Skip to main content
Advertisement
  • Loading metrics

Crosstalk between circadian clocks and pathogen niche

  • Helene Borrmann,

    Roles Conceptualization, Funding acquisition, Visualization, Writing – original draft

    Affiliation Berkeley Public Health, Molecular and Cell Biology Department, University of California Berkeley, Berkeley, California, United States of America

  • Filipa Rijo-Ferreira

    Roles Conceptualization, Funding acquisition, Project administration, Visualization, Writing – review & editing

    [email protected]

    Affiliations Berkeley Public Health, Molecular and Cell Biology Department, University of California Berkeley, Berkeley, California, United States of America, Chan Zuckerberg Biohub–San Francisco, San Francisco, California, United States of America

Abstract

Circadian rhythms are intrinsic 24-hour oscillations found in nearly all life forms. They orchestrate key physiological and behavioral processes, allowing anticipation and response to daily environmental changes. These rhythms manifest across entire organisms, in various organs, and through intricate molecular feedback loops that govern cellular oscillations. Recent studies describe circadian regulation of pathogens, including parasites, bacteria, viruses, and fungi, some of which have their own circadian rhythms while others are influenced by the rhythmic environment of hosts. Pathogens target specific tissues and organs within the host to optimize their replication. Diverse cellular compositions and the interplay among various cell types create unique microenvironments in different tissues, and distinctive organs have unique circadian biology. Hence, residing pathogens are exposed to cyclic conditions, which can profoundly impact host–pathogen interactions. This review explores the influence of circadian rhythms and mammalian tissue-specific interactions on the dynamics of pathogen–host relationships. Overall, this demonstrates the intricate interplay between the body’s internal timekeeping system and its susceptibility to pathogens, which has implications for the future of infectious disease research and treatment.

1. Background on circadian rhythms in mammals

The Earth’s rotation governs day and night cycles, which shapes the evolutionary adaptation of all organisms on our planet to these rhythmic environmental variations. Circadian rhythms are 24-hour cycles that are widely observed across all domains of life. In mammals, the circadian system operates in a hierarchical framework, including central, peripheral, and molecular clocks. The suprachiasmatic nucleus (SCN) in the brain is the central pacemaker that integrates systemic cues, for instance, from light through the retinohypothalamic tract. It processes internal signals and synchronizes peripheral clocks in different tissues, causing daily changes in physiology and organ functions [14] (Fig 1).

thumbnail
Fig 1. Circadian regulation of the body.

Circadian rhythms impact diverse physiological processes across the body. The central clock in the brain integrates external and internal signals and communicates with tissue clocks to regulate organ functions. These include the nervous and endocrine system [4], cardiovascular system [1], respiratory tract [95], digestive system [61], reproductive [119] and urinary system [128], muscle skeletal system [2], and skin [111].

https://doi.org/10.1371/journal.ppat.1012157.g001

At the molecular level, each cell has a clock [5] that is governed by self-sustained transcriptional translational feedback loops that generate oscillations in gene expression (Fig 2A) [6]. The 2 main activators BMAL1 (brain and muscle ARNT-like 1) and CLOCK (circadian locomotor output cycles kaput) heterodimerize and bind to genomic elements called E-boxes, thereby activating the expression of circadian regulated genes, including their own repressors. Period (PER1/2/3) and Cryptochrome (CYR1/2) form a repressive complex that inhibits the activity of BMAL1 and CLOCK, creating a negative feedback loop [7]. A second interlocked feedback loop comprises the repressors REV-ERBα/β and activators RORα/β/γ (RAR-related orphan receptors), which themselves contain E-boxes in their promoters and are regulated by BMAL1 and CLOCK. REV-ERBs and RORs compete for binding to ROR response elements (RORE) in regulatory regions of genes, including the Bmal1 promoter [6,8]. BMAL1/CLOCK further regulate the expression of PAR-bZip transcription factors (proline and acidic amino acid-rich basic leucine zipper), which bind to D-box elements and form a third arm of the cellular clock. These factors include DBP (D-box binding protein), TEF (thyrotroph embryonic factor), and HLF (hepatic leukemia factor) that interact with the REV-ERB/ROR-driven repressor NFIL3 (nuclear factor interleukin-3 regulated). In addition to transcriptional regulation, posttranscriptional and posttranslational cellular processes contribute to circadian oscillations [9,10]. Cellular rhythms can also persist without transcription, such as the oxidation–reduction cycles of peroxiredoxin proteins [11,12].

thumbnail
Fig 2. Molecular clock and circadian criteria.

(A) Every cell in the body has an intrinsic circadian clock, in which transcriptional/translational feedback loops coordinate rhythmic gene expression. The activators BMAL1:CLOCK bind to E-boxes in promoter regions of genes and activate the expression of REV-ERB, ROR, PER, and CRY. PER:CRY inhibit BMAL1 activity, and ROR and REV-ERB compete for binding to RORE’s, including in the Bmal1 promoter. The figure represents 2 out of the 3 interconnected loops that regulate the clock, focusing on those that have been connected to host–pathogens interactions. (B) Criteria for biological rhythms to classify as circadian rhythms: (1) endogenous free-running period of approximately 24 hours; (2) entrainment to cues called Zeitgebers, for instance, light pulse causing phase shift; and (3) temperature compensation.

https://doi.org/10.1371/journal.ppat.1012157.g002

The term “circadian” derives from “circa” and “diem,” Latin for “about” and “a day,” but it’s important to note that not all daily rhythms are considered circadian. To qualify as a circadian rhythm, a biological pattern must meet the following 3 criteria (Fig 2B): (1) It must have an endogenous free-running period of approximately 24 hours, which is maintained in the absence of environmental cues. (2) Circadian oscillations can be entrained by exposure to external stimuli, called Zeitgebers, that cause readjustment of the timing (phase and period) of the oscillations. (3) Circadian periodicity is temperature compensated, hence maintained over a range of physiological temperatures [13]. Circadian rhythms provide organisms with the ability to predict and adjust to recurrent environmental variations. This enables optimized resource utilization, including light and food, thereby enhancing their evolutionary fitness [1417]. There is increasing evidence that pathogens that infect mammals are regulated through circadian mechanisms of their hosts or have intrinsic clocks themselves [1820].

2. The importance of host environments for infections

Many pathogens, including bacteria, viruses, parasites, and fungi, exhibit dependency on hosts for survival. Through evolutionary adaptations, they have fine-tuned their replication strategies to maximize efficiency within specific niches in hosts. Variations in host microenvironments can either facilitate or hinder pathogen colonization, replication, and transmission, thus critically impacting disease outcomes [21,22]. These environments encompass a diverse interplay of physiological, immunological, and microbiological factors (Fig 3).

thumbnail
Fig 3. Pathogens are influenced by the host’s microenvironments.

Bacteria, parasites, viruses, and fungi have evolved to maximize their colonization, replication, and transmission in specific niches within hosts. Multiple factors can vary depending on the infected organ. These include nutrient availability, variations in oxygen levels, endocrine regulation, and the presence of tissue-resident cells. The circadian clock can influence all these elements.

https://doi.org/10.1371/journal.ppat.1012157.g003

Circadian rhythms are important in shaping organ-specific niches, for instance, by regulating differences in nutrient availability depending on the time of the day [23,24]. While this is primarily a mechanism to ensure optimal organ function, pathogens that infect the tissue may have an advantage from adopting and utilizing this cyclic nutrient availability. Specialized immune cells, such as tissue-resident macrophages and dendritic cells, serve as a first line of defense against pathogens entering specific tissues [25]. For instance, alveolar macrophages in the lung act against inhaled pathogens [26] and microglia in the brain play a crucial role in protecting the central nervous system from infections [27]. It has been shown that multiple arms of the immune system are under circadian regulation and that immune cell intrinsic clocks are important for an effective response to pathogens [28,29]. Around 8% of the transcriptome from murine peritoneal macrophages is under circadian regulation [30], and deletion of Bmal1 in T cells reduced rhythmic cell-intrinsic receptor expression in mice [31]. Moreover, the rhythmic recruitment of immune cells to tissues [32,33] can influence temporal regulation of the protection against pathogens in organs. Loss of Bmal1 reduced rhythmic recruitment of myeloid cells to arteries and veins [34]. Additionally, Bmal1 knockout (KO) increased dendritic cell migration into skin lymphatic vessels during the morning in mice [35]. Thus, circadian fluctuations in immune responses may differ among distinct organs, emphasizing the role of circadian regulation in host environments as an important factor in the context of infections.

Selective pressures often drive the emergence of new pathogen variants and strains with distinct virulence profiles and host specificities. Therefore, a comprehensive understanding of the intricate relationships within host environments, including circadian rhythms, is indispensable for advancing our strategies for infectious disease prevention and treatment.

3. Interplay between clocks and pathogens in different host niches

Circadian rhythms manifest within host tissue environments, and numerous pathogens target comparable host niches, thereby experiencing similar circadian environments. Here, we will summarize the literature advances in the field over the past years, mostly focusing on each pathogenic infection when encountering the tissue (Fig 4). However, it is likely that infections also change the circadian regulation of the tissue and thus would be interesting to study it in the context of coinfections in the future.

thumbnail
Fig 4. Interplay between body clocks and pathogens.

Different pathogens target specific organs for replication, and some are capable of infecting multiple organs at various life stages. An interplay with circadian rhythms has been demonstrated for the pathogens shown. Liver: hepatitis B virus [44,45,51], hepatitis C virus [46,48, 50]. Blood: dengue virus [46], HIV [7477], Leishmania [78], Plasmodium [8690], Trypanosoma [91,92]. Respiratory tract: Aspergillus fumigatus [109,110], influenza virus [31,97100], respiratory syncytial virus [104], SARS-CoV-2 [101103], Sendai virus [98], Streptococcus pneumoniae [107,108]. Reproductive and urinary system: Candida albicans [129], Chlamydia muridarum [124126]. Gastrointestinal tract: Citobacter rodentium [71], Clostridium difficile [70], Helicobacter pylori [31], Klebsiella aerogenes [65], Listeria monocytogenes [72], microbiome [53,62,67], Salmonella [68,69], Trichuris muris [73]. Skin: herpes virus [97,115117], Staphylococcus aureus [114]. Brain: herpes virus [131], vesicular stomatitis virus [135], Zika virus [46], Trypanosoma [133].

https://doi.org/10.1371/journal.ppat.1012157.g004

The liver

The liver is the body’s largest solid internal organ and plays a central role in metabolism, detoxification, and the synthesis of essential proteins. Various cell types in the liver, including hepatocytes, sinusoidal endothelial cells, and Kupffer cells, cooperate spatiotemporally to shape and maintain liver functions [36]. A highly organized architectural pattern and vascularization receiving blood from both the hepatic artery and portal vein results in liver zonation. It causes specific microenvironments with varying oxygen tensions, nutrients, and toxins [37,38], which can influence colonization by pathogens.

The liver is one of the most circadian regulated organs with 20% of the transcriptome showing rhythmic expression [39,40], which impacts various of its functions [41,42]. Circadian disruption results in dysregulation of bile acid homeostasis in the liver [43], and transcription of the transmembrane mediator of bile acid transport (sodium taurocholate cotransporting polypeptide, NTCP) is regulated by the clock transcription factors REV-ERBα and β [44]. Interestingly, NTCP is required for entry of hepatitis B virus (HBV) into cells, and HBV is further regulated by the circadian machinery through BMAL1 binding to the viral promoter [44]. Similarly, entry of hepatitis C virus (HCV) into hepatocytes is regulated by BMAL1, and Bmal1 silencing or activation of REV-ERBα/β with synthetic agonists inhibits the replication of HCV in vitro. Lipids are essential for generation of new viral particles, and REV-ERBα was shown to regulate lipid and cholesterol metabolism [45]. Consistently, activation of REV-ERBα reduced expression of stearoyl-CoA-desaturase, a rate limiting enzyme for HCV replication [46]. Circadian rhythms and liver metabolism are connected through peroxisome proliferator-activated receptors (PPAR), which are rhythmically expressed, and control glucose and lipid metabolism [47]. Liver metabolism was shown to be disturbed in HCV-related cirrhosis through deregulation of BMAL1 [48]. Hepatic viruses can promote the formation of hepatocellular carcinoma, and the expression of Per1-3 and Cry2 was reduced in this tissue, which indicates that the crosstalk is bidirectional [4951]. Interestingly, the initiation and progression of virus-related liver cirrhosis was partially regulated by the gut microbiota through the gut–liver axis [52], and microbiota regulated the circadian liver transcriptome and detoxification pattern [53].

Malaria infection of the host is initiated in the liver where Plasmodium parasites replicate during various days, but there is limited knowledge about the circadian regulation of this phase in the parasite life cycle. Hypoxia promotes liver-stage malaria infection [54], and an interplay of hypoxic and circadian pathways has been described [55,56], raising the question if this could influence hepatic Plasmodium replication. Neutrophils were shown to infiltrate the liver in a time-of-day–dependent manner in uninfected mice, where neutrophil numbers peaked at the onset of the rest phase and were lowest at the beginning of the night [57]. Neutrophils are known to be important for the response to malaria [58], and while rhythmicity in neutrophil infiltration could result in daily oscillations of parasites in the liver, further studies are required to validate these hypotheses.

The gastrointestinal tract

The gastrointestinal tract is formed by various segments including the stomach, small intestine, and large intestine and functions as an interface connecting the external environment with the body. Its multifaceted role, encompassing digestion and protection, is evident in its distinctive microenvironments. These include high acidity, which aids in the digestive process, the presence of microvilli to enhance nutrient absorption, and the formation of gut-associated lymphoid tissue through tissue-resident immune cells. Additionally, the commensal gut microbiome, a complex community of microorganisms, contributes significantly to metabolism and highlights the importance of microbes for overall gut health [59,60]. Multiple metabolic processes are regulated by circadian rhythms, and metabolic signals provide feedback to the circadian timing system [24,61].

The intestinal microbiome exhibits daily oscillations [62] and levels of bacteria colonizing the epithelial layer were up to 10-fold higher during the active phase compared to the rest phase in mice [53]. Because these microbiome rhythms are almost absent in circadian clock mutant animals, there has not been a lot of research on the rhythms of the bacteria themselves. However, recent studies discovered that the commensal gut bacterium Bacillus subtilis has properties typically found in complex, multicellular circadian systems, including a free-running period, entrainment, and temperature compensation [63,64]. Similarly, the human gut bacterium Klebsiella aerogenes exhibited circadian rhythms in vitro, demonstrating entrainment to temperature cycles of different durations and amplitudes [65]. Rhythmic microbial and circadian cues are integrated by histone deacetylase 3, resulting in oscillations in histone acetylation, metabolic gene expression, and nutrient uptake [66]. Homeostasis in the intestinal epithelium is orchestrated by the circadian clock in conjunction with microbiota cues, which signal through rhythmic Toll-like receptor expression on intestinal epithelial cells [67]. Intestinal surface attachment of segmented filamentous bacteria in the mouse intestinal microbiota was shown to drive oscillations in innate immune response activation, resulting in periodic antimicrobial protein expression and rhythms in resistance to Salmonella typhimurium infection [68]. This is in line with findings of an increased proinflammatory response to Salmonella enterica and higher colonization levels when infection occurred during the rest period in mice [69].

The molecular clock of gut-resident immune cells, specifically type 3 innate lymphoid cells (ILC3s) was shown to influence circadian regulation of pathogens in the gastrointestinal tract. Mice with Bmal1-deficient ILC3s had more severe inflammation and higher bacterial burden of Clostridium difficile [70] and Citrobacter rodentium [71]. Moreover, clearance of Listeria monocytogenes was more effective when mice were infected intraperitoneally during the morning compared to the afternoon, which was regulated by diurnal oscillations of inflammatory monocytes [72]. The circadian immune system further contributed to the response against the rodent gut parasite Trichuris muris, with the circadian regulator BMAL1 governing the clock response in dendritic cells [73]. Mice were infected with Helicobacter pylori at 3 different times of the day and T cell counts in lymph nodes were quantified 3 weeks later. Cell numbers showed a circadian pattern with highest counts when infection occurred during the day, highlighting the role of circadian rhythms in modulating adaptive immune responses [31].

Generally, commensal and pathogenic gut bacteria are exposed to circadian rhythms in the gastrointestinal tract, and alterations in the gut microbiome can cause dysbiosis, which facilitates increased infections. This is likely to impact not only bacterial infections but also viruses, such as Norovirus, and parasitic infections like Giardiasis and Cryptosporidiosis in the gastrointestinal tract [19], which have not yet been explored within the circadian context.

The blood

The importance of blood is obvious considering its components: red blood cells for gas exchange, white blood cells for immune defense, platelets for blood clotting and transport of nutrients, hormones, and waste products inside the plasma. As blood travels throughout the body, it encounters various organs and is exposed to different peripheral clocks. Additionally, blood is important for systemic circadian clock synchronization, as it transmits information through soluble messenger signals and daily variations in blood pressure and temperature. Just like all other cells in the body, blood cells have intrinsic circadian rhythms and can be infected by pathogens.

The main replication site of human immunodeficiency virus (HIV) are T cells, and it was demonstrated that clock transcription factors regulate viral gene expression [74]. This results in rhythmic viral replication in vitro [75] and in patients [76,77]. Macrophages and neutrophils can be infected by the intracellular parasite Leishmania. The susceptibility to Leishmania infection varied over 24 hours in mice, which was regulated by the circadian expression of chemokines and subsequent rhythmic infiltration of immune cells at the infection site. These rhythms of higher susceptibility to Leishmania major at night were abolished in mice with clock-deficient immune cells, indicating that immune cell clocks control the magnitude of Leishmania parasite infection [78]. Another pathogen infecting immune cells is dengue virus, and it has been shown that REV-ERBα/β agonists can reduce viral replication [46].

Red blood cells do not have nuclei, yet they possess intrinsic 24 hours rhythms through posttranslational modification of peroxiredoxins and redox changes in hemoglobin [79,80]. During their asexual stages, malaria parasites undergo replication within red blood cells, resulting in periodic rupturing of these cells. This process is responsible for the characteristic cyclic fevers that are a defining feature of the disease [81,82]. The temporal dynamics of malaria biology have been investigated by various studies [8385], many of which found that the rhythmic behavior of Plasmodium is intricately governed by metabolic processes of the host [8689]. Recent evidence shows that malaria parasites are not driven by the host’s circadian rhythms but rather possess their own intrinsic rhythms, which are synchronized to the host. Approximately 80% of parasite genes were expressed rhythmically with a 24-hour period, and oscillations were only reduced to approximately 60% of cycling genes in the absence of host rhythmic cues, demonstrating that rhythms in malaria parasites are, in fact, intrinsic [90]. However, the complex interplay and coordination of circadian rhythms between the parasite and host remain to be elucidated.

The causative agent of sleeping sickness, Trypanosoma brucei, is another example of a blood-borne parasite under circadian regulation. In vitro culture revealed inherent circadian rhythms in T. brucei gene expression. Many of the parasite cycling genes were involved in metabolic pathways and parasite rhythms were not dependent on the cell cycle [91]. Interestingly, early studies found that Trypanosoma populations remained stable throughout the day in infected animals [92]. It is currently unknown whether there are circadian rhythms in infections of other blood-borne pathogens such as those that cause Chagas, Babesiosis, or Toxoplasmosis. Nonetheless, it is important to note that the timing of blood collection can impact the detection of immune markers and other pathogens [77], which should be considered in clinical settings.

The respiratory tract

The respiratory tract encompasses complex structures extending from the nose down to delicate bronchioles in the lungs. It is lined with mucosal membranes and ciliated cells, which facilitate gas exchange between the body and the external environment. Early observations of daily fluctuations in lung function were made by ancient Greek physicians around 100 CE, who described that symptoms of asthma manifested at night [93]. These findings were confirmed, and later studies revealed that exacerbated asthma symptoms at night are regulated through the endogenous circadian system independent of sleep and other daily cycles [94]. Airways exhibit daily rhythms in caliber, being most constricted in the early morning (2 AM to 5 AM) when individuals with asthma are at a higher risk of experiencing respiratory symptoms. The heightened airway resistance coincides with an increased tolerance to CO2 buildup, likely reducing the effort required for breathing during sleep [95]. The respiratory tract is exposed to dynamic fluxes of microbial colonization and clearance, and upper and lower parts have a topographically distinct microbial composition [96].

Several pathogens infecting respiratory organs are regulated by circadian rhythms, one of the best studied of which is influenza virus. Replication of the flu virus was increased in arrhythmic Bmal1 KO cells in vitro [97] and Bmal1-deficient mice in vivo [98]. Importantly, infection of mice at different time points did not result in changes in influenza viral load, and rhythms were conferred by rhythmicity in the immune response [31,99]. Infection at the start of the active phase promoted lung inflammation [99], further highlighting the importance of the tissue-specific environments on infection outcomes. Respiration largely defines the microenvironment in lungs, and, interestingly, exposure to neonatal excess oxygen abolished the clock-mediated time of day protection from influenza in mice. It was independent of viral burden, through host tolerance pathways, and loss of Bmal1 in alveolar type 2 cells recapitulated increased mortality. This suggests that disruption is mediated by the lung’s internal clock, rather than central or immune systemic clocks [100].

Circadian disruption altered the lung transcriptome in mice, which predisposes animals to viral infection, including coronavirus SARS-CoV-2 [101]. SARS-CoV-2 enters cells through the angiotensin-converting enzyme type 2 receptor, which is expressed rhythmically and controlled by posttranscriptional circadian mechanisms. This causes variations in SARS-CoV-2 entry depending on the time of day. Additionally, the replication of various SARS-CoV-2 strains was reduced in Bmal1 KO cells, which is likely regulated through an enhanced interferon-stimulated gene response [102,103]. The innate immune response in the lung further contributes to the circadian response to other viruses, for instance, pulmonary infection of Bmal1 KO mice with respiratory syncytial virus [104] and Sendai virus [98] produced more severe disease outcomes than in wild-type (WT) animals.

Circadian regulation in the lung has been tested using lipopolysaccharides (LPS)-induced inflammation to mimic gram-negative bacteria infection. Interestingly, the circadian repressor REV-ERBα plays a central role in the regulation of pulmonary inflammation, involving both myeloid and bronchial epithelial cells [105]. In airway epithelial cells, the circadian variation in pulmonary inflammatory responses was shown to be independent of rhythmic glucocorticoid signaling [106]. Mice had higher antibacterial immunity when infected with Streptococcus pneumoniae during the active phase, and genetic ablation of Bmal1 in bronchiolar cells disrupted rhythmic chemokine expression causing worse disease outcomes [107]. It was shown that BMAL1 inhibits macrophage motility and phagocytosis through regulation of a network of cell movement genes, thereby impairing the response against pneumonia in mice [108].

There is limited evidence regarding the circadian regulation of fungal infections in mammals; however, it has been studied in the context of the lung fungus Aspergillus fumigatus. In vitro, there was no variation in the phagocytic activity of macrophages over time, however, mice infected at night exhibited a 2-fold enhancement in lung clearance a few hours after the challenge compared with those inoculated in the morning [109]. Conversely, another study found that maximum colonization occurred when infection took place at the beginning of the active phase, when assessing fungal load at 1 or 3 days post-infection. This study further demonstrated that the circadian oscillation of tryptophan metabolism leads to diurnal changes in the immune response in the lung, regulating the outcome of A. fumigatus infection [110]. Variations in the timing of infection and organ collection between both studies underscore the importance of considering these timings in experimental designs.

The skin

As the largest organ exposed to daily rhythms in sunlight and radiation, oscillations are observed in multiple cell types across all layers of the skin [111]. The skin is colonized with microbiota, which showed daily changes that correlated with human activity [112]. There are only a few studied examples of circadian regulated pathogens that infect the skin, including herpesvirus and Staphylococcus aureus [113]. Survival of S. aureus on mouse skin was reduced when infection occurred at the start of the active phase compared to the end [114]. Cutaneous herpes simplex virus 2 (HSV-2) infection was less severe when mice were infected during the rest phase compared to the active phase [115], and acute HSV-1 infection was enhanced in arrhythmic Bmal1 KO mice [97]. On a molecular level, CLOCK interacts with viral proteins to remodel HSV chromatin [116] and was identified to be a component of the viral transcriptional complex [117].

Murine skin displays circadian-regulated expression of interferon-sensitive genes [118], and it is likely that this will affect the response to various other pathogens that infect the skin. Evidence is limited, but considering the circadian rhythms observed in the commensal microbiomes of other organs, it is plausible that a similar phenomenon holds true for the skin. Moreover, it is likely that circadian rhythms in the skin will not only affect direct pathogen infection but may also influence the invasion of pathogens transmitted by insect vectors.

The reproductive and urinary system

The unique microenvironment in the reproductive tract undergoes distinct changes affecting factors such as pH, mucosal integrity, and hormone levels. These changes are multi-oscillatory and follow seasonal, ovulatory, and circadian cycles and are mediated through both the hypothalamic–pituitary–gonadal and hypothalamic–pituitary–adrenal axes, where communication is bidirectional [119121]. Importantly, sex differences can contribute to variance of rhythms, for instance, in body temperature [122] and pubertal development [123], which should encourage inclusion of sex as a biological variable in circadian experiments.

Rhythms in the reproductive system can affect the susceptibility to pathogens that may impact fertility and reproductive health. Circadian disruption caused higher bacterial load, more pathology, and lower fertility rate in mice infected with Chlamydia muridarum [124]. Mice infected at the beginning of their rest phase shed more Chlamydia and were less fertile compared to infection at the beginning of the active phase [125]. Interestingly, different times of infection translated into differences in the vaginal, uterine, and ovary/oviduct microbiome, which were maintained even multiple weeks post-infection [126]. It is likely that these diurnal changes in the genital tract microbiome extend to infections of other pathogens and further research is warranted.

The circadian system can influence rhythms in the urinary tract, including bladder function [127] and kidney physiology. Renal plasma flow, glomerular filtration rate, and tubular reabsorption/secretion processes were shown to be rhythmic and peak during the active phase [128]. Despite limited research on the circadian regulation of pathogens infecting the urinary tract, findings suggest an impact on Candida albicans in the kidney [129]. Mice infected during the night exhibited reduced kidney colonization and weight loss, along with an increased survival rate compared to those infected during the day. This regulation was attributed to the neutrophil-intrinsic circadian clock and may extend to other kidney-infecting pathogens. C. albicans can infect various other organs, including the genitalia, which may also be subject to circadian regulation and warrants further investigation. This suggests that circadian immune processes may play a role in protecting against fungal infections, which is an intriguing and largely understudied topic for future research.

The brain

Considering that the SCN in the hypothalamus serves as the central clock [130], it is intriguing to explore how pathogens influence the brain, even though there are constraints related to the blood–brain barrier.

Early studies show that herpes simplex virus 1 can invade the suprachiasmatic nuclei in mice with depleted CD4+ and CD8+ T cells, suggesting a potential mechanism to disrupt the circadian clock [131]. Additionally, during advanced stages of T. brucei infection, parasites and inflammatory cells can infiltrate the brain, which causes the characteristic symptoms of altered sleep and impairment of circadian-regulated processes [132134]. Another pathogen crossing the blood–brain barrier is Zika virus, and viral replication is regulated by REV-ERBα/β through lipid signaling pathways [46]. However, these experiments were performed in a liver cell line, and the importance for brain-specific Zika virus infections remain to be elucidated. Encephalitis can be induced by vesicular stomatitis virus, and infection of mice at the beginning of the rest period caused higher mortality and increased inflammatory response compared to infection in the active cycle. This is likely mediated through a clock-regulated inflammatory response, as REV-ERBɑ inhibition at the start of the active phase abolished the phenotype [135].

It is worth considering the role of circadian rhythms on other neurotrophic pathogens, many of which can establish chronic infection and alter sleep rhythms, as demonstrated for Toxoplasma gondii [136]. Interestingly, crosstalk between the brain and daily changes in gut microbiota was shown to contribute to aging-associated neurodegenerative diseases [137]. However, how the microbiota–gut–brain axis and other pathogens interplay with brain circadian rhythms requires further investigation.

In summary, these examples of pathogen adjustments to the host niches underscore the significance of taking tissue-specific circadian control into account. It is likely that these extend even further to tissues that have not been studied in the circadian-pathogen context yet. For instance, it is interesting to speculate about the circadian rhythms of adipose tissue [138,139], which is a major reservoir of clock-regulated Trypanosoma parasites [91,140]. In addition to different environments within 1 host, it is fascinating how pathogens can adapt to various intermediate hosts and vectors [84,141]. Changes in cycling genes among different pathogen phases suggest the capability to adjust to diverse host settings, a characteristic feature of circadian clocks.

4. Therapeutic insights and future directions

There are several strategies for utilizing the insights on circadian pathogen regulation to enhance therapy [142145]. Administering medication or vaccines at specific times can strategically target the pathogen when it is most vulnerable and optimize the immune response at the right time of day [115,146].

The discovery that many pathogens have their own internal clocks could pave the way for novel treatment strategies [20]. If the pathogen’s clock regulators are different from the host’s, targeting the pathogen’s clock could result in a “jet-lagged” microorganism, making it more vulnerable to elimination by host defense mechanisms. It could provide a targeted approach without collateral disruption of the host clock; however, the feasibility of selectively manipulating the pathogen’s clock remains unclear, and our understanding of pathogen-intrinsic clocks is still limited, hindering the design of such compounds.

Another strategy is to pharmacologically modulate the circadian system of the host, using compounds that target clock transcription factors. This has the limitation that these drugs may exert systemic effects on various biological rhythms and pathways, leading to off-target effects. Nonetheless, combining broad circadian modifiers with pathogen-specific drugs could offer a more targeted approach. A better understanding of organ-specific and host–niche interactions between circadian rhythms and pathogens will help to refine and characterize these strategies.

A topic that has received limited attention is the interplay between circadian rhythms and coinfections involving multiple pathogens. These infections frequently involve pathogens replicating in the same host niche due to a locally compromised immune defense, facilitating the infection by other pathogens. This is particularly significant as it can lead to more severe outcomes. Exploring approaches to selectively target circadian clocks within specific host niches could provide a novel and efficient way to target multiple pathogens. Although this concept is in its early stages, the comprehensive understanding of the interplay between pathogens and circadian clocks across various host environments can guide future studies. We hope that this review provides a holistic view on this intricate relationship and stimulates a new perspective on circadian pathogen research.

Acknowledgments

We thank Goncalo Brito with assistance in figure design.

References

  1. 1. Crnko S, Du Pré BC, Sluijter JPG, Van Laake LW. Circadian rhythms and the molecular clock in cardiovascular biology and disease. Nat Rev Cardiol. 2019;16(7):437–447. Epub 2019/02/24. pmid:30796369.
  2. 2. Morrison M, Halson SL, Weakley J, Hawley JA. Sleep, circadian biology and skeletal muscle interactions: Implications for metabolic health. Sleep Med Rev. 2022;66:101700. pmid:36272396
  3. 3. Rijo-Ferreira F, Takahashi JS. Genomics of circadian rhythms in health and disease. Genome Med. 2019;11(1):82. Epub 2019/12/19. pmid:31847894; PubMed Central PMCID: PMC6916512.
  4. 4. Gamble KL, Berry R, Frank SJ, Young ME. Circadian clock control of endocrine factors. Nat Rev Endocrinol. 2014;10(8):466–475. Epub 2014/05/28. pmid:24863387; PubMed Central PMCID: PMC4304769.
  5. 5. Schibler U, Gotic I, Saini C, Gos P, Curie T, Emmenegger Y, et al. Clock-Talk: Interactions between Central and Peripheral Circadian Oscillators in Mammals. Cold Spring Harb Symp Quant Biol. 2015;80:223–232. Epub 2015/12/20. pmid:26683231.
  6. 6. Takahashi JS. Transcriptional architecture of the mammalian circadian clock. Nat Rev Genet. 2017;18(3):164–179. Epub 2016/12/19. pmid:27990019; PubMed Central PMCID: PMC5501165.
  7. 7. Partch CL, Green CB, Takahashi JS. Molecular architecture of the mammalian circadian clock. Trends Cell Biol. 2014;24(2):90–99. Epub 2013/08/07. pmid:23916625; PubMed Central PMCID: PMC3946763.
  8. 8. Guillaumond F, Dardente H, Giguere V, Cermakian N. Differential control of Bmal1 circadian transcription by REV-ERB and ROR nuclear receptors. J Biol Rhythms. 2005;20(5):391–403. Epub 2005/11/04. pmid:16267379.
  9. 9. Gallego M, Virshup DM. Post-translational modifications regulate the ticking of the circadian clock. Nat Rev Mol Cell Biol. 2007;8(2):139–148. Epub 2007/01/25. pmid:17245414.
  10. 10. Preußner M, Heyd F. Post-transcriptional control of the mammalian circadian clock: implications for health and disease. Pflugers Arch. 2016;468(6):983–991. Epub 2016/04/25. pmid:27108448; PubMed Central PMCID: PMC4893061.
  11. 11. O’Neill JS, van Ooijen G, Dixon LE, Troein C, Corellou F, Bouget FY, et al. Circadian rhythms persist without transcription in a eukaryote. Nature. 2011;469(7331):554–558. Epub 2011/01/29. pmid:21270895; PubMed Central PMCID: PMC3040569.
  12. 12. Edgar RS, Green EW, Zhao Y, van Ooijen G, Olmedo M, Qin X, et al. Peroxiredoxins are conserved markers of circadian rhythms. Nature. 2012;485(7399):459–464. Epub 2012/05/25. pmid:22622569; PubMed Central PMCID: PMC3398137.
  13. 13. Tsuchiya Y, Akashi M, Nishida E. Temperature compensation and temperature resetting of circadian rhythms in mammalian cultured fibroblasts. Genes Cells. 2003;8(8):713–720. Epub 2003/07/24. pmid:12875656.
  14. 14. Woelfle MA, Ouyang Y, Phanvijhitsiri K, Johnson CH. The adaptive value of circadian clocks: an experimental assessment in cyanobacteria. Curr Biol. 2004;14(16):1481–1486. Epub 2004/08/25. pmid:15324665.
  15. 15. Dodd AN, Salathia N, Hall A, Kévei E, Tóth R, Nagy F, et al. Plant circadian clocks increase photosynthesis, growth, survival, and competitive advantage. Science. 2005;309(5734):630–633. Epub 2005/07/26. pmid:16040710.
  16. 16. Acosta-Rodríguez V, Rijo-Ferreira F, Izumo M, Xu P, Wight-Carter M, Green CB, et al. Circadian alignment of early onset caloric restriction promotes longevity in male C57BL/6J mice. Science. 2022;376(6598):1192–1202. Epub 2022/05/06. pmid:35511946; PubMed Central PMCID: PMC9262309.
  17. 17. Turek FW, Joshu C, Kohsaka A, Lin E, Ivanova G, McDearmon E, et al. Obesity and metabolic syndrome in circadian Clock mutant mice. Science. 2005;308(5724):1043–1045. Epub 2005/04/23. pmid:15845877; PubMed Central PMCID: PMC3764501.
  18. 18. Borrmann H, McKeating JA, Zhuang X. The Circadian Clock and Viral Infections. J Biol Rhythms. 2021;36(1):9–22. Epub 2020/11/10. PubMed Central PMCID: PMC7924106. pmid:33161818
  19. 19. Pearson JA, Voisey AC, Boest-Bjerg K, Wong FS, Wen L. Circadian Rhythm Modulation of Microbes During Health and Infection. Front Microbiol. 2021;12:721004. Epub 2021/09/14. pmid:34512600; PubMed Central PMCID: PMC8430216.
  20. 20. Rijo-Ferreira F, Takahashi JS. Circadian rhythms in infectious diseases and symbiosis. Semin Cell Dev Biol. 2022;126:37–44. Epub 2021/10/10. pmid:34625370; PubMed Central PMCID: PMC9183220.
  21. 21. Casadevall A, Pirofski LA. Host-pathogen interactions: redefining the basic concepts of virulence and pathogenicity. Infect Immun. 1999;67(8):3703–3713. Epub 1999/07/23. pmid:10417127; PubMed Central PMCID: PMC96643.
  22. 22. Morgan AD, Koskella B. Coevolution of Host and Pathogen. In: Tibayrenc M, editor. Genetics and Evolution of Infectious Disease. London: Elsevier; 2011. p. 147–171.
  23. 23. Bass J. Circadian topology of metabolism. Nature. 2012;491(7424):348–356. pmid:23151577
  24. 24. Reinke H, Asher G. Crosstalk between metabolism and circadian clocks. Nat Rev Mol Cell Biol. 2019;20(4):227–241. pmid:30635659
  25. 25. Sun H, Sun C, Xiao W, Sun R. Tissue-resident lymphocytes: from adaptive to innate immunity. Cell Mol Immunol. 2019;16(3):205–215. Epub 2019/01/13. pmid:30635650; PubMed Central PMCID: PMC6460493.
  26. 26. Zazara DE, Belios I, Lücke J, Zhang T, Giannou AD. Tissue-resident immunity in the lung: a first-line defense at the environmental interface. Semin Immunopathol. 2022;44(6):827–854. pmid:36305904
  27. 27. Rock RB, Gekker G, Hu S, Sheng WS, Cheeran M, Lokensgard JR, et al. Role of microglia in central nervous system infections. Clin Microbiol Rev. 2004;17(4):942–964, table of contents. Epub 2004/10/19. pmid:15489356; PubMed Central PMCID: PMC523558.
  28. 28. Scheiermann C, Gibbs J, Ince L, Loudon A. Clocking in to immunity. Nat Rev Immunol. 2018;18(7):423–437. Epub 2018/04/18. pmid:29662121.
  29. 29. Wang C, Lutes LK, Barnoud C, Scheiermann C. The circadian immune system. Science Immunology. 2022;7(72):eabm2465. pmid:35658012
  30. 30. Keller M, Mazuch J, Abraham U, Eom GD, Herzog ED, Volk HD, et al. A circadian clock in macrophages controls inflammatory immune responses. Proc Natl Acad Sci U S A. 2009;106(50):21407–21412. Epub 2009/12/04. pmid:19955445; PubMed Central PMCID: PMC2795539.
  31. 31. Druzd D, Matveeva O, Ince L, Harrison U, He W, Schmal C, et al. Lymphocyte Circadian Clocks Control Lymph Node Trafficking and Adaptive Immune Responses. Immunity. 2017;46(1):120–132. Epub 2017/01/15. pmid:28087238; PubMed Central PMCID: PMC5263259.
  32. 32. Curtis AM, Bellet MM, Sassone-Corsi P, O’Neill LA. Circadian clock proteins and immunity. Immunity. 2014;40(2):178–186. Epub 2014/02/25. pmid:24560196.
  33. 33. Baxter M, Ray DW. Circadian rhythms in innate immunity and stress responses. Immunology. 2019. Epub 2019/12/11. pmid:31820826.
  34. 34. de Juan A, Ince LM, Pick R, Chen CS, Molica F, Zuchtriegel G, et al. Artery-Associated Sympathetic Innervation Drives Rhythmic Vascular Inflammation of Arteries and Veins. Circulation. 2019;140(13):1100–1114. Epub 2019/08/14. pmid:31401849; PubMed Central PMCID: PMC6756975.
  35. 35. Holtkamp SJ, Ince LM, Barnoud C, Schmitt MT, Sinturel F, Pilorz V, et al. Circadian clocks guide dendritic cells into skin lymphatics. Nat Immunol. 2021;22(11):1375–1381. Epub 2021/10/20. pmid:34663979; PubMed Central PMCID: PMC8553624.
  36. 36. Ding C, Li Y, Guo F, Jiang Y, Ying W, Li D, et al. A Cell-type-resolved Liver Proteome. Mol Cell Proteomics. 2016;15(10):3190–3202. Epub 2016/08/27. pmid:27562671; PubMed Central PMCID: PMC5054343.
  37. 37. Jungermann K, Kietzmann T. Oxygen: modulator of metabolic zonation and disease of the liver. Hepatology. 2000;31(2):255–260. Epub 2000/02/03. pmid:10655244.
  38. 38. Ben-Moshe S, Itzkovitz S. Spatial heterogeneity in the mammalian liver. Nat Rev Gastroenterol Hepatol. 2019;16(7):395–410. Epub 2019/04/03. pmid:30936469.
  39. 39. Nobuya Koike S-HY, Hung-Chung Huang, Vivek Kumar, Choogon Lee,2 Tae-Kyung Kim, Joseph S. Takahashi. Transcriptional Architecture and Chromatin Landscape of the Core Circadian Clock in Mammals. Science. 2012;338. PubMed Central PMCID: PMC3694775. pmid:22936566
  40. 40. Zhang R, Lahens NF, Ballance HI, Hughes ME, Hogenesch JB. A circadian gene expression atlas in mammals: implications for biology and medicine. Proc Natl Acad Sci U S A. 2014;111(45):16219–16224. Epub 2014/10/29. pmid:25349387; PubMed Central PMCID: PMC4234565.
  41. 41. Mukherji A, Bailey SM, Staels B, Baumert TF. The circadian clock and liver function in health and disease. J Hepatol. 2019;71(1):200–211. Epub 2019/04/02. pmid:30930223; PubMed Central PMCID: PMC7613420.
  42. 42. Bolshette N, Ibrahim H, Reinke H, Asher G. Circadian regulation of liver function: from molecular mechanisms to disease pathophysiology. Nat Rev Gastroenterol Hepatol. 2023. pmid:37291279
  43. 43. Ma K, Xiao R, Tseng H-T, Shan L, Fu L, Moore DD. Circadian Dysregulation Disrupts Bile Acid Homeostasis. PLoS ONE. 2009;4(8):e6843. pmid:19718444
  44. 44. Zhuang X, Forde D, Tsukuda S, D’Arienzo V, Mailly L, Harris JM, et al. Circadian control of hepatitis B virus replication. Nat Commun. 2021;12(1):1658. Epub 2021/03/14. pmid:33712578; PubMed Central PMCID: PMC7955118.
  45. 45. Zhang Y, Fang B, Emmett MJ, Damle M, Sun Z, Feng D, et al. Discrete functions of nuclear receptor Rev-erbα couple metabolism to the clock. Science. 2015;348(6242):1488–1492. pmid:26044300
  46. 46. Zhuang X, Magri A, Hill M, Lai AG, Kumar A, Rambhatla SB, et al. The circadian clock components BMAL1 and REV-ERBalpha regulate flavivirus replication. Nat Commun. 2019;10(1):377. Epub 2019/01/24. pmid:30670689; PubMed Central PMCID: PMC6343007.
  47. 47. Chen L, Yang G. PPARs Integrate the Mammalian Clock and Energy Metabolism. PPAR Res. 2014;2014:653017. Epub 2014/04/03. pmid:24693278; PubMed Central PMCID: PMC3945976.
  48. 48. Horii R, Honda M, Shirasaki T, Shimakami T, Shimizu R, Yamanaka S, et al. MicroRNA-10a Impairs Liver Metabolism in Hepatitis C Virus-Related Cirrhosis Through Deregulation of the Circadian Clock Gene Brain and Muscle Aryl Hydrocarbon Receptor Nuclear Translocator-Like 1. Hepatol Commun. 2019;3(12):1687–1703. Epub 2019/12/14. pmid:31832575; PubMed Central PMCID: PMC6887665.
  49. 49. Lin YM, Chang JH, Yeh KT, Yang MY, Liu TC, Lin SF, et al. Disturbance of circadian gene expression in hepatocellular carcinoma. Mol Carcinog. 2008;47(12):925–933. Epub 2008/04/30. pmid:18444243.
  50. 50. Benegiamo G, Mazzoccoli G, Cappello F, Rappa F, Scibetta N, Oben J, et al. Mutual antagonism between circadian protein period 2 and hepatitis C virus replication in hepatocytes. PLoS ONE. 2013;8(4):e60527. Epub 2013/04/18. pmid:23593233; PubMed Central PMCID: PMC3620463.
  51. 51. Yang SL, Yu C, Jiang JX, Liu LP, Fang X, Wu C. Hepatitis B virus X protein disrupts the balance of the expression of circadian rhythm genes in hepatocellular carcinoma. Oncol Lett. 2014;8(6):2715–2720. Epub 2014/11/02. pmid:25360177; PubMed Central PMCID: PMC4214404.
  52. 52. Wang T, Rong X, Zhao C. Circadian Rhythms Coordinated With Gut Microbiota Partially Account for Individual Differences in Hepatitis B-Related Cirrhosis. Front Cell Infect Microbiol. 2022;12:936815. Epub 2022/07/19. pmid:35846774; PubMed Central PMCID: PMC9283756.
  53. 53. Thaiss CA, Levy M, Korem T, Dohnalová L, Shapiro H, Jaitin DA, et al. Microbiota Diurnal Rhythmicity Programs Host Transcriptome Oscillations. Cell. 2016;167(6):1495–1510.e12. Epub 2016/12/03. pmid:27912059.
  54. 54. Ng S, March S, Galstian A, Hanson K, Carvalho T, Mota MM, et al. Hypoxia promotes liver-stage malaria infection in primary human hepatocytes in vitro. Dis Model Mech. 2014;7(2):215–224. Epub 2013/12/03. pmid:24291761; PubMed Central PMCID: PMC3917242.
  55. 55. Adamovich Y, Ladeuix B, Golik M, Koeners MP, Asher G. Rhythmic Oxygen Levels Reset Circadian Clocks through HIF1alpha. Cell Metab. 2017;25(1):93–101. Epub 2016/10/25. pmid:27773695.
  56. 56. O’Connell EJ, Martinez C-A, Liang YG, Cistulli PA, Cook KM. Out of Breath, Out of Time: Interactions between HIF and Circadian Rhythms. Am J Physiol Cell Physiol. 2020. pmid:32726159.
  57. 57. Crespo M, Gonzalez-Teran B, Nikolic I, Mora A, Folgueira C, Rodríguez E, et al. Neutrophil infiltration regulates clock-gene expression to organize daily hepatic metabolism. Elife. 2020;9:e59258. pmid:33287957
  58. 58. Rocha BC, Marques PE, Leoratti FMS, Junqueira C, Pereira DB, Antonelli L, et al. Type I Interferon Transcriptional Signature in Neutrophils and Low-Density Granulocytes Are Associated with Tissue Damage in Malaria. Cell Rep. 2015;13(12):2829–2841. Epub 2015/12/30. pmid:26711347; PubMed Central PMCID: PMC4698035.
  59. 59. Schmidt TSB, Raes J, Bork P. The Human Gut Microbiome: From Association to Modulation. Cell. 2018;172(6):1198–1215. Epub 2018/03/10. pmid:29522742.
  60. 60. Cho I, Blaser MJ. The human microbiome: at the interface of health and disease. Nat Rev Genet. 2012;13(4):260–270. pmid:22411464
  61. 61. Segers A, Depoortere I. Circadian clocks in the digestive system. Nat Rev Gastroenterol Hepatol. 2021;18(4):239–251. pmid:33531700
  62. 62. Gutierrez Lopez DE, Lashinger LM, Weinstock GM, Bray MS. Circadian rhythms and the gut microbiome synchronize the host’s metabolic response to diet. Cell Metab. 2021;33(5):873–887. Epub 2021/04/01. pmid:33789092.
  63. 63. Eelderink-Chen Z, Bosman J, Sartor F, Dodd AN, Kovács Á T, Merrow M. A circadian clock in a nonphotosynthetic prokaryote. Sci Adv. 2021;7(2). Epub 2021/02/02. pmid:33523996; PubMed Central PMCID: PMC7793578.
  64. 64. Sengupta S, Ince L, Sartor F, Borrmann H, Zhuang X, Naik A, et al. Clocks, Viruses, and Immunity: Lessons for the COVID-19 Pandemic. J Biol Rhythms. 2021;36(1):23–34. pmid:33480287.
  65. 65. Paulose JK, Cassone CV, Graniczkowska KB, Cassone VM. Entrainment of the Circadian Clock of the Enteric Bacterium Klebsiella aerogenes by Temperature Cycles. iScience. 2019;19:1202–1213. Epub 2019/09/26. pmid:31551197; PubMed Central PMCID: PMC6831877.
  66. 66. Kuang Z, Wang Y, Li Y, Ye C, Ruhn KA, Behrendt CL, et al. The intestinal microbiota programs diurnal rhythms in host metabolism through histone deacetylase 3. Science. 2019;365(6460):1428–1434. Epub 2019/10/12. pmid:31604271; PubMed Central PMCID: PMC7158748.
  67. 67. Mukherji A, Kobiita A, Ye T, Chambon P. Homeostasis in intestinal epithelium is orchestrated by the circadian clock and microbiota cues transduced by TLRs. Cell. 2013;153(4):812–827. Epub 2013/05/15. pmid:23663780.
  68. 68. Brooks JF 2nd, Behrendt CL, Ruhn KA, Lee S, Raj P, Takahashi JS, et al. The microbiota coordinates diurnal rhythms in innate immunity with the circadian clock. Cell. 2021;184(16):4154–4167.e12. Epub 2021/07/30. pmid:34324837; PubMed Central PMCID: PMC8967342.
  69. 69. Bellet MM, Deriu E, Liu JZ, Grimaldi B, Blaschitz C, Zeller M, et al. Circadian clock regulates the host response to Salmonella. Proc Natl Acad Sci U S A. 2013;110(24):9897–9902. Epub 2013/05/30. pmid:23716692; PubMed Central PMCID: PMC3683799.
  70. 70. Wang Q, Robinette ML, Billon C, Collins PL, Bando JK, Fachi JL, et al. Circadian rhythm-dependent and circadian rhythm-independent impacts of the molecular clock on type 3 innate lymphoid cells. Sci Immunol. 2019;4(40). Epub 2019/10/06. pmid:31586012; PubMed Central PMCID: PMC6911370.
  71. 71. Godinho-Silva C, Domingues RG, Rendas M, Raposo B, Ribeiro H, da Silva JA, et al. Light-entrained and brain-tuned circadian circuits regulate ILC3s and gut homeostasis. Nature. 2019;574(7777):254–258. Epub 2019/09/20. pmid:31534216; PubMed Central PMCID: PMC6788927.
  72. 72. Nguyen KD, Fentress SJ, Qiu Y, Yun K, Cox JS, Chawla A. Circadian gene Bmal1 regulates diurnal oscillations of Ly6C(hi) inflammatory monocytes. Science. 2013;341(6153):1483–1488. Epub 2013/08/24. pmid:23970558; PubMed Central PMCID: PMC3836670.
  73. 73. Hopwood TW, Hall S, Begley N, Forman R, Brown S, Vonslow R, et al. The circadian regulator BMAL1 programmes responses to parasitic worm infection via a dendritic cell clock. Sci Rep. 2018;8(1):3782. Epub 2018/03/02. pmid:29491349; PubMed Central PMCID: PMC5830501.
  74. 74. Borrmann H, Davies R, Dickinson M, Pedroza-Pacheco I, Schilling M, Vaughan-Jackson A, et al. Pharmacological activation of the circadian component REV-ERB inhibits HIV-1 replication. Sci Rep. 2020;10(1):13271. Epub 2020/08/09. pmid:32764708; PubMed Central PMCID: PMC7413328.
  75. 75. Borrmann H, Ulkar G, Kliszczak AE, Ismed D, Schilling M, Magri A, et al. Molecular components of the circadian clock regulate HIV-1 replication. iScience. 2023;26(7):107007. pmid:37534138
  76. 76. Stern J, Solomon A, Dantanarayana A, Pascoe R, Reynaldi A, Davenport MP, et al. Cell-Associated Human Immunodeficiency Virus (HIV) Ribonucleic Acid Has a Circadian Cycle in Males With HIV on Antiretroviral Therapy. J Infect Dis. 2022;225(10):1721–1730. Epub 2021/10/17. pmid:34655216.
  77. 77. Chang CC, Naranbhai V, Stern J, Roche M, Dantanarayana A, Ke R, et al. Variation in cell-associated unspliced HIV RNA on antiretroviral therapy is associated with the circadian regulator brain-and-muscle-ARNT-like-1. AIDS. 2018;32(15):2119–2128. Epub 2018/07/14. pmid:30005017; PubMed Central PMCID: PMC6173794.
  78. 78. Kiessling S, Dubeau-Laramée G, Ohm H, Labrecque N, Olivier M, Cermakian N. The circadian clock in immune cells controls the magnitude of Leishmania parasite infection. Sci Rep. 2017;7(1):10892. Epub 2017/09/09. pmid:28883509; PubMed Central PMCID: PMC5589941.
  79. 79. O’Neill JS, Reddy AB. Circadian clocks in human red blood cells. Nature. 2011;469(7331):498–503. Epub 2011/01/29. pmid:21270888; PubMed Central PMCID: PMC3040566.
  80. 80. Beale AD, Hayter EA, Crosby P, Valekunja UK, Edgar RS, Chesham JE, et al. Mechanisms and physiological function of daily haemoglobin oxidation rhythms in red blood cells. EMBO J. 2023;42(19):e114164. Epub 2023/08/09. pmid:37554073; PubMed Central PMCID: PMC10548169.
  81. 81. Mideo N, Reece SE, Smith AL, Metcalf CJE. The Cinderella syndrome: why do malaria-infected cells burst at midnight? Trends Parasitol. 2013;29(1):10–16. pmid:23253515
  82. 82. Cowman AF, Healer J, Marapana D, Marsh K. Malaria: Biology and Disease. Cell. 2016;167(3):610–624. Epub 2016/10/22. pmid:27768886.
  83. 83. Hunter FK, Butler TD, Gibbs JE. Circadian rhythms in immunity and host-parasite interactions. Parasite Immunol. 2022;44(3):e12904. Epub 2022/01/01. pmid:34971451; PubMed Central PMCID: PMC9285061.
  84. 84. Prior KF, Rijo-Ferreira F, Assis PA, Hirako IC, Weaver DR, Gazzinelli RT, et al. Periodic Parasites and Daily Host Rhythms. Cell Host Microbe. 2020;27(2):176–187. Epub 2020/02/14. pmid:32053788; PubMed Central PMCID: PMC7137616.
  85. 85. Rijo-Ferreira F, Takahashi JS, Figueiredo LM. Circadian rhythms in parasites. PLoS Pathog. 2017;13(10):e1006590. Epub 2017/10/13. pmid:29023533; PubMed Central PMCID: PMC5638552.
  86. 86. O’Donnell AJ, Mideo N, Reece SE. Disrupting rhythms in Plasmodium chabaudi: costs accrue quickly and independently of how infections are initiated. Malar J. 2013;12:372. Epub 2013/10/29. pmid:24160251; PubMed Central PMCID: PMC3819465.
  87. 87. Hirako IC, Assis PA, Hojo-Souza NS, Reed G, Nakaya H, Golenbock DT, et al. Daily Rhythms of TNFα Expression and Food Intake Regulate Synchrony of Plasmodium Stages with the Host Circadian Cycle. Cell Host Microbe. 2018;23(6):796–808.e6. Epub 2018/05/29. pmid:29805094; PubMed Central PMCID: PMC6014587.
  88. 88. Prior KF, van der Veen DR, O’Donnell AJ, Cumnock K, Schneider D, Pain A, et al. Timing of host feeding drives rhythms in parasite replication. PLoS Pathog. 2018;14(2):e1006900. pmid:29481559
  89. 89. O’Donnell AJ, Greischar MA, Reece SE. Mistimed malaria parasites re-synchronize with host feeding-fasting rhythms by shortening the duration of intra-erythrocytic development. Parasite Immunol. 2022;44(3):e12898. Epub 2021/11/16. pmid:34778983; PubMed Central PMCID: PMC9285586.
  90. 90. Rijo-Ferreira F, Acosta-Rodriguez VA, Abel JH, Kornblum I, Bento I, Kilaru G, et al. The malaria parasite has an intrinsic clock. Science. 2020;368(6492):746–753. pmid:32409471
  91. 91. Rijo-Ferreira F, Pinto-Neves D, Barbosa-Morais NL, Takahashi JS, Figueiredo LM. Trypanosoma brucei metabolism is under circadian control. Nat Microbiol. 2017;2:17032. Epub 2017/03/14. pmid:28288095; PubMed Central PMCID: PMC5398093.
  92. 92. Hawking F. Circadian rhythms of Trypanosoma congolense in laboratory rodents. Trans R Soc Trop Med Hyg. 1978;72(6):592–595. Epub 1978/01/01. pmid:366815
  93. 93. Adams F. The extant works of aretaeus: the cappadocian. Sydenham Society; 1856.
  94. 94. Scheer FAJL, Hilton MF, Evoniuk HL, Shiels SA, Malhotra A, Sugarbaker R, et al. The endogenous circadian system worsens asthma at night independent of sleep and other daily behavioral or environmental cycles. Proc Natl Acad Sci. 2021;118(37):e2018486118. pmid:34493686
  95. 95. Nosal C, Ehlers A, Haspel JA. Why Lungs Keep Time: Circadian Rhythms and Lung Immunity. Annu Rev Physiol. 2020;82:391–412. Epub 2019/09/29. pmid:31561746; PubMed Central PMCID: PMC8818323.
  96. 96. Natalini JG, Singh S, Segal LN. The dynamic lung microbiome in health and disease. Nat Rev Microbiol. 2023;21(4):222–235. pmid:36385637
  97. 97. Edgar RS, Stangherlin A, Nagy AD, Nicoll MP, Efstathiou S, O’Neill JS, et al. Cell autonomous regulation of herpes and influenza virus infection by the circadian clock. Proc Natl Acad Sci U S A. 2016;113(36):10085–10090. Epub 2016/08/17. pmid:27528682; PubMed Central PMCID: PMC5018795.
  98. 98. Ehlers A, Xie W, Agapov E, Brown S, Steinberg D, Tidwell R, et al. BMAL1 links the circadian clock to viral airway pathology and asthma phenotypes. Mucosal Immunol. 2018;11(1):97–111. Epub 2017/04/13. pmid:28401936; PubMed Central PMCID: PMC5638664.
  99. 99. Sengupta S, Tang SY, Devine JC, Anderson ST, Nayak S, Zhang SL, et al. Circadian control of lung inflammation in influenza infection. Nat Commun. 2019;10(1):4107. Epub 2019/09/13. pmid:31511530; PubMed Central PMCID: PMC6739310.
  100. 100. Issah Y, Naik A, Tang SY, Forrest K, Brooks TG, Lahens N, et al. Loss of circadian protection against influenza infection in adult mice exposed to hyperoxia as neonates. Elife. 2021;10. Epub 2021/03/03. pmid:33650487; PubMed Central PMCID: PMC7924938.
  101. 101. Taylor L, Von Lendenfeld F, Ashton A, Sanghani H, Di Pretoro S, Usselmann L, et al. Sleep and circadian rhythm disruption alters the lung transcriptome to predispose to viral infection. iScience. 2023;26(2):105877. pmid:36590897
  102. 102. Zhuang X, Tsukuda S, Wrensch F, Wing PAC, Schilling M, Harris JM, et al. The circadian clock component BMAL1 regulates SARS-CoV-2 entry and replication in lung epithelial cells. iScience. 2021;24(10):103144. Epub 2021/09/22. pmid:34545347; PubMed Central PMCID: PMC8443536.
  103. 103. Park A, Iwasaki A. Type I and Type III Interferons—Induction, Signaling, Evasion, and Application to Combat COVID-19. Cell Host Microbe. 2020;27(6):870–878. Epub 2020/05/29. pmid:32464097; PubMed Central PMCID: PMC7255347.
  104. 104. Majumdar T, Dhar J, Patel S, Kondratov R, Barik S. Circadian transcription factor BMAL1 regulates innate immunity against select RNA viruses. Innate Immun. 2017;23(2):147–154. Epub 2016/12/04. pmid:27913791.
  105. 105. Pariollaud M, Gibbs JE, Hopwood TW, Brown S, Begley N, Vonslow R, et al. Circadian clock component REV-ERBalpha controls homeostatic regulation of pulmonary inflammation. J Clin Invest. 2018;128(6):2281–2296. Epub 2018/03/14. pmid:29533925; PubMed Central PMCID: PMC5983347.
  106. 106. Ince LM, Zhang Z, Beesley S, Vonslow RM, Saer BR, Matthews LC, et al. Circadian variation in pulmonary inflammatory responses is independent of rhythmic glucocorticoid signaling in airway epithelial cells. FASEB J. 2019;33(1):126–139. Epub 2018/07/03. pmid:29965797; PubMed Central PMCID: PMC6355062.
  107. 107. Gibbs J, Ince L, Matthews L, Mei J, Bell T, Yang N, et al. An epithelial circadian clock controls pulmonary inflammation and glucocorticoid action. Nat Med. 2014;20(8):919–926. Epub 2014/07/30. pmid:25064128; PubMed Central PMCID: PMC4268501.
  108. 108. Kitchen GB, Cunningham PS, Poolman TM, Iqbal M, Maidstone R, Baxter M, 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):1543–1551. Epub 2020/01/05. pmid:31900362; PubMed Central PMCID: PMC6983378.
  109. 109. Chen S, Fuller KK, Dunlap JC, Loros JJ. Circadian Clearance of a Fungal Pathogen from the Lung Is Not Based on Cell-intrinsic Macrophage Rhythms. J Biol Rhythms. 2018;33(1):99–105. Epub 2017/12/29. pmid:29281921; PubMed Central PMCID: PMC5858702.
  110. 110. Stincardini C, Pariano M, D’Onofrio F, Renga G, Orecchini E, Orabona C, et al. The circadian control of tryptophan metabolism regulates the host response to pulmonary fungal infections. PNAS Nexus. 2023;2(3):pgad036. pmid:36896128
  111. 111. Lyons AB, Moy L, Moy R, Tung R. Circadian Rhythm and the Skin: A Review of the Literature. J Clin Aesthet Dermatol. 2019;12(9):42–45. Epub 2019/10/24. pmid:31641418; PubMed Central PMCID: PMC6777699.
  112. 112. Wilkins D, Tong X, Leung MHY, Mason CE, Lee PKH. Diurnal variation in the human skin microbiome affects accuracy of forensic microbiome matching. Microbiome. 2021;9(1):129. Epub 2021/06/07. pmid:34090519; PubMed Central PMCID: PMC8180031.
  113. 113. Duan J, Greenberg EN, Karri SS, Andersen B. The circadian clock and diseases of the skin. FEBS Lett. 2021;595(19):2413–2436. pmid:34535902
  114. 114. Bilska B, Zegar A, Slominski AT, Kleszczyński K, Cichy J, Pyza E. Expression of antimicrobial peptide genes oscillates along day/night rhythm protecting mice skin from bacteria. Exp Dermatol. 2021;30(10):1418–1427. Epub 2020/11/02. pmid:33131146; PubMed Central PMCID: PMC8085171.
  115. 115. Matsuzawa T, Nakamura Y, Ogawa Y, Ishimaru K, Goshima F, Shimada S, et al. Differential Day-Night Outcome to HSV-2 Cutaneous Infection. J Invest Dermatol. 2018;138(1):233–236. Epub 2017/08/27. pmid:28842321.
  116. 116. Kalamvoki M, Roizman B. Circadian CLOCK histone acetyl transferase localizes at ND10 nuclear bodies and enables herpes simplex virus gene expression. Proc Natl Acad Sci U S A. 2010;107(41):17721–17726. Epub 2010/09/30. pmid:20876123; PubMed Central PMCID: PMC2955081.
  117. 117. Kalamvoki M, Roizman B. The histone acetyltransferase CLOCK is an essential component of the herpes simplex virus 1 transcriptome that includes TFIID, ICP4, ICP27, and ICP22. J Virol. 2011;85(18):9472–9477. Epub 2011/07/08. pmid:21734043; PubMed Central PMCID: PMC3165755.
  118. 118. Greenberg EN, Marshall ME, Jin S, Venkatesh S, Dragan M, Tsoi LC, et al. Circadian control of interferon-sensitive gene expression in murine skin. Proc Natl Acad Sci U S A. 2020;117(11):5761–5771. Epub 2020/03/07. pmid:32132203; PubMed Central PMCID: PMC7084068.
  119. 119. Simonneaux V, Bahougne T. A Multi-Oscillatory Circadian System Times Female Reproduction. Front Endocrinol (Lausanne). 2015;6:157. Epub 2015/11/06. pmid:26539161; PubMed Central PMCID: PMC4611855.
  120. 120. Moeller JS, Bever SR, Finn SL, Phumsatitpong C, Browne MF, Kriegsfeld LJ. Circadian Regulation of Hormonal Timing and the Pathophysiology of Circadian Dysregulation Compr Physiol. 2022;12(4):4185–214.
  121. 121. Angelopoulou E, Quignon C, Kriegsfeld LJ, Simonneaux V. Functional Implications of RFRP-3 in the Central Control of Daily and Seasonal Rhythms in Reproduction. Front Endocrinol. 2019;10. pmid:31024442
  122. 122. Smarr B, Kriegsfeld LJ. Female mice exhibit less overall variance, with a higher proportion of structured variance, than males at multiple timescales of continuous body temperature and locomotive activity records. Biol Sex Differ. 2022;13(1):41. pmid:35870975
  123. 123. Grant AD, Wilbrecht L, Kriegsfeld LJ. Sex Differences in Pubertal Circadian and Ultradian Rhythmic Development Under Semi-naturalistic Conditions. J Biol Rhythms. 2022;37(4):442–454. Epub 2022/05/04. pmid:35502708; PubMed Central PMCID: PMC9329191.
  124. 124. Lundy SR, Richardson S, Ramsey A, Ellerson D, Fengxia Y, Onyeabor S, et al. Shift work influences the outcomes of Chlamydia infection and pathogenesis. Sci Rep. 2020;10(1):15389. pmid:32958779
  125. 125. Lundy SR, Ahmad T, Simoneaux T, Benyeogor I, Robinson Y, George Z, et al. Effect of Time of Day of Infection on Chlamydia Infectivity and Pathogenesis. Sci Rep. 2019;9(1):11405. Epub 2019/08/08. pmid:31388084; PubMed Central PMCID: PMC6684580.
  126. 126. Zhao L, Lundy SR, Eko FO, Igietseme JU, Omosun YO. Genital tract microbiome dynamics are associated with time of Chlamydia infection in mice. Sci Rep. 2023;13(1):9006. Epub 2023/06/03. pmid:37268696; PubMed Central PMCID: PMC10238418.
  127. 127. Ramsay S, Zagorodnyuk V. Role of circadian rhythms and melatonin in bladder function in heath and diseases. Auton Neurosci. 2023;246:103083. Epub 2023/03/06. pmid:36871511.
  128. 128. Firsov D, Bonny O. Circadian rhythms and the kidney. Nat Rev Nephrol. 2018;14(10):626–635. pmid:30143787
  129. 129. Adrover JM, Del Fresno C, Crainiciuc G, Cuartero MI, Casanova-Acebes M, Weiss LA, et al. A Neutrophil Timer Coordinates Immune Defense and Vascular Protection. Immunity. 2019;50(2):390–402.e10. Epub 2019/02/03. pmid:30709741.
  130. 130. Hastings MH, Maywood ES, Brancaccio M. Generation of circadian rhythms in the suprachiasmatic nucleus. Nat Rev Neurosci. 2018;19(8):453–469. pmid:29934559
  131. 131. Matsubara S, Atherton SS. Spread of HSV-1 to the suprachiasmatic nuclei and retina in T cell depleted BALB/c mice. J Neuroimmunol. 1997;80(1–2):165–171. Epub 1997/12/31. pmid:9413273.
  132. 132. Lundkvist GB, Kristensson K, Bentivoglio M. Why trypanosomes cause sleeping sickness. Physiology (Bethesda). 2004;19:198–206. Epub 2004/08/12. pmid:15304634.
  133. 133. Rijo-Ferreira F, Carvalho T, Afonso C, Sanches-Vaz M, Costa RM, Figueiredo LM, et al. Sleeping sickness is a circadian disorder. Nat Commun. 2018;9(1):62. pmid:29302035
  134. 134. Rijo-Ferreira F, Takahashi JS. Sleeping Sickness: A Tale of Two Clocks. Front Cell Infect Microbiol. 2020;10:525097. Epub 2020/11/03. pmid:33134186; PubMed Central PMCID: PMC7562814.
  135. 135. Gagnidze K, Hajdarovic KH, Moskalenko M, Karatsoreos IN, McEwen BS, Bulloch K. Nuclear receptor REV-ERBα mediates circadian sensitivity to mortality in murine vesicular stomatitis virus-induced encephalitis. Proc Natl Acad Sci. 2016;113(20):5730–5735. pmid:27143721
  136. 136. Dupont D, Lin JS, Peyron F, Akaoka H, Wallon M. Chronic Toxoplasma gondii infection and sleep-wake alterations in mice. CNS Neurosci Ther. 2021;27(8):895–907. Epub 2021/06/05. pmid:34085752; PubMed Central PMCID: PMC8265947.
  137. 137. Cheng WY, Ho YS, Chang RC. Linking circadian rhythms to microbiome-gut-brain axis in aging-associated neurodegenerative diseases. Ageing Res Rev. 2022;78:101620. Epub 2022/04/12. pmid:35405323.
  138. 138. Froy O, Garaulet M. The Circadian Clock in White and Brown Adipose Tissue: Mechanistic, Endocrine, and Clinical Aspects. Endocr Rev. 2018;39(3):261–273. Epub 2018/03/01. pmid:29490014; PubMed Central PMCID: PMC6456924.
  139. 139. Ribas-Latre A, Santos RB, Fekry B, Tamim YM, Shivshankar S, Mohamed AMT, et al. Cellular and physiological circadian mechanisms drive diurnal cell proliferation and expansion of white adipose tissue. Nat Commun. 2021;12(1):3482. pmid:34108488
  140. 140. Trindade S, Rijo-Ferreira F, Carvalho T, Pinto-Neves D, Guegan F, Aresta-Branco F, et al. Trypanosoma brucei Parasites Occupy and Functionally Adapt to the Adipose Tissue in Mice. Cell Host Microbe. 2016;19(6):837–848. Epub 2016/05/31. pmid:27237364; PubMed Central PMCID: PMC4906371.
  141. 141. Diallo AB, Coiffard B, Leone M, Mezouar S, Mege J-L. For Whom the Clock Ticks: Clinical Chronobiology for Infectious Diseases. Front Immunol. 2020;11. pmid:32733482
  142. 142. Ruben MD, Smith DF, FitzGerald GA, Hogenesch JB. Dosing time matters. Science. 2019;365(6453):547–549. pmid:31395773
  143. 143. Smith DF, Ruben MD, Francey LJ, Walch OJ, Hogenesch JB. When Should You Take Your Medicines? J Biol Rhythms. 2019;34(6):582–583. Epub 2019/12/10. pmid:31813349.
  144. 144. Miller S, Hirota T. Pharmacological Interventions to Circadian Clocks and Their Molecular Bases. J Mol Biol. 2020;432(12):3498–3514. Epub 2020/01/14. pmid:31931005.
  145. 145. Kojetin DJ, Burris TP. REV-ERB and ROR nuclear receptors as drug targets. Nat Rev Drug Discov. 2014;13(3):197–216. Epub 2014/03/01. pmid:24577401; PubMed Central PMCID: PMC4865262.
  146. 146. Cermakian N, Stegeman SK, Tekade K, Labrecque N. Circadian rhythms in adaptive immunity and vaccination. Semin Immunopathol. 2022;44(2):193–207. pmid:34825270
OSZAR »