Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 357
Filter
1.
Nat Commun ; 15(1): 3712, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38697963

ABSTRACT

The discovery of nitrogen fixation in unicellular cyanobacteria provided the first clues for the existence of a circadian clock in prokaryotes. However, recalcitrance to genetic manipulation barred their use as model systems for deciphering the clock function. Here, we explore the circadian clock in the now genetically amenable Cyanothece 51142, a unicellular, nitrogen-fixing cyanobacterium. Unlike non-diazotrophic clock models, Cyanothece 51142 exhibits conspicuous self-sustained rhythms in various discernable phenotypes, offering a platform to directly study the effects of the clock on the physiology of an organism. Deletion of kaiA, an essential clock component in the cyanobacterial system, impacted the regulation of oxygen cycling and hindered nitrogenase activity. Our findings imply a role for the KaiA component of the clock in regulating the intracellular oxygen dynamics in unicellular diazotrophic cyanobacteria and suggest that its addition to the KaiBC clock was likely an adaptive strategy that ensured optimal nitrogen fixation as microbes evolved from an anaerobic to an aerobic atmosphere under nitrogen constraints.


Subject(s)
Bacterial Proteins , Circadian Clocks , Cyanothece , Nitrogen Fixation , Oxygen , Oxygen/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Circadian Clocks/genetics , Circadian Clocks/physiology , Cyanothece/metabolism , Cyanothece/genetics , Nitrogenase/metabolism , Nitrogenase/genetics , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Circadian Rhythm Signaling Peptides and Proteins/genetics , Gene Expression Regulation, Bacterial , Cyanobacteria/metabolism , Cyanobacteria/genetics
2.
Biopolymers ; 115(2): e23559, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37421636

ABSTRACT

Circadian clocks are intracellular systems that orchestrate metabolic processes in anticipation of sunrise and sunset by providing an internal representation of local time. Because the ~24-h metabolic rhythms they produce are important to health across diverse life forms there is growing interest in their mechanisms. However, mechanistic studies are challenging in vivo due to the complex, that is, poorly defined, milieu of live cells. Recently, we reconstituted the intact circadian clock of cyanobacteria in vitro. It oscillates autonomously and remains phase coherent for many days with a fluorescence-based readout that enables real-time observation of individual clock proteins and promoter DNA simultaneously under defined conditions without user intervention. We found that reproducibility of the reactions required strict adherence to the quality of each recombinant clock protein purified from Escherichia coli. Here, we provide protocols for preparing in vitro clock samples so that other labs can ask questions about how changing environments, like temperature, metabolites, and protein levels are reflected in the core oscillator and propagated to regulation of transcription, providing deeper mechanistic insights into clock biology.


Subject(s)
Circadian Clocks , Cyanobacteria , Circadian Clocks/genetics , Reproducibility of Results , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Circadian Rhythm Signaling Peptides and Proteins/genetics , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Cyanobacteria/genetics , Cyanobacteria/metabolism
3.
Proc Natl Acad Sci U S A ; 120(13): e2221453120, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36940340

ABSTRACT

The circadian system of the cyanobacterium Synechococcus elongatus PCC 7942 relies on a three-protein nanomachine (KaiA, KaiB, and KaiC) that undergoes an oscillatory phosphorylation cycle with a period of ~24 h. This core oscillator can be reconstituted in vitro and is used to study the molecular mechanisms of circadian timekeeping and entrainment. Previous studies showed that two key metabolic changes that occur in cells during the transition into darkness, changes in the ATP/ADP ratio and redox status of the quinone pool, are cues that entrain the circadian clock. By changing the ATP/ADP ratio or adding oxidized quinone, one can shift the phase of the phosphorylation cycle of the core oscillator in vitro. However, the in vitro oscillator cannot explain gene expression patterns because the simple mixture lacks the output components that connect the clock to genes. Recently, a high-throughput in vitro system termed the in vitro clock (IVC) that contains both the core oscillator and the output components was developed. Here, we used IVC reactions and performed massively parallel experiments to study entrainment, the synchronization of the clock with the environment, in the presence of output components. Our results indicate that the IVC better explains the in vivo clock-resetting phenotypes of wild-type and mutant strains and that the output components are deeply engaged with the core oscillator, affecting the way input signals entrain the core pacemaker. These findings blur the line between input and output pathways and support our previous demonstration that key output components are fundamental parts of the clock.


Subject(s)
Circadian Clocks , Synechococcus , Circadian Clocks/genetics , Circadian Rhythm/genetics , Circadian Rhythm Signaling Peptides and Proteins/genetics , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Synechococcus/genetics , Synechococcus/metabolism , Phosphorylation , Adenosine Triphosphate/metabolism
4.
Environ Microbiol ; 25(4): 894-913, 2023 04.
Article in English | MEDLINE | ID: mdl-36579711

ABSTRACT

KaiC is the central cog of the circadian clock in Cyanobacteria. Close homologues of this protein are widespread among nonphotosynthetic bacteria, but the function, interaction network, and mechanism of action of these proteins are still largely unknown. Here, we focus on KaiC homologues found in environmental Pseudomonas species. Using bioinformatics, we describe the distribution of this protein family in the genus and reveal a conserved interaction network comprising a histidine kinase and response regulator. We characterize experimentally the only KaiC homologue present in Pseudomonas putida KT2440 and Pseudomonas protegens CHA0. Through phenotypic assays and transcriptomics, we show that KaiC is involved in osmotic and oxidative stress resistance in P. putida and in biofilm production in both species. KaiC homologues are found in different phosphorylation states and physically interact with a cognate histidine kinase and response regulator. In contrast with cyanobacterial counterparts, the expression and phosphorylation of KaiC homologues do not correlate with light variations under 12:12 light: dark cycles in either Pseudomonas species, and KaiC itself is not required to support a light-driven behaviour in P. putida. Overall, this suggests that KaiC homologues in Pseudomonas species are involved in environmental stress resistance but not in responses to diurnal rhythms.


Subject(s)
Bacterial Proteins , Cyanobacteria , Bacterial Proteins/metabolism , Histidine Kinase/genetics , Histidine Kinase/metabolism , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Circadian Rhythm/physiology , Cyanobacteria/metabolism , Phosphorylation , Pseudomonas/genetics , Pseudomonas/metabolism
5.
PLoS Comput Biol ; 18(9): e1010494, 2022 09.
Article in English | MEDLINE | ID: mdl-36067222

ABSTRACT

When the mixture solution of cyanobacterial proteins, KaiA, KaiB, and KaiC, is incubated with ATP in vitro, the phosphorylation level of KaiC shows stable oscillations with the temperature-compensated circadian period. Elucidating this temperature compensation is essential for understanding the KaiABC circadian clock, but its mechanism has remained a mystery. We analyzed the KaiABC temperature compensation by developing a theoretical model describing the feedback relations among reactions and structural transitions in the KaiC molecule. The model showed that the reduced structural cooperativity should weaken the negative feedback coupling among reactions and structural transitions, which enlarges the oscillation amplitude and period, explaining the observed significant period extension upon single amino-acid residue substitution. We propose that an increase in thermal fluctuations similarly attenuates the reaction-structure feedback, explaining the temperature compensation in the KaiABC clock. The model explained the experimentally observed responses of the oscillation phase to the temperature shift or the ADP-concentration change and suggested that the ATPase reactions in the CI domain of KaiC affect the period depending on how the reaction rates are modulated. The KaiABC clock provides a unique opportunity to analyze how the reaction-structure coupling regulates the system-level synchronized oscillations of molecules.


Subject(s)
Circadian Clocks , Circadian Rhythm Signaling Peptides and Proteins , Adenosine Diphosphate/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Bacterial Proteins/metabolism , Circadian Clocks/physiology , Circadian Rhythm , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Phosphorylation , Temperature
6.
Proc Natl Acad Sci U S A ; 119(37): e2202426119, 2022 09 13.
Article in English | MEDLINE | ID: mdl-36067319

ABSTRACT

The cyanobacterial clock presents a unique opportunity to understand the biochemical basis of circadian rhythms. The core oscillator, composed of the KaiA, KaiB, and KaiC proteins, has been extensively studied, but a complete picture of its connection to the physiology of the cell is lacking. To identify previously unknown components of the clock, we used KaiB locked in its active fold as bait in an immunoprecipitation/mass spectrometry approach. We found that the most abundant interactor, other than KaiC, was a putative diguanylate cyclase protein predicted to contain multiple Per-Arnt-Sim (PAS) domains, which we propose to name KidA. Here we show that KidA directly binds to the fold-switched active form of KaiB through its N-terminal PAS domains. We found that KidA shortens the period of the circadian clock both in vivo and in vitro and alters the ability of the clock to entrain to light-dark cycles. The dose-dependent effect of KidA on the clock period could be quantitatively recapitulated by a mathematical model in which KidA stabilizes the fold-switched form of KaiB, favoring rebinding to KaiC. Put together, our results show that the period and amplitude of the clock can be modulated by regulating the access of KaiB to the fold-switched form.


Subject(s)
Bacterial Proteins , Circadian Clocks , Circadian Rhythm Signaling Peptides and Proteins , Circadian Rhythm , Synechococcus , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Circadian Rhythm Signaling Peptides and Proteins/chemistry , Circadian Rhythm Signaling Peptides and Proteins/genetics , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Phosphorylation , Protein Domains , Synechococcus/physiology
7.
Nat Struct Mol Biol ; 29(8): 759-766, 2022 08.
Article in English | MEDLINE | ID: mdl-35864165

ABSTRACT

The AAA+ family member KaiC is the central pacemaker for circadian rhythms in the cyanobacterium Synechococcus elongatus. Composed of two hexameric rings of adenosine triphosphatase (ATPase) domains with tightly coupled activities, KaiC undergoes a cycle of autophosphorylation and autodephosphorylation on its C-terminal (CII) domain that restricts binding of clock proteins on its N-terminal (CI) domain to the evening. Here, we use cryogenic-electron microscopy to investigate how daytime and nighttime states of CII regulate KaiB binding on CI. We find that the CII hexamer is destabilized during the day but takes on a rigidified C2-symmetric state at night, concomitant with ring-ring compression. Residues at the CI-CII interface are required for phospho-dependent KaiB association, coupling ATPase activity on CI to cooperative KaiB recruitment. Together, these studies clarify a key step in the regulation of cyanobacterial circadian rhythms by KaiC phosphorylation.


Subject(s)
Circadian Clocks , Synechococcus , Adenosine Triphosphatases/metabolism , Bacterial Proteins/chemistry , CLOCK Proteins/metabolism , Circadian Rhythm , Circadian Rhythm Signaling Peptides and Proteins/genetics , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Phosphorylation , Synechococcus/metabolism
8.
Biochem J ; 479(14): 1505-1515, 2022 07 29.
Article in English | MEDLINE | ID: mdl-35771042

ABSTRACT

KaiC, a core protein of the cyanobacterial circadian clock, consists of an N-terminal CI domain and a C-terminal CII domain, and assembles into a double-ring hexamer upon binding with ATP. KaiC rhythmically phosphorylates and dephosphorylates its own two adjacent residues Ser431 and Thr432 at the CII domain with a period of ∼24 h through assembly and disassembly with the other clock proteins, KaiA and/or KaiB. In this study, to understand how KaiC alters its conformation as the source of circadian rhythm, we investigated structural changes of an inner-radius side of the CII ring using time-resolved Trp fluorescence spectroscopy. A KaiC mutant harboring a Trp fluorescence probe at a position of 419 exhibited a robust circadian rhythm with little temperature sensitivity in the presence of KaiA and KaiB. Our fluorescence observations show a remarkable environmental change at the inner-radius side of the CII ring during circadian oscillation. Crystallographic analysis revealed that a side chain of Trp at the position of 419 was oriented toward a region undergoing a helix-coil transition, which is considered to be a key event to allosterically regulate the CI ring that plays a crucial role in determining the cycle period. The present study provides a dynamical insight into how KaiC generates circadian oscillation.


Subject(s)
Circadian Clocks , Cyanobacteria , Bacterial Proteins/metabolism , Circadian Rhythm , Circadian Rhythm Signaling Peptides and Proteins/genetics , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Cyanobacteria/genetics , Cyanobacteria/metabolism , Fluorescence , Fluorescent Dyes/metabolism , Phosphorylation , Tryptophan/metabolism
9.
Int J Mol Sci ; 23(11)2022 May 30.
Article in English | MEDLINE | ID: mdl-35682819

ABSTRACT

Circadian rhythms are present in almost all living organisms, and their activity relies on molecular clocks. In prokaryotes, a functional molecular clock has been defined only in cyanobacteria. Here, we investigated the presence of circadian rhythms in non-cyanobacterial prokaryotes. The bioinformatic approach was used to identify a homologue of KaiC (circadian gene in cyanobacteria) in Escherichia coli. Then, strains of E. coli (wild type and mutants) were grown on blood agar, and sampling was made every 3 h for 24 h at constant conditions. Gene expression was determined by qRT-PCR, and the rhythmicity was analyzed using the Cosinor model. We identified RadA as a KaiC homologue in E. coli. Expression of radA showed a circadian rhythm persisting at least 3 days, with a peak in the morning. The circadian expression of other E. coli genes was also observed. Gene circadian oscillations were lost in radA mutants of E. coli. This study provides evidence of molecular clock gene expression in E. coli with a circadian rhythm. Such a finding paves the way for new perspectives in antibacterial treatment.


Subject(s)
Circadian Clocks , Cyanobacteria , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Circadian Clocks/genetics , Circadian Rhythm/genetics , Circadian Rhythm Signaling Peptides and Proteins/genetics , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Cyanobacteria/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Phosphorylation
10.
Proc Natl Acad Sci U S A ; 119(19): e2119627119, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35507871

ABSTRACT

KaiC is a dual adenosine triphosphatase (ATPase), with one active site in its N-terminal domain and another in its C-terminal domain, that drives the circadian clock system of cyanobacteria through sophisticated coordination of the two sites. To elucidate the coordination mechanism, we studied the contribution of the dual-ATPase activities in the ring-shaped KaiC hexamer and these structural bases for activation and inactivation. At the N-terminal active site, a lytic water molecule is sequestered between the N-terminal domains, and its reactivity to adenosine triphosphate (ATP) is controlled by the quaternary structure of the N-terminal ring. The C-terminal ATPase activity is regulated mostly by water-incorporating voids between the C-terminal domains, and the size of these voids is sensitive to phosphoryl modification of S431. The up-regulatory effect on the N-terminal ATPase activity inversely correlates with the affinity of KaiC for KaiB, a clock protein constitutes the circadian oscillator together with KaiC and KaiA, and the complete dissociation of KaiB from KaiC requires KaiA-assisted activation of the dual ATPase. Delicate interactions between the N-terminal and C-terminal rings make it possible for the components of the dual ATPase to work together, thereby driving the assembly and disassembly cycle of KaiA and KaiB.


Subject(s)
Circadian Clocks , Cyanobacteria , Adenosine Triphosphatases/metabolism , Bacterial Proteins/metabolism , CLOCK Proteins/metabolism , Circadian Rhythm , Circadian Rhythm Signaling Peptides and Proteins/genetics , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Cyanobacteria/metabolism , Phosphorylation
11.
Sci Adv ; 8(15): eabm8990, 2022 04 15.
Article in English | MEDLINE | ID: mdl-35427168

ABSTRACT

Spatiotemporal allostery is the source of complex but ordered biological phenomena. To identify the structural basis for allostery that drives the cyanobacterial circadian clock, we crystallized the clock protein KaiC in four distinct states, which cover a whole cycle of phosphor-transfer events at Ser431 and Thr432. The minimal set of allosteric events required for oscillatory nature is a bidirectional coupling between the coil-to-helix transition of the Ser431-dependent phospho-switch in the C-terminal domain of KaiC and adenosine 5'-diphosphate release from its N-terminal domain during adenosine triphosphatase cycle. An engineered KaiC protein oscillator consisting of a minimal set of the identified master allosteric events exhibited a monophosphorylation cycle of Ser431 with a temperature-compensated circadian period, providing design principles for simple posttranslational biochemical circadian oscillators.


Subject(s)
Circadian Clocks , Cyanobacteria , Adenosine Diphosphate/metabolism , Bacterial Proteins/metabolism , Circadian Rhythm , Circadian Rhythm Signaling Peptides and Proteins/genetics , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Cyanobacteria/metabolism , Phosphorylation
12.
PLoS Comput Biol ; 18(3): e1009243, 2022 03.
Article in English | MEDLINE | ID: mdl-35255087

ABSTRACT

Circadian clocks tick a rhythm with a nearly 24-hour period in a variety of organisms. In the clock proteins of cyanobacteria, KaiA, KaiB, and KaiC, known as a minimum circadian clock, the slow KaiB-KaiC complex formation is essential in determining the clock period. This complex formation, occurring when the C1 domain of KaiC hexamer binds ADP molecules produced by the ATPase activity of C1, is considered to be promoted by accumulating ADP molecules in C1 through inhibiting the ADP/ATP exchange (ADP release) rather than activating the ATP hydrolysis (ADP production). Significantly, this ADP/ATP exchange inhibition accelerates the complex formation together with its promotion, implying a potential role in the period robustness under environmental perturbations. However, the molecular mechanism of this simultaneous promotion and acceleration remains elusive because inhibition of a backward process generally slows down the whole process. In this article, to investigate the mechanism, we build several reaction models of the complex formation with the pre-binding process concerning the ATPase activity. In these models, six KaiB monomers cooperatively and rapidly bind to C1 when C1 binds ADP molecules more than a given threshold while stabilizing the binding-competent conformation of C1. Through comparison among the models proposed here, we then extract three requirements for the simultaneous promotion and acceleration: the stabilization of the binding-competent C1 by KaiB binding, slow ADP/ATP exchange in the binding-competent C1, and relatively fast ADP/ATP exchange occurring in the binding-incompetent C1 in the presence of KaiB. The last two requirements oblige KaiC to form a multimer. Moreover, as a natural consequence, the present models can also explain why the binding of KaiB to C1 reduces the ATPase activity of C1.


Subject(s)
Bacterial Proteins , Circadian Rhythm Signaling Peptides and Proteins , Acceleration , Adenosine Diphosphate/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Bacterial Proteins/metabolism , Circadian Rhythm , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Phosphorylation , Protein Binding
13.
Toxins (Basel) ; 14(2)2022 01 19.
Article in English | MEDLINE | ID: mdl-35202097

ABSTRACT

Shiga toxin-producing Escherichia coli (STEC) causes proximal tubular defects in the kidney. However, factors altered by Shiga toxin (Stx) within the proximal tubules are yet to be shown. We determined Stx receptor Gb3 in murine and human kidneys and confirmed the receptor expression in the proximal tubules. Stx2-injected mouse kidney tissues and Stx2-treated human primary renal proximal tubular epithelial cell (RPTEC) were collected and microarray analysis was performed. We compared murine kidney and RPTEC arrays and selected common 58 genes that are differentially expressed vs. control (0 h, no toxin-treated). We found that the most highly expressed gene was GDF15, which may be involved in Stx2-induced weight loss. Genes associated with previously reported Stx2 activities such as src kinase Yes phosphorylation pathway activation, unfolded protein response (UPR) and ribotoxic stress response (RSR) showed differential expressions. Moreover, circadian clock genes were differentially expressed, suggesting Stx2-induced renal circadian rhythm disturbance. Circadian rhythm-regulated proximal tubular Na+-glucose transporter SGLT1 (SLC5A1) was down-regulated, indicating proximal tubular functional deterioration, and mice developed glucosuria confirming proximal tubular dysfunction. Stx2 alters gene expression in murine and human proximal tubules through known activities and newly investigated circadian rhythm disturbance, which may result in proximal tubular dysfunctions.


Subject(s)
Circadian Rhythm Signaling Peptides and Proteins/metabolism , Circadian Rhythm , Gene Expression Regulation/drug effects , Kidney Tubules, Proximal/drug effects , Shiga Toxin 2/toxicity , Animals , Circadian Rhythm Signaling Peptides and Proteins/genetics , Epithelial Cells/drug effects , Glycosuria/chemically induced , Humans , Kidney Tubules, Proximal/cytology , Lipopolysaccharides/toxicity , Mice , Protein Array Analysis
14.
Biophys J ; 121(6): 943-955, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35151633

ABSTRACT

Cyanobacteria possesses the simplest circadian clock, composed of three proteins that act as a phosphorylation oscillator: KaiA, KaiB, and KaiC. The timing of this oscillator is determined by the fold-switch of KaiB, a structural rearrangement of its C-terminal half that is accompanied by a change in the oligomerization state. During the day, KaiB forms a stable tetramer (gsKaiB), whereas it adopts a monomeric thioredoxin-like fold during the night (fsKaiB). Although the structures and functions of both native states are well studied, little is known about the sequence and structure determinants that control their structural interconversion. Here, we used confinement molecular dynamics (CCR-MD) and folding simulations using structure-based models to show that the dissociation of the gsKaiB dimer is a key energetic event for the fold-switch. Hydrogen-deuterium exchange mass spectrometry (HDXMS) recapitulates the local stability of protein regions reported by CCR-MD, with both approaches consistently indicating that the energy and backbone flexibility changes are solely associated with the region that fold-switches between gsKaiB and fsKaiB and that the localized regions that differentially stabilize gsKaiB also involve regions outside the dimer interface. Moreover, two mutants (R23C and R75C) previously reported to be relevant for altering the rhythmicity of the Kai clock were also studied by HDXMS. Particularly, R75C populates dimeric and monomeric states with a deuterium incorporation profile comparable to the one observed for fsKaiB, emphasizing the importance of the oligomerization state of KaiB for the fold-switch. These findings suggest that the information necessary to control the rhythmicity of the cyanobacterial biological clock is, to a great extent, encoded within the KaiB sequence.


Subject(s)
Circadian Clocks , Cyanobacteria , Bacterial Proteins/metabolism , Circadian Rhythm , Circadian Rhythm Signaling Peptides and Proteins/genetics , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Deuterium , Phosphorylation
15.
Sci Rep ; 12(1): 114, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34997092

ABSTRACT

Microglia are subject to change in tandem with the endogenously generated biological oscillations known as our circadian rhythm. Studies have shown microglia harbor an intrinsic molecular clock which regulates diurnal changes in morphology and influences inflammatory responses. In the adult brain, microglia play an important role in the regulation of condensed extracellular matrix structures called perineuronal nets (PNNs), and it has been suggested that PNNs are also regulated in a circadian and diurnal manner. We sought to determine whether microglia mediate the diurnal regulation of PNNs via CSF1R inhibitor dependent microglial depletion in C57BL/6J mice, and how the absence of microglia might affect cortical diurnal gene expression rhythms. While we observe diurnal differences in microglial morphology, where microglia are most ramified at the onset of the dark phase, we do not find diurnal differences in PNN intensity. However, PNN intensity increases across many brain regions in the absence of microglia, supporting a role for microglia in the regulation of PNNs. Here, we also show that cortical diurnal gene expression rhythms are intact, with no cycling gene changes without microglia. These findings demonstrate a role for microglia in the maintenance of PNNs, but not in the maintenance of diurnal rhythms.


Subject(s)
Brain Waves , Circadian Rhythm , Microglia/pathology , Nerve Net/pathology , Somatosensory Cortex/pathology , Animals , Brain Waves/drug effects , Circadian Rhythm/drug effects , Circadian Rhythm/genetics , Circadian Rhythm Signaling Peptides and Proteins/genetics , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Gene Expression Regulation , Male , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Nerve Net/drug effects , Nerve Net/metabolism , Nerve Net/physiopathology , Organic Chemicals/pharmacology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Somatosensory Cortex/drug effects , Somatosensory Cortex/metabolism , Somatosensory Cortex/physiopathology , Time Factors
16.
PLoS Comput Biol ; 17(12): e1009698, 2021 12.
Article in English | MEDLINE | ID: mdl-34919546

ABSTRACT

We propose a stochastic distributed delay model together with a Markov random field prior and a measurement model for bioluminescence-reporting to analyse spatio-temporal gene expression in intact networks of cells. The model describes the oscillating time evolution of molecular mRNA counts through a negative transcriptional-translational feedback loop encoded in a chemical Langevin equation with a probabilistic delay distribution. The model is extended spatially by means of a multiplicative random effects model with a first order Markov random field prior distribution. Our methodology effectively separates intrinsic molecular noise, measurement noise, and extrinsic noise and phenotypic variation driving cell heterogeneity, while being amenable to parameter identification and inference. Based on the single-cell model we propose a novel computational stability analysis that allows us to infer two key characteristics, namely the robustness of the oscillations, i.e. whether the reaction network exhibits sustained or damped oscillations, and the profile of the regulation, i.e. whether the inhibition occurs over time in a more distributed versus a more direct manner, which affects the cells' ability to phase-shift to new schedules. We show how insight into the spatio-temporal characteristics of the circadian feedback loop in the suprachiasmatic nucleus (SCN) can be gained by applying the methodology to bioluminescence-reported expression of the circadian core clock gene Cry1 across mouse SCN tissue. We find that while (almost) all SCN neurons exhibit robust cell-autonomous oscillations, the parameters that are associated with the regulatory transcription profile give rise to a spatial division of the tissue between the central region whose oscillations are resilient to perturbation in the sense that they maintain a high degree of synchronicity, and the dorsal region which appears to phase shift in a more diversified way as a response to large perturbations and thus could be more amenable to entrainment.


Subject(s)
Circadian Clocks/genetics , Circadian Rhythm Signaling Peptides and Proteins , Gene Expression Regulation/genetics , Models, Biological , Transcription, Genetic/genetics , Animals , Circadian Rhythm Signaling Peptides and Proteins/genetics , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Cryptochromes/genetics , Cryptochromes/metabolism , Mice , Phenotype , Single-Cell Analysis , Stochastic Processes , Suprachiasmatic Nucleus/cytology , Suprachiasmatic Nucleus/metabolism
17.
STAR Protoc ; 2(4): 100958, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34841277

ABSTRACT

Our backward simulation (BS) is an approach to infer the dynamics of individual components in ordinary differential equation (ODE) models, given the information on relatively downstream components or their sums. Here, we demonstrate the use of BS to infer protein synthesis rates with a given profile of protein concentrations over time in a circadian system. This protocol can also be applied to a wide range of problems with undetermined dynamics at the upstream levels. For complete details on the use and execution of this protocol, please refer to Lim et al. (2021).


Subject(s)
Computer Simulation , Models, Biological , Systems Biology/methods , Circadian Rhythm Signaling Peptides and Proteins/genetics , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Kinetics
18.
Oxid Med Cell Longev ; 2021: 4044606, 2021.
Article in English | MEDLINE | ID: mdl-34697563

ABSTRACT

Hepatocellular carcinoma (HCC) is among the most common and lethal form of cancer worldwide. However, its diagnosis and treatment are still dissatisfactory, due to limitations in the understanding of its pathogenic mechanism. Therefore, it is important to elucidate the molecular mechanisms and identify novel therapeutic targets for HCC. Circadian rhythm-related genes control a variety of biological processes. These genes play pivotal roles in the initiation and progression of HCC and are potential diagnostic markers and therapeutic targets. This review gives an update on the research progress of circadian rhythms, their effects on the initiation, progression, and prognosis of HCC, in a bid to provide new insights for the research and treatment of HCC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/metabolism , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Circadian Rhythm , Liver Neoplasms/metabolism , Animals , Antineoplastic Agents/administration & dosage , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Circadian Rhythm Signaling Peptides and Proteins/genetics , Drug Chronotherapy , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Prognosis , Signal Transduction , Time Factors
19.
Science ; 374(6564): eabd4453, 2021 Oct 08.
Article in English | MEDLINE | ID: mdl-34618577

ABSTRACT

Circadian clocks control gene expression to provide an internal representation of local time. We report reconstitution of a complete cyanobacterial circadian clock in vitro, including the central oscillator, signal transduction pathways, downstream transcription factor, and promoter DNA. The entire system oscillates autonomously and remains phase coherent for many days with a fluorescence-based readout that enables real-time observation of each component simultaneously without user intervention. We identified the molecular basis for loss of cycling in an arrhythmic mutant and explored fundamental mechanisms of timekeeping in the cyanobacterial clock. We find that SasA, a circadian sensor histidine kinase associated with clock output, engages directly with KaiB on the KaiC hexamer to regulate period and amplitude of the central oscillator. SasA uses structural mimicry to cooperatively recruit the rare, fold-switched conformation of KaiB to the KaiC hexamer to form the nighttime repressive complex and enhance rhythmicity of the oscillator, particularly under limiting concentrations of KaiB. Thus, the expanded in vitro clock reveals previously unknown mechanisms by which the circadian system of cyanobacteria maintains the pace and rhythmicity under variable protein concentrations.


Subject(s)
Bacterial Proteins/metabolism , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Circadian Rhythm/physiology , Phosphotransferases/metabolism , Synechococcus/physiology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Circadian Rhythm/genetics , Circadian Rhythm Signaling Peptides and Proteins/chemistry , Circadian Rhythm Signaling Peptides and Proteins/genetics , Gene Expression Regulation, Bacterial , Molecular Mimicry , Mutation , Phosphotransferases/chemistry , Phosphotransferases/genetics , Promoter Regions, Genetic , Protein Domains , Protein Folding , Protein Kinases/metabolism , Protein Multimerization , Synechococcus/genetics , Synechococcus/metabolism , Transcription, Genetic
20.
Nat Immunol ; 22(11): 1375-1381, 2021 11.
Article in English | MEDLINE | ID: mdl-34663979

ABSTRACT

Migration of leukocytes from the skin to lymph nodes (LNs) via afferent lymphatic vessels (LVs) is pivotal for adaptive immune responses1,2. Circadian rhythms have emerged as important regulators of leukocyte trafficking to LNs via the blood3,4. Here, we demonstrate that dendritic cells (DCs) have a circadian migration pattern into LVs, which peaks during the rest phase in mice. This migration pattern is determined by rhythmic gradients in the expression of the chemokine CCL21 and of adhesion molecules in both mice and humans. Chronopharmacological targeting of the involved factors abrogates circadian migration of DCs. We identify cell-intrinsic circadian oscillations in skin lymphatic endothelial cells (LECs) and DCs that cogovern these rhythms, as their genetic disruption in either cell type ablates circadian trafficking. These observations indicate that circadian clocks control the infiltration of DCs into skin lymphatics, a process that is essential for many adaptive immune responses and relevant for vaccination and immunotherapies.


Subject(s)
Adaptive Immunity , Chemotaxis , Circadian Clocks , Dendritic Cells/immunology , Lymph Nodes/immunology , Lymphatic Vessels/immunology , Skin/immunology , Aged , Animals , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cells, Cultured , Chemokine CCL21/genetics , Chemokine CCL21/metabolism , Circadian Rhythm Signaling Peptides and Proteins/genetics , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Dendritic Cells/metabolism , Female , Humans , Lymph Nodes/metabolism , Lymphatic Vessels/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Skin/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...