Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
Add more filters

Publication year range
1.
Cell ; 177(6): 1436-1447.e12, 2019 05 30.
Article in English | MEDLINE | ID: mdl-31150620

ABSTRACT

Circadian rhythms control organismal physiology throughout the day. At the cellular level, clock regulation is established by a self-sustained Bmal1-dependent transcriptional oscillator network. However, it is still unclear how different tissues achieve a synchronized rhythmic physiology. That is, do they respond independently to environmental signals, or require interactions with each other to do so? We show that unexpectedly, light synchronizes the Bmal1-dependent circadian machinery in single tissues in the absence of Bmal1 in all other tissues. Strikingly, light-driven tissue autonomous clocks occur without rhythmic feeding behavior and are lost in constant darkness. Importantly, tissue-autonomous Bmal1 partially sustains homeostasis in otherwise arrhythmic and prematurely aging animals. Our results therefore support a two-branched model for the daily synchronization of tissues: an autonomous response branch, whereby light entrains circadian clocks without any commitment of other Bmal1-dependent clocks, and a memory branch using other Bmal1-dependent clocks to "remember" time in the absence of external cues.


Subject(s)
ARNTL Transcription Factors/physiology , Circadian Clocks/genetics , ARNTL Transcription Factors/metabolism , Animals , CLOCK Proteins/metabolism , Circadian Clocks/physiology , Circadian Rhythm/genetics , Feeding Behavior/physiology , Female , Homeostasis , Light , Male , Mice , Mice, Knockout , Models, Animal , Organ Specificity/physiology , Photoperiod , Suprachiasmatic Nucleus/metabolism
2.
Cell ; 177(6): 1448-1462.e14, 2019 05 30.
Article in English | MEDLINE | ID: mdl-31150621

ABSTRACT

Mammals rely on a network of circadian clocks to control daily systemic metabolism and physiology. The central pacemaker in the suprachiasmatic nucleus (SCN) is considered hierarchically dominant over peripheral clocks, whose degree of independence, or tissue-level autonomy, has never been ascertained in vivo. Using arrhythmic Bmal1-null mice, we generated animals with reconstituted circadian expression of BMAL1 exclusively in the liver (Liver-RE). High-throughput transcriptomics and metabolomics show that the liver has independent circadian functions specific for metabolic processes such as the NAD+ salvage pathway and glycogen turnover. However, although BMAL1 occupies chromatin at most genomic targets in Liver-RE mice, circadian expression is restricted to ∼10% of normally rhythmic transcripts. Finally, rhythmic clock gene expression is lost in Liver-RE mice under constant darkness. Hence, full circadian function in the liver depends on signals emanating from other clocks, and light contributes to tissue-autonomous clock function.


Subject(s)
ARNTL Transcription Factors/physiology , Circadian Clocks/genetics , Liver/metabolism , ARNTL Transcription Factors/metabolism , Animals , CLOCK Proteins/metabolism , Circadian Clocks/physiology , Circadian Rhythm/genetics , Female , Gene Expression Regulation , Homeostasis , Light , Male , Mice , Mice, Knockout , Models, Animal , Organ Specificity/physiology , Photoperiod , Suprachiasmatic Nucleus/metabolism
3.
Genes Dev ; 34(15-16): 1089-1105, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32616519

ABSTRACT

The circadian clock is encoded by a negative transcriptional feedback loop that coordinates physiology and behavior through molecular programs that remain incompletely understood. Here, we reveal rhythmic genome-wide alternative splicing (AS) of pre-mRNAs encoding regulators of peptidergic secretion within pancreatic ß cells that are perturbed in Clock-/- and Bmal1-/- ß-cell lines. We show that the RNA-binding protein THRAP3 (thyroid hormone receptor-associated protein 3) regulates circadian clock-dependent AS by binding to exons at coding sequences flanking exons that are more frequently skipped in clock mutant ß cells, including transcripts encoding Cask (calcium/calmodulin-dependent serine protein kinase) and Madd (MAP kinase-activating death domain). Depletion of THRAP3 restores expression of the long isoforms of Cask and Madd, and mimicking exon skipping in these transcripts through antisense oligonucleotide delivery in wild-type islets reduces glucose-stimulated insulin secretion. Finally, we identify shared networks of alternatively spliced exocytic genes from islets of rodent models of diet-induced obesity that significantly overlap with clock mutants. Our results establish a role for pre-mRNA alternative splicing in ß-cell function across the sleep/wake cycle.


Subject(s)
Alternative Splicing , Circadian Clocks/genetics , Exocytosis , Glucose/metabolism , Insulin Secretion/genetics , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/physiology , Animals , CLOCK Proteins/genetics , CLOCK Proteins/physiology , Cells, Cultured , Death Domain Receptor Signaling Adaptor Proteins/genetics , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Guanylate Kinases/genetics , Guanylate Kinases/metabolism , Homeostasis , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Male , Mice, Inbred C57BL , Nuclear Proteins/physiology , Obesity/genetics , Obesity/metabolism , Synaptosomal-Associated Protein 25/genetics , Synaptosomal-Associated Protein 25/metabolism , Transcription Factors/physiology
4.
Proc Natl Acad Sci U S A ; 119(10): e2200083119, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35238641

ABSTRACT

SignificanceWhile increasing evidence associates the disruption of circadian rhythms with pathologic conditions, including obesity, type 2 diabetes, and nonalcoholic fatty liver diseases (NAFLD), the involved mechanisms are still poorly described. Here, we show that, in both humans and mice, the pathogenesis of NAFLD is associated with the disruption of the circadian clock combined with perturbations of the growth hormone and sex hormone pathways. However, while this condition protects mice from the development of fibrosis and insulin resistance, it correlates with increased fibrosis in humans. This suggests that the perturbation of the circadian clock and its associated disruption of the growth hormone and sex hormone pathways are critical for the pathogenesis of metabolic and liver diseases.


Subject(s)
ARNTL Transcription Factors/physiology , Circadian Clocks , Insulin Resistance , Non-alcoholic Fatty Liver Disease/etiology , ARNTL Transcription Factors/genetics , Animals , Diet, High-Fat , Gene Deletion , Gene Expression Regulation , Humans , Leptin/genetics , Lipid Metabolism/genetics , Male , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/genetics , Obesity/genetics
5.
Proc Natl Acad Sci U S A ; 118(35)2021 08 31.
Article in English | MEDLINE | ID: mdl-34426495

ABSTRACT

Exercise and circadian biology are closely intertwined with physiology and metabolism, yet the functional interaction between circadian clocks and exercise capacity is only partially characterized. Here, we tested different clock mutant mouse models to examine the effect of the circadian clock and clock proteins, namely PERIODs and BMAL1, on exercise capacity. We found that daytime variance in endurance exercise capacity is circadian clock controlled. Unlike wild-type mice, which outperform in the late compared with the early part of their active phase, PERIODs- and BMAL1-null mice do not show daytime variance in exercise capacity. It appears that BMAL1 impairs and PERIODs enhance exercise capacity in a daytime-dependent manner. An analysis of liver and muscle glycogen stores as well as muscle lipid utilization suggested that these daytime effects mostly relate to liver glycogen levels and correspond to the animals' feeding behavior. Furthermore, given that exercise capacity responds to training, we tested the effect of training at different times of the day and found that training in the late compared with the early part of the active phase improves exercise performance. Overall, our findings suggest that clock proteins shape exercise capacity in a daytime-dependent manner through changes in liver glycogen levels, likely due to their effect on animals' feeding behavior.


Subject(s)
CLOCK Proteins/physiology , Exercise Tolerance/physiology , Physical Conditioning, Animal/physiology , ARNTL Transcription Factors/physiology , Animals , CLOCK Proteins/genetics , Feeding Behavior , Female , Light , Liver Glycogen/metabolism , Male , Mice , Mice, Inbred C57BL , Muscles/metabolism , Mutation , Period Circadian Proteins/physiology , Photoperiod , Sex Characteristics , Time Factors
6.
J Neurosci ; 41(3): 502-512, 2021 01 20.
Article in English | MEDLINE | ID: mdl-33234609

ABSTRACT

Circadian (approximately daily) rhythms pervade mammalian behavior. They are generated by cell-autonomous, transcriptional/translational feedback loops (TTFLs), active in all tissues. This distributed clock network is coordinated by the principal circadian pacemaker, the hypothalamic suprachiasmatic nucleus (SCN). Its robust and accurate time-keeping arises from circuit-level interactions that bind its individual cellular clocks into a coherent time-keeper. Cells that express the neuropeptide vasoactive intestinal peptide (VIP) mediate retinal entrainment of the SCN; and in the absence of VIP, or its cognate receptor VPAC2, circadian behavior is compromised because SCN cells cannot synchronize. The contributions to pace-making of other cell types, including VPAC2-expressing target cells of VIP, are, however, not understood. We therefore used intersectional genetics to manipulate the cell-autonomous TTFLs of VPAC2-expressing cells. Measuring circadian behavioral and SCN rhythmicity in these temporally chimeric male mice thus enabled us to determine the contribution of VPAC2-expressing cells (∼35% of SCN cells) to SCN time-keeping. Lengthening of the intrinsic TTFL period of VPAC2 cells by deletion of the CK1εTau allele concomitantly lengthened the period of circadian behavioral rhythms. It also increased the variability of the circadian period of bioluminescent TTFL rhythms in SCN slices recorded ex vivo Abrogation of circadian competence in VPAC2 cells by deletion of Bmal1 severely disrupted circadian behavioral rhythms and compromised TTFL time-keeping in the corresponding SCN slices. Thus, VPAC2-expressing cells are a distinct, functionally powerful subset of the SCN circuit, contributing to computation of ensemble period and maintenance of circadian robustness. These findings extend our understanding of SCN circuit topology.


Subject(s)
Behavior, Animal/physiology , Circadian Rhythm/physiology , Periodicity , Receptors, Vasoactive Intestinal Peptide, Type II/physiology , Receptors, Vasoactive Intestinal Peptide/physiology , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/physiology , Animals , Circadian Rhythm/genetics , Feedback, Physiological , Male , Mice , Mice, Knockout , Motor Activity/physiology , Mutant Chimeric Proteins/genetics , Receptors, Vasoactive Intestinal Peptide/genetics , Receptors, Vasoactive Intestinal Peptide, Type II/genetics , Suprachiasmatic Nucleus/physiology
7.
FASEB J ; 35(5): e21530, 2021 05.
Article in English | MEDLINE | ID: mdl-33813752

ABSTRACT

Circadian clock is involved in regulating most renal physiological functions, including water and electrolyte balance and blood pressure homeostasis, however, the role of circadian clock in renal pathophysiology remains largely unknown. Here we aimed to investigate the role of Bmal1, a core clock component, in the development of renal fibrosis, the hallmark of pathological features in many renal diseases. The inducible Bmal1 knockout mice (iKO) whose gene deletion occurred in adulthood were used in the study. Analysis of the urinary water, sodium and potassium excretion showed that the iKO mice exhibit abolished diurnal variations. In the model of renal fibrosis induced by unilateral ureteral obstruction, the iKO mice displayed significantly decreased tubulointerstitial fibrosis reflected by attenuated collagen deposition and mitigated expression of fibrotic markers α-SMA and fibronectin. The hedgehog pathway transcriptional effectors Gli1 and Gli2, which have been reported to be involved in the pathogenesis of renal fibrosis, were significantly decreased in the iKO mice. Mechanistically, ChIP assay and luciferase reporter assay revealed that BMAL1 bound to the promoter of and activate the transcription of Gli2, but not Gli1, suggesting that the involvement of Bmal1 in renal fibrosis was possibly mediated via Gli2-dependent mechanisms. Furthermore, treatment with TGF-ß increased Bmal1 in cultured murine proximal tubular cells. Knockdown of Bmal1 abolished, while overexpression of Bmal1 increased, Gli2 and the expression of fibrosis-related genes. Collectively, these results revealed a prominent role of the core clock gene Bmal1 in tubulointerstitial fibrosis. Moreover, we identified Gli2 as a novel target of Bmal1, which may mediate the adverse effect of Bmal1 in obstructive nephropathy.


Subject(s)
ARNTL Transcription Factors/physiology , Fibrosis/prevention & control , Gene Expression Regulation , Kidney Diseases/prevention & control , Period Circadian Proteins/physiology , Zinc Finger Protein Gli2/antagonists & inhibitors , Animals , Animals, Newborn , Fibrosis/etiology , Fibrosis/metabolism , Fibrosis/pathology , Kidney Diseases/etiology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Zinc Finger Protein Gli2/genetics , Zinc Finger Protein Gli2/metabolism
8.
Acta Pharmacol Sin ; 43(2): 316-329, 2022 Feb.
Article in English | MEDLINE | ID: mdl-33850278

ABSTRACT

Hepatic stellate cells (HSCs) play an important role in the initiation and development of liver fibrogenesis, and abnormal glucose metabolism is increasingly being considered a crucial factor controlling phenotypic transformation in HSCs. However, the role of the factors affecting glycolysis in HSCs in the experimental models of liver fibrosis has not been completely elucidated. In this study, we showed that glycolysis was significantly enhanced, while the expression of brain and muscle arnt-like protein-1 (Bmal1) was downregulated in fibrotic liver tissues of mice, primary HSCs, and transforming growth factor-ß1 (TGF-ß1)-induced LX2 cells. Overexpression of Bmal1 in TGF-ß1-induced LX2 cells blocked glycolysis and inhibited the proliferation and phenotypic transformation of activated HSCs. We further confirmed the protective effect of Bmal1 in liver fibrosis by overexpressing Bmal1 from hepatic adeno-associated virus 8 in mice. In addition, we also showed that the regulation of glycolysis by Bmal1 is mediated by the isocitrate dehydrogenase 1/α-ketoglutarate (IDH1/α-KG) pathway. Collectively, our results indicated that a novel Bmal1-IDH1/α-KG axis may be involved in regulating glycolysis of activated HSCs and might hence be used as a therapeutic target for alleviating liver fibrosis.


Subject(s)
ARNTL Transcription Factors/metabolism , Glycolysis , Hepatic Stellate Cells/metabolism , Isocitrate Dehydrogenase/metabolism , Liver Cirrhosis/metabolism , ARNTL Transcription Factors/physiology , Animals , Blotting, Western , Chromatography, High Pressure Liquid , Flow Cytometry , Hepatic Stellate Cells/pathology , Liver Cirrhosis/physiopathology , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL
9.
Am J Physiol Endocrinol Metab ; 320(4): E747-E759, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33554778

ABSTRACT

Prostaglandin G/H synthase 2 (PTGS2) is a rate-limiting enzyme in prostaglandin synthesis. The present study assessed the role of the uterine circadian clock on Ptgs2 transcription in response to steroid hormones during early pregnancy. We demonstrated that the core clock genes (Bmal1, Per2, Nr1d1, and Dbp), Vegf, and Ptgs2, and their encoded proteins, have rhythmic expression in the mouse uterus from days 3.5 to 4.5 (D3.5-4.5) of pregnancy. Progesterone (P4) treatment of cultured uterus endometrial stromal cells (UESCs) isolated from mPer2Luciferase reporter gene knock-in mice on D4 induced a phase shift in PER2::LUCIFERASE oscillations. This P4-induced phase shift of PER2::LUCIFERASE oscillations was significantly attenuated by the P4 antagonist RU486. Additionally, the amplitude of PER2::LUCIFERASE oscillations was increased by estradiol (E2) treatment in the presence of P4. Consistently, the mRNA levels of clock genes (Bmal1 and Per2), Vegf, and Ptgs2 were markedly increased by E2 treatment of UESCs in the presence of P4. Treatment with E2 also promoted prostaglandin E2 (PGE2) synthesis by UESCs. Depletion of Bmal1 in UESCs by small-interfering RNA (siRNA) decreased the transcript levels of clock genes (Nr1d1 and Dbp), Vegf, and Ptgs2 compared with nonsilencing siRNA treatment. Bmal1 knockdown also inhibited PGE2 synthesis. Moreover, the mRNA expression levels of clock genes (Nr1d1 and Dbp), Vegf, and Ptgs2, and their respective proteins were significantly decreased in the uterus of Bmal1-/- mice. Thus, these data suggest that Bmal1 in mice promotes PGE2 synthesis by upregulating Ptgs2 in response to increases in E2 on D4 of pregnancy.NEW & NOTEWORTHY Rhythmic expression of Bmal1 and Ptgs2 was observed in the uterus isolated from D3.5-4.5 of pregnant mice. E2 increased the expression of Bmal1 and Ptg2 in UESCs isolated from mice on D4. The expression of Ptgs2 was significantly decreased in Bmal1-siRNA treated UESCs. Bmal1 knockdown also inhibited PGE2 synthesis. Thus, these data suggest that Bmal1 in mice promotes PGE2 synthesis by upregulating Ptgs2 in response to increases in E2 on D4 of pregnancy.


Subject(s)
ARNTL Transcription Factors/physiology , Cyclooxygenase 2/genetics , Dinoprostone/biosynthesis , Estradiol/blood , ARNTL Transcription Factors/genetics , Animals , Cells, Cultured , Cyclooxygenase 2/metabolism , Estradiol/pharmacology , Female , Gestational Age , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Progesterone/pharmacology , Transcriptional Activation/drug effects , Uterus/drug effects , Uterus/metabolism
10.
FASEB J ; 34(5): 6613-6627, 2020 05.
Article in English | MEDLINE | ID: mdl-32212194

ABSTRACT

Circadian clock confers temporal control in metabolism, with its disruption leading to the development of insulin resistance. Metabolic substrate utilization in skeletal muscle is coordinated with diurnal nutrient cycles. However, whether the molecular clock is involved in this coordination is largely unknown. Using a myocyte-selective genetic ablation mouse model of the essential clock activator Bmal1, here we identify muscle-intrinsic clock as a sensor of feeding cues to orchestrate skeletal muscle oxidation required for global nutrient flux. Bmal1 in skeletal muscle responds robustly to feeding in vivo and insulin induces its expression. Muscle Bmal1 deficiency impaired the transcriptional control of glucose metabolic pathway, resulting in markedly attenuated glucose utilization and fasting hyperglycemia. Notably, the loss of Bmal1 response to feeding abolished fasting-to-feeding metabolic fuel switch from fatty acids to glucose in skeletal muscle, leading to the activation of energy-sensing pathways for fatty acid oxidation. These altered metabolic substrate oxidations in Bmal1-deficient muscle ultimately depleted circulating lipid levels that prevented hepatic steatosis. Collectively, our findings highlight the key role of the metabolic-sensing function of skeletal muscle clock in partitioning nutrient flux between muscle and liver to maintain whole-body lipid and glucose homeostasis.


Subject(s)
ARNTL Transcription Factors/physiology , Circadian Clocks , Glucose/chemistry , Glucose/metabolism , Homeostasis , Liver/metabolism , Muscle, Skeletal/metabolism , Animals , Fatty Acids/chemistry , Fatty Acids/metabolism , Gene Expression Regulation , Mice , Mice, Transgenic
11.
FASEB J ; 34(5): 6570-6581, 2020 05.
Article in English | MEDLINE | ID: mdl-32246801

ABSTRACT

Dysfunction of the circadian rhythm is one of most common nonmotor symptoms in Parkinson's disease (PD), but the molecular role of the circadian rhythm in PD is unclear. We here showed that inactivation of brain and muscle ARNT-like 1 (BMAL1) in 1-methyl-4-phenyl-1,2,4,5-tetrahydropyridine (MPTP)-treated mice resulted in obvious motor functional deficit, loss of dopaminergic neurons (DANs) in the substantia nigra pars compacta (SNpc), decrease of dopamine (DA) transmitter, and increased activation of microglia and astrocytes in the striatum. Time on the rotarod or calorie consumption, and food and water intake were reduced in the Bmal1-/- mice after MPTP treatment, suggesting that absence of Bmal1 may exacerbate circadian and PD motor function. We observed a significant reduction of DANs (~35%) in the SNpc, the tyrosine hydroxylase protein level in the striatum (~60%), the DA (~22%), and 3,4-dihydroxyphenylacetic acid content (~29%), respectively, in MPTP-treated Bmal1-/- mice. Loss of Bmal1 aggravated the inflammatory reaction both in vivo and in vitro. These findings suggest that BMAL1 may play an essential role in the survival of DANs and maintain normal function of the DA signaling pathway via regulating microglia-mediated neuroinflammation in the brain.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , ARNTL Transcription Factors/physiology , Disease Models, Animal , Dopaminergic Neurons/immunology , Inflammation/pathology , Microglia/pathology , Parkinson Disease/pathology , Animals , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Inflammation/etiology , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/drug effects , Microglia/metabolism , Neurotoxins/toxicity , Parkinson Disease/etiology , Parkinson Disease/metabolism
12.
PLoS Biol ; 16(8): e2005886, 2018 08.
Article in English | MEDLINE | ID: mdl-30096135

ABSTRACT

Circadian clocks are fundamental physiological regulators of energy homeostasis, but direct transcriptional targets of the muscle clock machinery are unknown. To understand how the muscle clock directs rhythmic metabolism, we determined genome-wide binding of the master clock regulators brain and muscle ARNT-like protein 1 (BMAL1) and REV-ERBα in murine muscles. Integrating occupancy with 24-hr gene expression and metabolomics after muscle-specific loss of BMAL1 and REV-ERBα, here we unravel novel molecular mechanisms connecting muscle clock function to daily cycles of lipid and protein metabolism. Validating BMAL1 and REV-ERBα targets using luciferase assays and in vivo rescue, we demonstrate how a major role of the muscle clock is to promote diurnal cycles of neutral lipid storage while coordinately inhibiting lipid and protein catabolism prior to awakening. This occurs by BMAL1-dependent activation of Dgat2 and REV-ERBα-dependent repression of major targets involved in lipid metabolism and protein turnover (MuRF-1, Atrogin-1). Accordingly, muscle-specific loss of BMAL1 is associated with metabolic inefficiency, impaired muscle triglyceride biosynthesis, and accumulation of bioactive lipids and amino acids. Taken together, our data provide a comprehensive overview of how genomic binding of BMAL1 and REV-ERBα is related to temporal changes in gene expression and metabolite fluctuations.


Subject(s)
ARNTL Transcription Factors/physiology , Circadian Clocks/physiology , Muscle, Skeletal/physiology , Amino Acids/metabolism , Amino Acids/physiology , Animals , CLOCK Proteins/genetics , Circadian Rhythm/genetics , Gene Expression , Homeostasis , Humans , Lipid Metabolism/physiology , Lipids , Mice , Mice, Knockout , RNA, Messenger/metabolism
13.
Arterioscler Thromb Vasc Biol ; 40(6): 1523-1532, 2020 06.
Article in English | MEDLINE | ID: mdl-32321308

ABSTRACT

OBJECTIVE: Although the molecular components of circadian rhythms oscillate in discrete cellular components of the vasculature and many aspects of vascular function display diurnal variation, the cellular connections between the molecular clock and inflammatory cardiovascular diseases remain to be elucidated. Previously we have shown that pre- versus postnatal deletion of Bmal1 (brain and muscle aryl hydrocarbon receptor nuclear translocator-like 1), the nonredundant core clock gene has contrasting effects on atherogenesis. Here we investigated the effect of myeloid cell Bmal1 deletion on atherogenesis and abdominal aortic aneurysm formation in mice. Approach and Results: Mice lacking Bmal1 in myeloid cells were generated by crossing Bmal1 flox/flox mice with lysozyme 2 promoter-driven Cre recombinase mice on a hyperlipidemic low-density lipoprotein receptor-deficient background and were fed on a high-fat diet to induce atherosclerosis. Atherogenesis was restrained, concomitant with a reduction of aortic proinflammatory gene expression in myeloid cell Bmal1 knockout mice. Body weight, blood pressure, blood glucose, triglycerides, and cholesterol were unaltered. Similarly, myeloid cell depletion of Bmal1 also restrained Ang II (angiotensin II) induced formation of abdominal aortic aneurysm in hyperlipidemic mice. In vitro, RNA-Seq analysis demonstrated a proinflammatory response in cultured macrophages in which there was overexpression of Bmal1. CONCLUSIONS: Myeloid cell Bmal1 deletion retards atherogenesis and restrains the formation of abdominal aortic aneurysm and may represent a potential therapeutic target for inflammatory cardiovascular diseases.


Subject(s)
ARNTL Transcription Factors/deficiency , ARNTL Transcription Factors/physiology , Aortic Aneurysm, Abdominal/prevention & control , Atherosclerosis/prevention & control , Hyperlipidemias/complications , Myeloid Cells/chemistry , ARNTL Transcription Factors/genetics , Angiotensin II/pharmacology , Animals , Aortic Aneurysm, Abdominal/chemically induced , Atherosclerosis/etiology , Atherosclerosis/pathology , Cells, Cultured , Crosses, Genetic , Diet, High-Fat , Gene Deletion , Gene Expression , Hyperlipidemias/etiology , Inflammation , Integrases/genetics , Macrophages, Peritoneal/chemistry , Macrophages, Peritoneal/physiology , Mice , Mice, Knockout , Muramidase/genetics , Promoter Regions, Genetic/genetics , Receptors, LDL/deficiency , Receptors, LDL/genetics
14.
PLoS Genet ; 14(1): e1007156, 2018 01.
Article in English | MEDLINE | ID: mdl-29300726

ABSTRACT

The mammalian circadian clock relies on the transcription factor CLOCK:BMAL1 to coordinate the rhythmic expression of 15% of the transcriptome and control the daily regulation of biological functions. The recent characterization of CLOCK:BMAL1 cistrome revealed that although CLOCK:BMAL1 binds synchronously to all of its target genes, its transcriptional output is highly heterogeneous. By performing a meta-analysis of several independent genome-wide datasets, we found that the binding of other transcription factors at CLOCK:BMAL1 enhancers likely contribute to the heterogeneity of CLOCK:BMAL1 transcriptional output. While CLOCK:BMAL1 rhythmic DNA binding promotes rhythmic nucleosome removal, it is not sufficient to generate transcriptionally active enhancers as assessed by H3K27ac signal, RNA Polymerase II recruitment, and eRNA expression. Instead, the transcriptional activity of CLOCK:BMAL1 enhancers appears to rely on the activity of ubiquitously expressed transcription factors, and not tissue-specific transcription factors, recruited at nearby binding sites. The contribution of other transcription factors is exemplified by how fasting, which effects several transcription factors but not CLOCK:BMAL1, either decreases or increases the amplitude of many rhythmically expressed CLOCK:BMAL1 target genes. Together, our analysis suggests that CLOCK:BMAL1 promotes a transcriptionally permissive chromatin landscape that primes its target genes for transcription activation rather than directly activating transcription, and provides a new framework to explain how environmental or pathological conditions can reprogram the rhythmic expression of clock-controlled genes.


Subject(s)
ARNTL Transcription Factors/physiology , CLOCK Proteins/physiology , Circadian Clocks/genetics , Gene Expression Regulation , Transcription, Genetic , ARNTL Transcription Factors/metabolism , Animals , Binding Sites/genetics , CLOCK Proteins/metabolism , Circadian Rhythm/genetics , Enhancer Elements, Genetic/genetics , Mice , Protein Binding
15.
Proc Natl Acad Sci U S A ; 115(51): 13099-13104, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30498030

ABSTRACT

The mammalian retina contains an autonomous circadian clock system that controls many physiological functions within this tissue. Previous studies on young mice have reported that removal of the key circadian clock gene Bmal1 from the retina affects the circadian regulation of visual function, but does not affect photoreceptor viability. Because dysfunction in the circadian system is known to affect cell viability during aging in other systems, we compared the effect of Bmal1 removal from the retina on visual function, inner retinal structure, and photoreceptor viability in young (1 to 3 months) and aged (24 to 26 months) mice. We found that removal of Bmal1 from the retina significantly affects visual information processing in both rod and cone pathways, reduces the thickness of inner retinal nuclear and plexiform layers, accelerates the decline of visual functions during aging, and reduces the viability of cone photoreceptors. Our results thus suggest that circadian clock dysfunction, caused by genetic or other means, may contribute to the decline of visual function during development and aging.


Subject(s)
ARNTL Transcription Factors/physiology , Aging/pathology , Circadian Rhythm , Retina/pathology , Retinal Cone Photoreceptor Cells/pathology , Vision, Ocular , Aging/metabolism , Animals , Circadian Clocks , Gene Expression Regulation , Mice , Mice, Inbred C57BL , Mice, Knockout , Retina/metabolism , Retinal Cone Photoreceptor Cells/metabolism
16.
Reproduction ; 160(6): 955-967, 2020 12.
Article in English | MEDLINE | ID: mdl-33112769

ABSTRACT

Luteinization is the event of corpus luteum formation, a way of follicle cells transformation and a process of steroidogenesis alteration. As the core clock gene, Bmal1 was involved in the regulation of ovulation process and luteal function afterwards. Till now, the underlying roles of luteinization played by Bmal1 remain unknown. To explore the unique role of Bmal1 in luteal steroidogenesis and its underlying pathway, we investigated the luteal hormone synthesis profile in Bmal1 knockout female mice. We found that luteal hormone synthesis was notably impaired, and phosphorylation of PI3K/NfκB pathway was significantly activated. Then, the results were verified in in vitro cultured cells, including isolated Bmal1 interference granulosa cells (GCs) and theca cells (TCs), respectively. Hormones levels of supernatant culture media and mRNA expressions of steroidogenesis-associated genes (star, Hsd3ß2, cyp19a1 in GCs, Lhcgr, star, Hsd3ß2, cyp17a1 in TCs) were mutually decreased, while the phosphorylation of PI3K/NfκB was promoted during in vitro luteinization. After PI3K specific-inhibitor LY294002 intervention, mRNA expressions of Lhcgr and Hsd3ß2 were partially rescued in Bmal1 interference TCs, together with significantly increased androstenedione and T synthesis. Further exploration in TCs demonstrated BMAL1 interacted directly but negatively with NfκB p65 (RelA), a subunit which was supposed as a mediator in Bmal1-governed PI3K signaling regulation. Taken together, we verified the novel role of Bmal1 in luteal steroidogenesis, achieving by negative interplay with RelA-mediated PI3K/NfκB pathway.


Subject(s)
ARNTL Transcription Factors/physiology , Gonadal Steroid Hormones/biosynthesis , Granulosa Cells/metabolism , Luteinization , Ovarian Follicle/metabolism , Theca Cells/metabolism , Androstenedione/biosynthesis , Animals , Estradiol/biosynthesis , Female , Granulosa Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovarian Follicle/pathology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Progesterone/biosynthesis , Testosterone/biosynthesis , Theca Cells/pathology , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism
17.
Int J Mol Sci ; 21(3)2020 Jan 30.
Article in English | MEDLINE | ID: mdl-32019183

ABSTRACT

The circadian rhythm regulates the physiology and behavior of living organisms in a time-dependent manner. Clock genes have distinct roles including the control over gene expression mediated by the transcriptional activators CLOCK and BMAL1, and the suppression of gene expression mediated by the transcriptional repressors PER1/2 and CRY1/2. The balance between gene expression and repression is key to the maintenance of tissue homeostasis that is disrupted in the event of an injury. In the skin, a compromised epithelial barrier triggers a cascade of events that culminate in the mobilization of epithelial cells and stem cells. Recruited epithelial cells migrate towards the wound and reestablish the protective epithelial layer of the skin. Although we have recently demonstrated the involvement of BMAL and the PI3K signaling in wound healing, the role of the circadian clock genes in tissue repair remains poorly understood. Here, we sought to understand the role of BMAL1 on skin healing in response to injury. We found that genetic depletion of BMAL1 resulted in delayed healing of the skin as compared to wild-type control mice. Furthermore, we found that loss of Bmal1 was associated with the accumulation of Reactive Oxygen Species Modulator 1 (ROMO1), a protein responsible for inducing the production of intracellular reactive oxygen species (ROS). The slow healing was associated with ROS and superoxide dismutase (SOD) production, and pharmacological inhibition of the oxidative stress signaling (ROS/SOD) led to cellular proliferation, upregulation of Sirtuin 1 (SIRT1), and rescued the skin healing phenotype of Bmal1-/- mice. Overall, our study points to BMAL1 as a key player in tissue regeneration and as a critical regulator of ROMO1 and oxidative stress in the skin.


Subject(s)
ARNTL Transcription Factors/physiology , Antioxidants/pharmacology , Epidermis/physiology , Gene Expression Regulation , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Wound Healing/drug effects , Animals , Circadian Rhythm , Epidermis/drug effects , Male , Membrane Proteins/metabolism , Mice , Mice, Knockout , Mitochondrial Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism
18.
Nucleic Acids Res ; 45(11): 6459-6470, 2017 Jun 20.
Article in English | MEDLINE | ID: mdl-28407113

ABSTRACT

Circadian clocks are autonomous daily timekeeping mechanisms that allow organisms to adapt to environmental rhythms as well as temporally organize biological functions. Clock-controlled timekeeping involves extensive regulation of rhythmic gene expression. To date, relatively few clock-associated promoter elements have been identified and characterized. In an unbiased search of core clock gene promoters from 12 species of Drosophila, we discovered a 29-bp consensus sequence that we designated as the Clock-Associated Transcriptional Activation Cassette or 'CATAC'. To experimentally address the spatiotemporal expression information associated with this element, we generated constructs with four separate native CATAC elements upstream of a basal promoter driving expression of either the yeast Gal4 or firefly luciferase reporter genes. Reporter assays showed that presence of wild-type, but not mutated CATAC elements, imparted increased expression levels as well as rhythmic regulation. Part of the CATAC consensus sequence resembles the E-box binding site for the core circadian transcription factor CLOCK/CYCLE (CLK/CYC), and CATAC-mediated expression rhythms are lost in the presence of null mutations in either cyc or the gene encoding the CLK/CYC inhibitor, period (per). Nevertheless, our results indicate that CATAC's enhancer function persists in the absence of CLK/CYC. Thus, CATAC represents a novel cis-regulatory element encoding clock-controlled regulation.


Subject(s)
Drosophila melanogaster/genetics , Promoter Regions, Genetic , ARNTL Transcription Factors/physiology , Animals , Base Sequence , CLOCK Proteins/physiology , Circadian Rhythm , Consensus Sequence , Drosophila Proteins/physiology , Drosophila melanogaster/metabolism , Gene Expression , Gene Expression Regulation , Genes, Reporter , Male , Sequence Analysis, DNA
19.
Yi Chuan ; 41(6): 524-533, 2019 Jun 20.
Article in Zh | MEDLINE | ID: mdl-31257200

ABSTRACT

Normal development of the cerebral cortex is a basis for the formation and function of mammalian brains. During this process, the radial migration of cortical neurons, as well as the axon projection into specific layers, are the most important steps regulated by some transcription factors, but the underlying molecular mechanisms are still obscure. BMAL1 (brain and muscle Arnt-like protein 1) is a newly identified transcription factor that plays important roles in the circadian rhythms. It was recently found to regulate the proliferation of hippocampal neuronal progenitor/precursor cells (NPCs), implicating Bmal1 in the brain development. Here we employed both RT-RCR and real-time PCR to explore the expression pattern of the Bmal1 gene in the developing brain. We found BMAl1 is enriched in the brain cortex during the perinatal stages and peaked in P3 mouse brains. Combined with in utero electroporation and interference with RNAi, we found that reducing the expression level of Bmal1 in neurons, the radial migration of embryonic cortical neurons was largely delayed, in a gene dose-effect pattern. Moreover, reducing the level of Bmal1 expression in mouse brains, the axonal projection in the corpus callosum was also disrupted from ipsilateral to the lateral cerebral hemisphere. These findings indicate that BMAL1 is essential for the radial migration of neurons in the cerebral cortex and the axonal projection of the corpus callosum, providing insights into the molecular mechanisms of cerebral cortex development.


Subject(s)
ARNTL Transcription Factors/physiology , Axons , Cell Movement , Cerebral Cortex/embryology , Neurogenesis , Animals , Female , Mice , Pregnancy
20.
Am J Pathol ; 187(6): 1426-1435, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28432873

ABSTRACT

The brain and muscle aryl hydrocarbon receptor nuclear translocator-like protein (BMAL)-1 constitutes a major transcriptional regulator of the circadian clock. Here, we explored the impact of conditional deletion of Bmal1 in endothelium and hematopoietic cells in murine models of microvascular and macrovascular injury. We used two models of Bmal1fx/fx;Tek-Cre mice, a retinal ischemia/reperfusion model and a neointimal hyperplasia model of the femoral artery. Eyes were enumerated for acellular capillaries and were stained for oxidative damage markers using nitrotyrosine immunohistochemistry. LSK (lineage-negative, stem cell antigen-1-positive, c-Kit-positive) cells were quantified and proliferation assessed. Hematopoiesis is influenced by innervation to the bone marrow, which we assessed using IHC analysis. The number of acellular capillaries increased threefold, and nitrotyrosine staining increased 1.5-fold, in the retinas of Bmal1fx/fx;Tek-Cre mice. The number of LSK cells from the Bmal1fx/fx;Tek-Cre mice decreased by 1.5-fold and was accompanied by a profound decrease in proliferative potential. Bmal1fx/fx;Tek-Cre mice also exhibited evidence of bone marrow denervation, demonstrating a loss of neurofilament-200 staining. Injured femoral arteries showed a 20% increase in neointimal hyperplasia compared with similarly injured wild-type controls. Our study highlights the importance of the circadian clock in maintaining vascular homeostasis and demonstrates that specific deletion of BMAL1 in endothelial and hematopoietic cells results in phenotypic features similar to those of diabetes.


Subject(s)
ARNTL Transcription Factors/physiology , Neointima/pathology , Reperfusion Injury/metabolism , Retinal Vessels/metabolism , ARNTL Transcription Factors/deficiency , ARNTL Transcription Factors/genetics , Animals , Bone Marrow/metabolism , Bone Marrow/pathology , Capillaries/pathology , Cell Proliferation , Circadian Rhythm/physiology , Disease Models, Animal , Endothelial Cells/metabolism , Femoral Artery/injuries , Femoral Artery/pathology , Gene Deletion , Hematopoietic Stem Cells/pathology , Hyperplasia , Leukocyte Common Antigens/analysis , Leukocyte Count , Mice, Transgenic , Nitric Oxide Synthase Type III/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reperfusion Injury/pathology , Retina/metabolism , Retinal Vessels/pathology
SELECTION OF CITATIONS
SEARCH DETAIL