (20:00–7:59)
+ (20:00–7:59)
[131]
Kidney 8:00–19:59
20:00–7:59
443 1 Year
  • Graft Survival

  • Patient Survival

[132]
Kidney 8:00–19:59
20:00–7:59
7 1 Year
  • Delayed Graft Function

  • Serum Creatinine

[133]
Kidney 8:00–19:59
20:00–7:59
4519 10 Years
  • Graft Survival

+(20:00–7:59) [134]
Kidney 8:00–19:59
20:00–7:59
873 5 Years
  • Delayed Graft Function

  • Acute Rejection

  • Graft Survival

+(20:00–7:59)

[135]

Table 3: Association between organ procurement time of day and transplant outcomes.

Times of day are expressed in military time.

Organ Procurement Time Bins Compared Sample Size Duration of Followup Outcomes Time Effect (Highest Risk Bin for Adverse Outcomes) Reference
Heart 12:00–23:59
0:00–11:59
266 (propensity score matched) 6 Years
  • Acute Rejection-Free Survival

  • Chronic Rejection-Free Survival

  • Overall Survival

+ (12:00–23:59)

+ (12:00–23:59)
[136]
Liver 7:00–18:59
19:00–6:59
8816 3 Years
  • Graft Survival

+/− (19:00–6:59 in unadjusted analysis) [127]
Kidney 0:00–5:59
6:00–11:59
12:00–17:59
18:00–23:59
726 7 Days
  • Delayed Graft Function

+ (6:00–11:59)
Note: association noted only with organs obtained from brain-dead donors (n=536).
[84]
Kidney 0:00–5:59
6:00–11:59
12:00–17:59
18:00–23:59
536 7 Days
  • Delayed Graft Function

[77]
Kidney 10:00–17:59
18:00–9:59
8962 3 Years
  • Patient Survival

+ (18:00–9:59) [129]
Kidney 12:00–23:59
0:00–11:59
5026 10 Years
  • Delayed Graft Function

  • eGFR at 1 Year

  • Patient and Graft Survival

+ (12:00–23:59)

+ (12:00–23:59)
[137]

Immunologically focused transplant studies rarely consider time-of-day as a biological variable. However, studies repeatedly demonstrate that there is circadian regulation of leukocyte trafficking, cytokine release, and immune cell surface marker upregulation [6771]. These fluctuations may result in differential immune measurements at different times of day, similar to how transplant patients tend to develop arrhythmia at night [7275]. Therefore, studies which consider differences in these immune markers may generate spurious results. For example, if a study is performed in mice, and the controls are measured in the morning and the transplant mice are measured in the afternoon, differences in the respective immune landscapes will be significant regardless of treatment. Conversely, replicate experiments performed at different times of day may yield variable results that can be dismissed as falsely negative. Beyond the direct time-of-day effect on the immune landscape, many metabolic systems are dependent on circadian regulated protein and expression levels which could affect drug metabolism and response [4, 5, 62].

Intriguingly, some studies indicate that organ procurement time of day independently correlates with transplant success, implying that circadian rhythms within the donor or donor organ may affect graft performance (Table 3). At first glance, this seems counterintuitive as most solid organ donors are brain-dead and presumably lack a functioning central circadian pacemaker. However, no literature assessing biological rhythms or circadian clock function in the organ donor population is currently available. The nearest correlate would be intensive care unit (ICU) patients, who routinely exhibit intact rhythms in core body temperature and even clock gene expression [76, 77]. A major distinction between ICU patients and healthy individuals is that circadian outputs like temperature rhythms appear unmoored from the day-night cycle and peak at different points of the day from patient to patient [76]. Whether this extends to organ donors is unknown. Of note, organ donors routinely receive supra-physiologic doses of corticosteroids as part of pre-procurement management [78]. Given that corticosteroids are a strong circadian clock synchronizer [79], it’s tempting to speculate this could enable donors to recapitulate physiologic circadian rhythms in the absence of a central pacemaker.

Altogether, clinical observations suggest a time-of-day effect in solid organ transplantation, both respect to the donor organs and the recipient. As with all real-world biological rhythms, the effects observed are likely an amalgam of environmental variations (like differences in hospital staffing or performance across the day) and periodicity in the underlying biology (circadian clocks). Regardless, there is an opportunity to improve transplantation success by better understanding time as a biological variable in organ donors and recipients.

Future Directions.

Clinicians already consider donor-recipient matching based on organ size, infectious history, blood group, and, in the case of hematopoietic stem cells, HLA typing. Circadian biology raises the possibility that temporally matching the circadian systems of donated organs and recipients may improve transplant acceptance. Getting to this point will require a much better mechanistic understanding of how circadian clocks relate to organ transplant biology. It will also require translational research advances to practically apply circadian biology to transplantation process of care. Several topics need parallel development:

Mechanistic research.

While clinical observations are suggestive, there are currently no direct mechanistic studies evaluating how the circadian clock affects transplant pathophysiology like primary graft dysfunction, acute rejection, or chronic rejection. However, multiple tools and model organisms can be applied to this question including clock gene mutant mice, many of which are commercially available. A useful quality of circadian clocks is that they can be experimentally manipulated by adjusting light or feeding cycles, thereby shifting organisms into different “time zones” before employing them as organ donors or recipients. The internal circadian synchrony of model organisms can be disrupted by providing a variable light schedule to create a state of “chronic jet lag”, which can serve to validate observations with clock mutant mice. The existing clinical literature and the prominent immune composition of the lung illustrates the need to explore circadian rhythm contributions to thoracic transplants and subsequent recovery.

Donor circadian biology.

Given the suggestion that the time of organ procurement may be clinically relevant, there may be an opportunity to employ donor circadian biology to affect outcomes. The extent to which biological rhythms persist in organ donors (brain dead or otherwise) is unknown. However, donors routinely receive corticosteroids, which have the potential to synchronize or reboot circadian rhythms in the donor. The pre-procurement period is one of the most practical points in the transplantation process to manipulate circadian rhythms in immune cell trafficking or ischemia tolerance given the degree of clinical control. Circadian clocks have a stereotypical reaction to synchronizing agents like steroids called a “phase response curve”, in which the clocks will either advance, fall back, or do nothing based on the internal clock time when stimulus occurs. To be able to predict how corticosteroid dosing affects circadian clocks in donors, it will be necessary to develop a means of estimating the internal clock time of individual donors. Assays already exist for predicting clock time using peripheral blood or skin samples from healthy individuals, and similar methodologies could be adapted to the donor population.

Organ preservation.

Evidence suggests that hypothermia as mild as 18°C synchronizes circadian clocks, making organ preservation conditions a factor in circadian biology within the allograft [80, 81]. With the increasing popularity of continuous perfusion systems, there is the potential to expose grafts to temperature or hormonal cycles during the period between organ procurement and transplantation. In so doing, it may be possible to shift circadian clocks so that their phases are aligned with the intended recipient. Research is needed to determine whether such an approach can optimize graft resilience against ischemia/reperfusion injury.

Circadian pharmacology.

Despite studies suggesting that time of day affects the pharmacodynamics of immunosuppressants, tailoring the dosing of these drugs around patient circadian rhythms to maximize their therapeutic index is not a part of routine practice. Likely, part of the reason is that the standard practice of tacrolimus drug level monitoring allows clinicians to empirically determine dosing, and there is the potential for increasing non-compliance by making dosing schedules too arduous or complicated for patients. One opportunity may be the dosing of prednisone, a once-a-day drug where night dosing may enable lower amounts to be given to patients based on asthma literature. Additionally, pulse steroid dosing for clinical deterioration may provide differential benefits based on morning versus evening dosing [64]. More research is warranted to investigate this.

Rhythms as a marker of transplant recipient health.

Finally, for transplant recipients to thrive socially they must regain circadian rhythms in activity, sleep, and food intake. Literature suggests that circadian rhythms connected to donor organ physiology, for example, heart rate variation and blood pressure, are lost initially after solid organ transplantation [8285]. Re-establishment of these rhythms could potentially represent a positive long-term prognostic sign [86]. Conversely, a loss of circadian rhythms in activity or sleep could presage clinical decompensation in transplant recipients, as this is already understood to be a general predictor of future hospitalization [87, 88]. Over-the-counter devices including cell phones can collect actigraphy data and represent a possible way to improve clinical follow-up, especially for recipients living far away from specialized transplant centers.

In summary, circadian rhythms and their underlying clock are likely to be relevant to solid organ transplantation on multiple biological and clinical levels. The topic is wide open for study, and future research will determine how circadian biology can be best leveraged to improve transplant success.

ACKNOWLEDGEMENTS

We thank Carrie Gierasch, Kim Harbert, and Robyn Haspel for their input. This work was funded by NIH R01 HL135846 and R01 HL152968.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflict of interest statement:

The authors have declared that no conflict of interest exists.

REFERENCES

  • 1.Nosal C, Ehlers A, and Haspel JA, Why Lungs Keep Time: Circadian Rhythms and Lung Immunity. Annu Rev Physiol, 2020. 82: p. 391–412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Allada R and Bass J, Circadian Mechanisms in Medicine. N Engl J Med, 2021. 384(6): p. 550–561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Cederroth CR, et al. , Medicine in the Fourth Dimension. Cell Metab, 2019. 30(2): p. 238–250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Cederroth CR, et al. , Medicine in the Fourth Dimension. Cell Metab, 2019. 30(2): p. 238–250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Panda S, The arrival of circadian medicine. Nat Rev Endocrinol, 2019. 15(2): p. 67–69. [DOI] [PubMed] [Google Scholar]
  • 6.Refinetti R, Integration of biological clocks and rhythms. Compr Physiol, 2012. 2(2): p. 1213–39. [DOI] [PubMed] [Google Scholar]
  • 7.Kuhlman SJ, Mackey SR, and Duffy JF, Biological Rhythms Workshop I: introduction to chronobiology. Cold Spring Harb Symp Quant Biol, 2007. 72: p. 1–6. [DOI] [PubMed] [Google Scholar]
  • 8.Klerman EB, et al. , Keeping an eye on circadian time in clinical research and medicine. Clin Transl Med, 2022. 12(12): p. e1131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Rosbash M, Circadian Rhythms and the Transcriptional Feedback Loop (Nobel Lecture)*. Angew Chem Int Ed Engl, 2021. 60(16): p. 8650–8666. [DOI] [PubMed] [Google Scholar]
  • 10.Huang RC, The discoveries of molecular mechanisms for the circadian rhythm: The 2017 Nobel Prize in Physiology or Medicine. Biomed J, 2018. 41(1): p. 5–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Patke A, et al. , Mutation of the Human Circadian Clock Gene CRY1 in Familial Delayed Sleep Phase Disorder. Cell, 2017. 169(2): p. 203–215 e13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Meyer N, et al. , Circadian rhythms and disorders of the timing of sleep. Lancet, 2022. 400(10357): p. 1061–1078. [DOI] [PubMed] [Google Scholar]
  • 13.Mackey SR, Biological Rhythms Workshop IA: molecular basis of rhythms generation. Cold Spring Harb Symp Quant Biol, 2007. 72: p. 7–19. [DOI] [PubMed] [Google Scholar]
  • 14.Trott AJ and Menet JS, Regulation of circadian clock transcriptional output by CLOCK:BMAL1. PLoS Genet, 2018. 14(1): p. e1007156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Virshup DM, et al. , Reversible protein phosphorylation regulates circadian rhythms. Cold Spring Harb Symp Quant Biol, 2007. 72: p. 413–20. [DOI] [PubMed] [Google Scholar]
  • 16.Zhang R, et al. , A circadian gene expression atlas in mammals: implications for biology and medicine. Proc Natl Acad Sci U S A, 2014. 111(45): p. 16219–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Mure LS, et al. , Diurnal transcriptome atlas of a primate across major neural and peripheral tissues. Science, 2018. 359(6381). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Cowell IG, E4BP4/NFIL3, a PAR-related bZIP factor with many roles. Bioessays, 2002. 24(11): p. 1023–9. [DOI] [PubMed] [Google Scholar]
  • 19.Gachon F, et al. , The circadian PAR-domain basic leucine zipper transcription factors DBP, TEF, and HLF modulate basal and inducible xenobiotic detoxification. Cell Metab, 2006. 4(1): p. 25–36. [DOI] [PubMed] [Google Scholar]
  • 20.Chen L and Yang G, PPARs Integrate the Mammalian Clock and Energy Metabolism. PPAR Res, 2014. 2014: p. 653017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Pekovic-Vaughan V, et al. , The circadian clock regulates rhythmic activation of the NRF2/glutathione-mediated antioxidant defense pathway to modulate pulmonary fibrosis. Genes Dev, 2014. 28(6): p. 548–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Sun Q, et al. , Mechanism of circadian regulation of the NRF2/ARE pathway in renal ischemia-reperfusion. Exp Ther Med, 2021. 21(3): p. 190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Early JO, et al. , Circadian clock protein BMAL1 regulates IL-1β in macrophages via NRF2. Proc Natl Acad Sci U S A, 2018. 115(36): p. E8460–e8468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Woelders T, et al. , Machine learning estimation of human body time using metabolomic profiling. Proc Natl Acad Sci U S A, 2023. 120(18): p. e2212685120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Wang Y, et al. , A proteomics landscape of circadian clock in mouse liver. Nature Communications, 2018. 9(1): p. 1553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Specht A, et al. , Circadian protein expression patterns in healthy young adults. Sleep Health, 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Robles MS, Humphrey SJ, and Mann M, Phosphorylation Is a Central Mechanism for Circadian Control of Metabolism and Physiology. Cell Metab, 2017. 25(1): p. 118–127. [DOI] [PubMed] [Google Scholar]
  • 28.Ryzhikov M, et al. , Diurnal Rhythms Spatially and Temporally Organize Autophagy. Cell Rep, 2019. 26(7): p. 1880–1892.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Hansen FM, et al. , Data-independent acquisition method for ubiquitinome analysis reveals regulation of circadian biology. Nature Communications, 2021. 12(1): p. 254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Green CB, Takahashi JS, and Bass J, The meter of metabolism. Cell, 2008. 134(5): p. 728–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Farshadi E, van der Horst GTJ, and Chaves I, Molecular Links between the Circadian Clock and the Cell Cycle. J Mol Biol, 2020. 432(12): p. 3515–3524. [DOI] [PubMed] [Google Scholar]
  • 32.Schibler U, et al. , Clock-Talk: Interactions between Central and Peripheral Circadian Oscillators in Mammals. Cold Spring Harb Symp Quant Biol, 2015. 80: p. 223–32. [DOI] [PubMed] [Google Scholar]
  • 33.Li JD, Hu WP, and Zhou QY, The circadian output signals from the suprachiasmatic nuclei. Prog Brain Res, 2012. 199: p. 119–127. [DOI] [PubMed] [Google Scholar]
  • 34.Qiu Z, et al. , Roles of HDAC3-orchestrated circadian clock gene oscillations in diabetic rats following myocardial ischaemia/reperfusion injury. Cell Death & Disease, 2021. 12(1): p. 43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Rabinovich-Nikitin I and Kirshenbaum LA, Circadian regulated control of myocardial ischemia-reperfusion injury. Trends in Cardiovascular Medicine, 2022. [DOI] [PubMed] [Google Scholar]
  • 36.Montaigne D, et al. , Daytime variation of perioperative myocardial injury in cardiac surgery and its prevention by Rev-Erbα antagonism: a single-centre propensity-matched cohort study and a randomised study. Lancet, 2018. 391(10115): p. 59–69. [DOI] [PubMed] [Google Scholar]
  • 37.Correa-Costa M, et al. , Carbon monoxide protects the kidney through the central circadian clock and CD39. Proceedings of the National Academy of Sciences of the United States of America, 2018. 115(10): p. E2302–E2310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Bulluck H, et al. , Circadian variation in acute myocardial infarct size assessed by cardiovascular magnetic resonance in reperfused STEMI patients. Int J Cardiol, 2017. 230: p. 149–154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Wu Y, et al. , Reciprocal Regulation between the Circadian Clock and Hypoxia Signaling at the Genome Level in Mammals. Cell Metab, 2017. 25(1): p. 73–85. [DOI] [PubMed] [Google Scholar]
  • 40.Peek CB, et al. , Circadian Clock Interaction with HIF1α Mediates Oxygenic Metabolism and Anaerobic Glycolysis in Skeletal Muscle. Cell Metab, 2017. 25(1): p. 86–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.!!! INVALID CITATION !!! [41–52].
  • 42.Deng W, et al. , The Circadian Clock Controls Immune Checkpoint Pathway in Sepsis. Cell Rep, 2018. 24(2): p. 366–378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Gibbs JE, et al. , The nuclear receptor REV-ERBα mediates circadian regulation of innate immunity through selective regulation of inflammatory cytokines. Proc Natl Acad Sci U S A, 2012. 109(2): p. 582–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Griffin P, et al. , Circadian clock protein Rev-erbα regulates neuroinflammation. Proceedings of the National Academy of Sciences, 2019. 116(11): p. 5102–5107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Narasimamurthy R, et al. , Circadian clock protein cryptochrome regulates the expression of proinflammatory cytokines. Proc Natl Acad Sci U S A, 2012. 109(31): p. 12662–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Pourcet B, et al. , Nuclear Receptor Subfamily 1 Group D Member 1 Regulates Circadian Activity of NLRP3 Inflammasome to Reduce the Severity of Fulminant Hepatitis in Mice. Gastroenterology, 2018. 154(5): p. 1449–1464.e20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Sato S, et al. , A circadian clock gene, Rev-erbα, modulates the inflammatory function of macrophages through the negative regulation of Ccl2 expression. J Immunol, 2014. 192(1): p. 407–17. [DOI] [PubMed] [Google Scholar]
  • 48.Tsuruta A, et al. , Diurnal Expression of PD-1 on Tumor-Associated Macrophages Underlies the Dosing Time-Dependent Antitumor Effects of the PD-1/PD-L1 Inhibitor BMS-1 in B16/BL6 Melanoma-Bearing Mice. Molecular Cancer Research, 2022. 20(6): p. 972–982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Shi-chun Shen JX, Cheng Cheng, Xiang Xin-jian, Hong Bao-yu, Zhang Meng, Gong Chen & Ma Li-kun, Macrophages promote the transition from myocardial ischemia reperfusion injury to cardiac fibrosis in mice through GMCSF/CCL2/CCR2 and phenotype switching. Acta Pharmacol Sin, 2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Wang H, et al. , Macrophage Polarization and Liver Ischemia-Reperfusion Injury. Int J Med Sci, 2021. 18(5): p. 1104–1113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Huen SC and Cantley LG, Macrophage-mediated injury and repair after ischemic kidney injury. Pediatr Nephrol, 2015. 30(2): p. 199–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Adrover JM, et al. , A Neutrophil Timer Coordinates Immune Defense and Vascular Protection. Immunity, 2019. 50(2): p. 390–402.e10. [DOI] [PubMed] [Google Scholar]
  • 53.Adrover JM, et al. , Programmed ‘disarming’ of the neutrophil proteome reduces the magnitude of inflammation. Nat Immunol, 2020. 21(2): p. 135–144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Aroca-Crevillén A, Adrover JM, and Hidalgo A, Circadian Features of Neutrophil Biology. Front Immunol, 2020. 11: p. 576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Ella K, Csépányi-Kömi R, and Káldi K, Circadian regulation of human peripheral neutrophils. Brain, Behavior, and Immunity, 2016. 57: p. 209–221. [DOI] [PubMed] [Google Scholar]
  • 56.Druzd D, et al. , Lymphocyte Circadian Clocks Control Lymph Node Trafficking and Adaptive Immune Responses. Immunity, 2017. 46(1): p. 120–132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.He W, et al. , Circadian Expression of Migratory Factors Establishes Lineage-Specific Signatures that Guide the Homing of Leukocyte Subsets to Tissues. Immunity, 2018. 49(6): p. 1175–1190 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Holtkamp SJ, et al. , Circadian clocks guide dendritic cells into skin lymphatics. Nat Immunol, 2021. 22(11): p. 1375–1381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Ruben MD, et al. , Dosing time matters. Science, 2019. 365(6453): p. 547–549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Chen M, et al. , Identification of rhythmic human CYPs and their circadian regulators using synchronized hepatoma cells. Xenobiotica, 2020. 50(9): p. 1052–1063. [DOI] [PubMed] [Google Scholar]
  • 61.Fontova P, et al. , Influence of the Circadian Timing System on Tacrolimus Pharmacokinetics and Pharmacodynamics After Kidney Transplantation. Front Pharmacol, 2021. 12: p. 636048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Park SI, et al. , Circadian and time-dependent variability in tacrolimus pharmacokinetics. Fundam Clin Pharmacol, 2007. 21(2): p. 191–7. [DOI] [PubMed] [Google Scholar]
  • 63.Wintzinger M, et al. , Impact of circadian time of dosing on cardiomyocyte-autonomous effects of glucocorticoids. Mol Metab, 2022. 62: p. 101528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Noonan M, et al. , Comparison of once-daily to twice-daily treatment with mometasone furoate dry powder inhaler. Annals of Allergy, Asthma & Immunology, 2001. 86(1): p. 36–43. [DOI] [PubMed] [Google Scholar]
  • 65.Ermert L, Seeger W, and Duncker HR, Computer-assisted morphometry of the intracapillary leukocyte pool in the rabbit lung. Cell Tissue Res, 1993. 271(3): p. 469–76. [DOI] [PubMed] [Google Scholar]
  • 66.Aulakh GK, Neutrophils in the lung: “the first responders”. Cell Tissue Res, 2018. 371(3): p. 577–588. [DOI] [PubMed] [Google Scholar]
  • 67.Man K, Loudon A, and Chawla A, Immunity around the clock. Science, 2016. 354(6315): p. 999–1003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Orozco-Solis R and Aguilar-Arnal L, Circadian Regulation of Immunity Through Epigenetic Mechanisms. Front Cell Infect Microbiol, 2020. 10: p. 96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Silver AC, et al. , The circadian clock controls toll-like receptor 9-mediated innate and adaptive immunity. Immunity, 2012. 36(2): p. 251–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Silver AC, et al. , Daily oscillations in expression and responsiveness of Toll-like receptors in splenic immune cells. Heliyon, 2018. 4(3): p. e00579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Wang C, et al. , The circadian immune system. Sci Immunol, 2022. 7(72): p. eabm2465. [DOI] [PubMed] [Google Scholar]
  • 72.Bansal N, et al. , Masked Hypertension in Pediatric Heart Transplant Recipients. Pediatr Cardiol, 2023. 44(5): p. 1003–1008. [DOI] [PubMed] [Google Scholar]
  • 73.Bracht C, et al. , Circadian variations of blood pressure and heart rate early and late after heart transplantation. Transplantation, 1996. 62(8): p. 1187–90. [DOI] [PubMed] [Google Scholar]
  • 74.Cashion AK, et al. , Changes in patterns of 24-hr heart rate variability after kidney and kidney-pancreas transplant. Transplantation, 1999. 68(12): p. 1846–50. [DOI] [PubMed] [Google Scholar]
  • 75.Parra DA, et al. , Endothelial dysfunction and circadian blood pressure rhythmicity in young heart transplant recipients. Pediatr Cardiol, 2007. 28(1): p. 1–7. [DOI] [PubMed] [Google Scholar]
  • 76.Knauert MP, Haspel JA, and Pisani MA, Sleep Loss and Circadian Rhythm Disruption in the Intensive Care Unit. Clin Chest Med, 2015. 36(3): p. 419–29. [DOI] [PubMed] [Google Scholar]
  • 77.Cunningham PS, et al. , ClinCirc identifies alterations of the circadian peripheral oscillator in critical care patients. J Clin Invest, 2023. 133(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.D’Aragon F, et al. , Effect of corticosteroid administration on neurologically deceased organ donors and transplant recipients: a systematic review and meta-analysis. BMJ Open, 2017. 7(6): p. e014436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Izumo M, et al. , Quantitative analyses of circadian gene expression in mammalian cell cultures. PLoS Comput Biol, 2006. 2(10): p. e136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Cunningham PS, et al. , Incidence of primary graft dysfunction after lung transplantation is altered by timing of allograft implantation. Thorax, 2019. 74(4): p. 413–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Fischl H, et al. , Cold-induced chromatin compaction and nuclear retention of clock mRNAs resets the circadian rhythm. The EMBO Journal, 2020. 39(22): p. e105604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Pisano A, et al. , Assessment of hypertension in kidney transplantation by ambulatory blood pressure monitoring: a systematic review and meta-analysis. Clinical Kidney Journal, 2021. 15(1): p. 31–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Lanuza DM, et al. , Circadian rhythm changes in blood pressure and heart rate during the first year after heart transplantation. J Heart Lung Transplant, 1994. 13(4): p. 614–23. [PubMed] [Google Scholar]
  • 84.van de Borne P, et al. , Reappearance of a normal circadian rhythm of blood pressure after cardiac transplantation. The American Journal of Cardiology, 1992. 69(8): p. 794–801. [DOI] [PubMed] [Google Scholar]
  • 85.Hryniewiecka E, et al. , Circadian and short-term blood pressure abnormalities after liver transplantation. Clin Exp Hypertens, 2018. 40(8): p. 730–733. [DOI] [PubMed] [Google Scholar]
  • 86.Jaques DA, et al. , Relationship between renal function and blood pressure dipping status in renal transplant recipients: a longitudinal study. BMC Nephrology, 2021. 22(1): p. 325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Brooks TG, et al. , Diurnal rhythms of wrist temperature are associated with future disease risk in the UK Biobank. Nature Communications, 2023. 14(1): p. 5172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Naik A, et al. , Circadian regulation of lung repair and regeneration. JCI Insight, 2023. 8(16). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Kostrzewski T, et al. , Multiple Levels of Control Determine How E4bp4/Nfil3 Regulates NK Cell Development. J Immunol, 2018. 200(4): p. 1370–1381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Geiger TL, et al. , Nfil3 is crucial for development of innate lymphoid cells and host protection against intestinal pathogens. J Exp Med, 2014. 211(9): p. 1723–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Seillet C, et al. , Nfil3 is required for the development of all innate lymphoid cell subsets. J Exp Med, 2014. 211(9): p. 1733–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Male V, et al. , The transcription factor E4bp4/Nfil3 controls commitment to the NK lineage and directly regulates Eomes and Id2 expression. J Exp Med, 2014. 211(4): p. 635–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Kamizono S, et al. , Nfil3/E4bp4 is required for the development and maturation of NK cells in vivo. J Exp Med, 2009. 206(13): p. 2977–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Yu X, et al. , The basic leucine zipper transcription factor NFIL3 directs the development of a common innate lymphoid cell precursor. Elife, 2014. 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Wong SH, et al. , Transcription factor RORα is critical for nuocyte development. Nat Immunol, 2012. 13(3): p. 229–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Yu X, et al. , TH17 cell differentiation is regulated by the circadian clock. Science, 2013. 342(6159): p. 727–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Amir M, et al. , REV-ERBalpha Regulates TH17 Cell Development and Autoimmunity. Cell Rep, 2018. 25(13): p. 3733–3749 e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Chang C, et al. , The nuclear receptor REV-ERBα modulates Th17 cell-mediated autoimmune disease. Proc Natl Acad Sci U S A, 2019. 116(37): p. 18528–18536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Kim HS, et al. , The transcription factor NFIL3 controls regulatory T-cell function and stability. Exp Mol Med, 2019. 51(7): p. 1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Nguyen KD, et al. , Circadian gene Bmal1 regulates diurnal oscillations of Ly6C(hi) inflammatory monocytes. Science, 2013. 341(6153): p. 1483–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Druzd D, et al. , Lymphocyte Circadian Clocks Control Lymph Node Trafficking and Adaptive Immune Responses. Immunity, 2017. 46(1): p. 120–132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Holtkamp SJ, et al. , Circadian clocks guide dendritic cells into skin lymphatics. Nature Immunology, 2021. 22(11): p. 1375–1381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Mendez-Ferrer S, et al. , Haematopoietic stem cell release is regulated by circadian oscillations. Nature, 2008. 452(7186): p. 442–7. [DOI] [PubMed] [Google Scholar]
  • 104.Gibbs J, et al. , An epithelial circadian clock controls pulmonary inflammation and glucocorticoid action. Nat Med, 2014. 20(8): p. 919–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Lam MT, et al. , Rev-Erbs repress macrophage gene expression by inhibiting enhancer-directed transcription. Nature, 2013. 498(7455): p. 511–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Wang S, et al. , REV-ERBα integrates colon clock with experimental colitis through regulation of NF-κB/NLRP3 axis. Nature Communications, 2018. 9(1): p. 4246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Allen NC, et al. , Desynchronization of the molecular clock contributes to the heterogeneity of the inflammatory response. Sci Signal, 2019. 12(571). [DOI] [PubMed] [Google Scholar]
  • 108.Oliva-Ramírez J, et al. , Crosstalk between circadian rhythmicity, mitochondrial dynamics and macrophage bactericidal activity. Immunology, 2014. 143(3): p. 490–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Blacher E, et al. , Aging disrupts circadian gene regulation and function in macrophages. Nat Immunol, 2022. 23(2): p. 229–236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.N AG, et al. , Phagocytosis imprints heterogeneity in tissue-resident macrophages. J Exp Med, 2017. 214(5): p. 1281–1296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Kitchen GB, et al. , The clock gene Bmal1 inhibits macrophage motility, phagocytosis, and impairs defense against pneumonia. Proc Natl Acad Sci U S A, 2020. 117(3): p. 1543–1551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Logan RW, et al. , Altered circadian expression of cytokines and cytolytic factors in splenic natural killer cells of Per1(−/‒) mutant mice. J Interferon Cytokine Res, 2013. 33(3): p. 108–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Arjona A and Sarkar DK, Circadian oscillations of clock genes, cytolytic factors, and cytokines in rat NK cells. J Immunol, 2005. 174(12): p. 7618–24. [DOI] [PubMed] [Google Scholar]
  • 114.Baumann A, et al. , IgE-dependent activation of human mast cells and fMLP-mediated activation of human eosinophils is controlled by the circadian clock. Mol Immunol, 2015. 64(1): p. 76–81. [DOI] [PubMed] [Google Scholar]
  • 115.Cervantes-Silva MP, et al. , The circadian clock influences T cell responses to vaccination by regulating dendritic cell antigen processing. Nat Commun, 2022. 13(1): p. 7217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Wang C, et al. , Dendritic cells direct circadian anti-tumour immune responses. Nature, 2023. 614(7946): p. 136–143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Nobis CC, et al. , The circadian clock of CD8 T cells modulates their early response to vaccination and the rhythmicity of related signaling pathways. Proc Natl Acad Sci U S A, 2019. 116(40): p. 20077–20086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Fortier EE, et al. , Circadian variation of the response of T cells to antigen. J Immunol, 2011. 187(12): p. 6291–300. [DOI] [PubMed] [Google Scholar]
  • 119.Suzuki K, et al. , Adrenergic control of the adaptive immune response by diurnal lymphocyte recirculation through lymph nodes. J Exp Med, 2016. 213(12): p. 2567–2574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Penny HA, et al. , Rhythmicity of intestinal IgA responses confers oscillatory commensal microbiota mutualism. Science Immunology, 2022. 7(75): p. eabk2541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Cao Q, et al. , Circadian clock cryptochrome proteins regulate autoimmunity. Proc Natl Acad Sci U S A, 2017. 114(47): p. 12548–12553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.George TJ, et al. , Association of operative time of day with outcomes after thoracic organ transplant. Jama, 2011. 305(21): p. 2193–9. [DOI] [PubMed] [Google Scholar]
  • 123.Yang Z, et al. , Impact of Nighttime Lung Transplantation on Outcomes and Costs. Ann Thorac Surg, 2021. 112(1): p. 206–213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Gouchoe Doug A., E.Y.C., Darcy Christine E., Whitson Bryan A., Ganapathi Asvin M., Zhu Hua, Is Timing Everything? Examining Operative Time in Lung Transplants from 2006–2023. JHLT Open, 2024(2950–1334). [Google Scholar]
  • 125.Immohr MB, et al. , Impact of circadian rhythm and daytime variation on outcome after heart transplantation. Clin Transplant, 2023. 37(5): p. e14939. [DOI] [PubMed] [Google Scholar]
  • 126.Ren SS, et al. , Circadian Rhythms Have Effects on Surgical Outcomes of Liver Transplantation for Patients With Hepatocellular Carcinoma: A Retrospective Analysis of 147 Cases in a Single Center. Transplant Proc, 2019. 51(6): p. 1913–1919. [DOI] [PubMed] [Google Scholar]
  • 127.Halliday N, et al. , Is liver transplantation ‘out-of-hours’ non-inferior to ‘in-hours’ transplantation? A retrospective analysis of the UK Transplant Registry. BMJ Open, 2019. 9(2): p. e024917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Becker F, et al. , Impact of nighttime procedures on outcomes after liver transplantation. PLoS One, 2019. 14(7): p. e0220124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Montaigne D, et al. , Day-Time Declamping Is Associated with Better Outcomes in Kidney Transplantation: The Circarein Study. J Clin Med, 2021. 10(11). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Sugünes N, et al. , Analysis of the Effects of Day-Time vs. Night-Time Surgery on Renal Transplant Patient Outcomes. J Clin Med, 2019. 8(7). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Fechner G, et al. , Kidney’s nightshift, kidney’s nightmare? Comparison of daylight and nighttime kidney transplantation: impact on complications and graft survival. Transplant Proc, 2008. 40(5): p. 1341–4. [DOI] [PubMed] [Google Scholar]
  • 132.Guo QH, et al. , Comparison of nighttime and daytime operation on outcomes of kidney transplant with deceased donors: a retrospective analysis. Chin Med J (Engl), 2019. 132(4): p. 395–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Guerrero ER, et al. , Nighttime Kidney Transplant From Donor With Controlled Cardiac Death: Greater Functionality at the Cost of More Complications? Transplant Proc, 2021. 53(9): p. 2666–2671. [DOI] [PubMed] [Google Scholar]
  • 134.Brunschot DM, et al. , Nighttime kidney transplantation is associated with less pure technical graft failure. World J Urol, 2016. 34(7): p. 955–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Kienzl-Wagner K, et al. , Nighttime procedures are not associated with adverse outcomes in kidney transplantation. Transpl Int, 2013. 26(9): p. 879–85. [DOI] [PubMed] [Google Scholar]
  • 136.Yim WY, et al. , Donor circadian clock influences the long-term survival of heart transplantation by immunoregulation. Cardiovasc Res, 2023. 119(12): p. 2202–2212. [DOI] [PubMed] [Google Scholar]
  • 137.Ville S, et al. , Timing of Kidney Clamping and Deceased Donor Kidney Transplant Outcomes. Clin J Am Soc Nephrol, 2021. 16(11): p. 1704–1714. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES