Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
ACS Nano ; 18(6): 4822-4839, 2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38285698

ABSTRACT

Efficiently delivering exogenous materials into primary neurons and neural stem cells (NSCs) has long been a challenge in neurobiology. Existing methods have struggled with complex protocols, unreliable reproducibility, high immunogenicity, and cytotoxicity, causing a huge conundrum and hindering in-depth analyses. Here, we establish a cutting-edge method for transfecting primary neurons and NSCs, named teleofection, by a two-step process to enhance the formation of biocompatible calcium phosphate (CaP) nanoparticles. Teleofection enables both nucleic acid and protein transfection into primary neurons and NSCs, eliminating the need for specialized skills and equipment. It can easily fine-tune transfection efficiency by adjusting the incubation time and nanoparticle quantity, catering to various experimental requirements. Teleofection's versatility allows for the delivery of different cargos into the same cell culture, whether simultaneously or sequentially. This flexibility proves invaluable for long-term studies, enabling the monitoring of neural development and synapse plasticity. Moreover, teleofection ensures the consistent and robust expression of delivered genes, facilitating molecular and biochemical investigations. Teleofection represents a significant advancement in neurobiology, which has promise to transcend the limitations of current gene delivery methods. It offers a user-friendly, cost-effective, and reproducible approach for researchers, potentially revolutionizing our understanding of brain function and development.


Subject(s)
Nanoparticles , Neural Stem Cells , Nucleic Acids , Nucleic Acids/metabolism , Reproducibility of Results , Neural Stem Cells/metabolism , Nanoparticles/chemistry , Transfection , Calcium Phosphates/chemistry
2.
iScience ; 26(2): 106002, 2023 Feb 17.
Article in English | MEDLINE | ID: mdl-36866044

ABSTRACT

Timed daily access to a running-wheel (scheduled voluntary exercise; SVE) synchronizes rodent circadian rhythms and promotes stable, 24h rhythms in animals with genetically targeted impairment of neuropeptide signaling (Vipr2 -/- mice). Here we used RNA-seq and/or qRT-PCR to assess how this neuropeptide signaling impairment as well as SVE shapes molecular programs in the brain clock (suprachiasmatic nuclei; SCN) and peripheral tissues (liver and lung). Compared to Vipr2 +/+ animals, the SCN transcriptome of Vipr2 -/- mice showed extensive dysregulation which included core clock components, transcription factors, and neurochemicals. Furthermore, although SVE stabilized behavioral rhythms in these animals, the SCN transcriptome remained dysregulated. The molecular programs in the lung and liver of Vipr2 -/- mice were partially intact, although their response to SVE differed to that of these peripheral tissues in the Vipr2 +/+ mice. These findings highlight that SVE can correct behavioral abnormalities in circadian rhythms without causing large scale alterations to the SCN transcriptome.

3.
J Biol Rhythms ; 37(3): 235-248, 2022 06.
Article in English | MEDLINE | ID: mdl-35382619

ABSTRACT

Methylation, that is, the transfer or synthesis of a -CH3 group onto a target molecule, is a pervasive biochemical modification found in organisms from bacteria to humans. In mammals, a complex metabolic pathway powered by the essential nutrients vitamin B9 and B12, methionine and choline, synthesizes S-adenosylmethionine, the methyl donor in the methylation of nucleic acids, proteins, fatty acids, and small molecules by over 200 substrate-specific methyltransferases described so far in humans. Methylations not only play a key role in scenarios for the origin and evolution of life, but they remain essential for the development and physiology of organisms alive today, and methylation deficiencies contribute to the etiology of many pathologies. The methylation of histones and DNA is important for circadian rhythms in many organisms, and global inhibition of methyl metabolism similarly affects biological rhythms in prokaryotes and eukaryotes. These observations, together with various pieces of evidence scattered in the literature on circadian gene expression and metabolism, indicate a close mutual interdependence between biological rhythms and methyl metabolism that may originate from prebiotic chemistry. This perspective first proposes an abiogenetic scenario for rhythmic methylations and then outlines mammalian methyl metabolism, before reanalyzing previously published data to draw a tentative map of its profound connections with the circadian clock.


Subject(s)
Circadian Rhythm , S-Adenosylmethionine , Animals , Folic Acid/metabolism , Humans , Mammals/metabolism , Methionine/metabolism , Methylation , S-Adenosylmethionine/chemistry , S-Adenosylmethionine/metabolism
4.
Commun Biol ; 5(1): 313, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35383287

ABSTRACT

The global dietary supplement market is valued at over USD 100 billion. One popular dietary supplement, S-adenosylmethionine, is marketed to improve joints, liver health and emotional well-being in the US since 1999, and has been a prescription drug in Europe to treat depression and arthritis since 1975, but recent studies questioned its efficacy. In our body, S-adenosylmethionine is critical for the methylation of nucleic acids, proteins and many other targets. The marketing of SAM implies that more S-adenosylmethionine is better since it would stimulate methylations and improve health. Previously, we have shown that methylation reactions regulate biological rhythms in many organisms. Here, using biological rhythms to assess the effects of exogenous S-adenosylmethionine, we reveal that excess S-adenosylmethionine disrupts rhythms and, rather than promoting methylation, is catabolized to adenine and methylthioadenosine, toxic methylation inhibitors. These findings further our understanding of methyl metabolism and question the safety of S-adenosylmethionine as a supplement.


Subject(s)
Adenine , S-Adenosylmethionine , Dietary Supplements , Liver/metabolism , Methylation , S-Adenosylmethionine/metabolism , S-Adenosylmethionine/pharmacology
5.
Nat Commun ; 12(1): 645, 2021 01 28.
Article in English | MEDLINE | ID: mdl-33510150

ABSTRACT

Hepatocellular carcinoma (HCC) is the most predominant primary malignancy in the liver. Genotoxic and genetic models have revealed that HCC cells are derived from hepatocytes, but where the critical region for tumor foci emergence is and how this transformation occurs are still unclear. Here, hyperpolyploidization of hepatocytes around the centrilobular (CL) region is demonstrated to be closely linked with the development of HCC cells after diethylnitrosamine treatment. We identify the CL region as a dominant lobule for accumulation of hyperpolyploid hepatocytes and preneoplastic tumor foci formation. We also demonstrate that upregulation of Aurkb plays a critical role in promoting hyperpolyploidization. Increase of AURKB phosphorylation is detected on the midbody during cytokinesis, causing abscission failure and hyperpolyploidization. Pharmacological inhibition of AURKB dramatically reduces nucleus size and tumor foci number surrounding the CL region in diethylnitrosamine-treated liver. Our work reveals an intimate molecular link between pathological hyperpolyploidy of CL hepatocytes and transformation into HCC cells.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cell Transformation, Neoplastic/genetics , Hepatocytes/metabolism , Liver Neoplasms/genetics , Liver/metabolism , Polyploidy , Precancerous Conditions/genetics , Animals , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/metabolism , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Diethylnitrosamine/toxicity , Female , Hepatocytes/drug effects , Humans , Liver/drug effects , Liver/pathology , Liver Neoplasms/chemically induced , Liver Neoplasms/metabolism , Male , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Microscopy, Confocal , Precancerous Conditions/chemically induced , Precancerous Conditions/metabolism
6.
Sci Adv ; 6(51)2020 12.
Article in English | MEDLINE | ID: mdl-33328229

ABSTRACT

Circadian gene expression driven by transcription activators CLOCK and BMAL1 is intimately associated with dynamic chromatin remodeling. However, how cellular metabolism directs circadian chromatin remodeling is virtually unexplored. We report that the S-adenosylhomocysteine (SAH) hydrolyzing enzyme adenosylhomocysteinase (AHCY) cyclically associates to CLOCK-BMAL1 at chromatin sites and promotes circadian transcriptional activity. SAH is a potent feedback inhibitor of S-adenosylmethionine (SAM)-dependent methyltransferases, and timely hydrolysis of SAH by AHCY is critical to sustain methylation reactions. We show that AHCY is essential for cyclic H3K4 trimethylation, genome-wide recruitment of BMAL1 to chromatin, and subsequent circadian transcription. Depletion or targeted pharmacological inhibition of AHCY in mammalian cells markedly decreases the amplitude of circadian gene expression. In mice, pharmacological inhibition of AHCY in the hypothalamus alters circadian locomotor activity and rhythmic transcription within the suprachiasmatic nucleus. These results reveal a previously unappreciated connection between cellular metabolism, chromatin dynamics, and circadian regulation.


Subject(s)
Adenosylhomocysteinase , Chromatin Assembly and Disassembly , Circadian Clocks , Methionine , ARNTL Transcription Factors/genetics , Adenosylhomocysteinase/genetics , Adenosylhomocysteinase/metabolism , Animals , CLOCK Proteins , Chromatin , Circadian Rhythm/genetics , Methionine/metabolism , Mice , S-Adenosylhomocysteine/metabolism
8.
Commun Biol ; 3(1): 211, 2020 05 06.
Article in English | MEDLINE | ID: mdl-32376902

ABSTRACT

The methyl cycle is a universal metabolic pathway providing methyl groups for the methylation of nuclei acids and proteins, regulating all aspects of cellular physiology. We have previously shown that methyl cycle inhibition in mammals strongly affects circadian rhythms. Since the methyl cycle and circadian clocks have evolved early during evolution and operate in organisms across the tree of life, we sought to determine whether the link between the two is also conserved. Here, we show that methyl cycle inhibition affects biological rhythms in species ranging from unicellular algae to humans, separated by more than 1 billion years of evolution. In contrast, the cyanobacterial clock is resistant to methyl cycle inhibition, although we demonstrate that methylations themselves regulate circadian rhythms in this organism. Mammalian cells with a rewired bacteria-like methyl cycle are protected, like cyanobacteria, from methyl cycle inhibition, providing interesting new possibilities for the treatment of methylation deficiencies.


Subject(s)
Circadian Rhythm , Methylation , Animals , Arabidopsis/physiology , Caenorhabditis elegans/physiology , Chlamydomonas reinhardtii/physiology , Chlorophyta/physiology , Drosophila melanogaster/physiology , Humans , Mice/physiology , Synechococcus/physiology , Zebrafish/physiology
9.
Curr Opin Pharmacol ; 48: 127-136, 2019 10.
Article in English | MEDLINE | ID: mdl-31600661

ABSTRACT

Organoids, bioprinted mini-tissues and body-on-a-chip technologies are poised to transform the practice of preclinical pharmacology, with a view to achieving better predictive value. We review the need for further refinement in static and dynamic biomechanical aspects of such microenvironments. Further consideration of the developments required in perfusion systems to enable delivery of an appropriate soluble microenvironment are argued. We place particular emphasis on a major deficiency in these systems, being the absence or aberrant circadian behaviour of cells used in such settings, and consider the technical challenges that are needing to be met in order to achieve rhythm-on-a-chip.


Subject(s)
Circadian Rhythm , Pharmacology/methods , Tissue Engineering , Animals , Humans , Organoids , Tissue Scaffolds
10.
Nat Commun ; 10(1): 2563, 2019 06 12.
Article in English | MEDLINE | ID: mdl-31189882

ABSTRACT

Non-coding cis-regulatory elements are essential determinants of development, but their exact impacts on behavior and physiology in adults remain elusive. Cis-element-based transcriptional regulation is believed to be crucial for generating circadian rhythms in behavior and physiology. However, genetic evidence supporting this model is based on mutations in the protein-coding sequences of clock genes. Here, we report generation of mutant mice carrying a mutation only at the E'-box cis-element in the promoter region of the core clock gene Per2. The Per2 E'-box mutation abolishes sustainable molecular clock oscillations and renders circadian locomotor activity and body temperature rhythms unstable. Without the E'-box, Per2 messenger RNA and protein expression remain at mid-to-high levels. Our work delineates the Per2 E'-box as a critical nodal element for keeping sustainable cell-autonomous circadian oscillation and reveals the extent of the impact of the non-coding cis-element in daily maintenance of animal locomotor activity and body temperature rhythmicity.


Subject(s)
Circadian Rhythm/genetics , E-Box Elements/genetics , Period Circadian Proteins/genetics , Promoter Regions, Genetic/genetics , Animals , Behavior, Animal/physiology , Body Temperature/physiology , Cells, Cultured , Fibroblasts , Locomotion/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Primary Cell Culture , RNA, Messenger/metabolism
11.
Proc Natl Acad Sci U S A ; 115(23): 5980-5985, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29784786

ABSTRACT

The N6-methylation of internal adenosines (m6A) in mRNA has been quantified and localized throughout the transcriptome. However, the physiological significance of m6A in most highly methylated mRNAs is unknown. It was demonstrated previously that the circadian clock, based on transcription-translation negative feedback loops, is sensitive to the general inhibition of m6A. Here, we show that the Casein Kinase 1 Delta mRNA (Ck1δ), coding for a critical kinase in the control of circadian rhythms, cellular growth, and survival, is negatively regulated by m6A. Inhibition of Ck1δ mRNA methylation leads to increased translation of two alternatively spliced CK1δ isoforms, CK1δ1 and CK1δ2, uncharacterized until now. The expression ratio between these isoforms is tissue-specific, CK1δ1 and CK1δ2 have different kinase activities, and they cooperate in the phosphorylation of the circadian clock protein PER2. While CK1δ1 accelerates the circadian clock by promoting the decay of PER2 proteins, CK1δ2 slows it down by stabilizing PER2 via increased phosphorylation at a key residue on PER2 protein. These observations challenge the previously established model of PER2 phosphorylation and, given the multiple functions and targets of CK1δ, the existence of two isoforms calls for a re-evaluation of past research when CK1δ1 and CK1δ2 were simply CK1δ.


Subject(s)
Casein Kinase Idelta/genetics , Circadian Clocks/genetics , Methylation , Methyltransferases/genetics , RNA, Messenger/genetics , Animals , Casein Kinase Idelta/metabolism , Male , Methyltransferases/metabolism , Mice , Mice, Inbred C57BL , Protein Isoforms , RNA Splicing/genetics , RNA, Messenger/metabolism
12.
Proc Natl Acad Sci U S A ; 115(23): 5986-5991, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29784789

ABSTRACT

Multisite phosphorylation of the PERIOD 2 (PER2) protein is the key step that determines the period of the mammalian circadian clock. Previous studies concluded that an unidentified kinase is required to prime PER2 for subsequent phosphorylation by casein kinase 1 (CK1), an essential clock component that is conserved from algae to humans. These subsequent phosphorylations stabilize PER2, delay its degradation, and lengthen the period of the circadian clock. Here, we perform a comprehensive biochemical and biophysical analysis of mouse PER2 (mPER2) priming phosphorylation and demonstrate, surprisingly, that CK1δ/ε is indeed the priming kinase. We find that both CK1ε and a recently characterized CK1δ2 splice variant more efficiently prime mPER2 for downstream phosphorylation in cells than the well-studied splice variant CK1δ1. While CK1 phosphorylation of PER2 was previously shown to be robust to changes in the cellular environment, our phosphoswitch mathematical model of circadian rhythms shows that the CK1 carboxyl-terminal tail can allow the period of the clock to be sensitive to cellular signaling. These studies implicate the extreme carboxyl terminus of CK1 as a key regulator of circadian timing.


Subject(s)
Casein Kinase 1 epsilon/metabolism , Casein Kinase Idelta/metabolism , Circadian Rhythm/physiology , Period Circadian Proteins/metabolism , Animals , HEK293 Cells , Humans , Mice , Period Circadian Proteins/genetics , Phosphorylation
13.
Biomed Res ; 39(2): 57-63, 2018.
Article in English | MEDLINE | ID: mdl-29669984

ABSTRACT

Micturition behavior follows regular day/night fluctuations, and unwanted increase in micturition could occur during night in jet lag condition. To clarify the effect of jet lag on micturition behavior, we simultaneously detected circadian micturition patterns and locomotor activity rhythms of mice under experimental jet lag conditions, by applying the improved automated Voided Stain on Paper (aVSOP) method. When wild-type (WT) mice were phase-advanced for 8 hours, day-night variation of micturition was disrupted suddenly, and this irregular daily micturition continued until 8 days, although their activity rhythms entrained gradually day by day until 8 days. We also examined how jet lag induced changes of micturition in Per-null mice lacking Per1, Per2 and Per3 genes, whose endogenous clock is completely disrupted. We found both micturition and locomotor activity of Per-null mice promptly entrained to the new LD cycle. These findings suggest that the irregular micturition during jet lag is caused along with the gradual shift of the endogenous clock, and paradoxically, jet lag-associated abnormality was absent when endogenous circadian oscillations were genetically disrupted.


Subject(s)
Circadian Rhythm , Jet Lag Syndrome/physiopathology , Urination , Animals , Behavior, Animal , Circadian Clocks , Disease Models, Animal , Jet Lag Syndrome/genetics , Locomotion , Male , Mice , Mice, Knockout , Motor Activity , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism
14.
Nat Commun ; 8(1): 2238, 2017 12 21.
Article in English | MEDLINE | ID: mdl-29269828

ABSTRACT

Liver metabolism undergoes robust circadian oscillations in gene expression and enzymatic activity essential for liver homeostasis, but whether the circadian clock controls homeostatic self-renewal of hepatocytes is unknown. Here we show that hepatocyte polyploidization is markedly accelerated around the central vein, the site of permanent cell self-renewal, in mice deficient in circadian Period genes. In these mice, a massive accumulation of hyperpolyploid mononuclear and binuclear hepatocytes occurs due to impaired mitogen-activated protein kinase phosphatase 1 (Mkp1)-mediated circadian modulation of the extracellular signal-regulated kinase (Erk1/2) activity. Time-lapse imaging of hepatocytes suggests that the reduced activity of Erk1/2 in the midbody during cytokinesis results in abscission failure, leading to polyploidization. Manipulation of Mkp1 phosphatase activity is sufficient to change the ploidy level of hepatocytes. These data provide clear evidence that the Period genes not only orchestrate dynamic changes in metabolic activity, but also regulate homeostatic self-renewal of hepatocytes through Mkp1-Erk1/2 signaling pathway.


Subject(s)
Dual Specificity Phosphatase 1/metabolism , Hepatocytes/metabolism , Liver/metabolism , MAP Kinase Signaling System/physiology , Period Circadian Proteins/genetics , Polyploidy , Animals , Circadian Clocks/genetics , Hepatocytes/cytology , Hepatocytes/pathology , Liver/cytology , Liver/pathology , Mice , Mice, Knockout , Microscopy , Time-Lapse Imaging
15.
J Biol Rhythms ; 32(6): 609-620, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29088992

ABSTRACT

The suprachiasmatic nucleus (SCN) is an extremely robust self-sustained oscillator, containing virtually the same molecular clock present in other tissues in the body but, in addition, endowed with tight intercellular coupling dependent on multiple neurotransmitter systems that allow the SCN to function as the "master clock." Several studies on the circadian SCN transcriptome have been published and compared with the transcriptome of other tissues, but the recent focus shift toward the circadian metabolome and the importance of small molecules for circadian timekeeping has so far been limited to macroscopic tissues such as the liver. Here, we report the successful use of laser capture microdissection coupled with liquid chromatography/tandem mass spectrometry for the circadian profiling of SCN amino acids. Among 18 amino acids detected, 10 (55.5%) showed significant variations, particularly marked for proline, lysine, and histidine, with higher levels during the subjective day. Moreover, we compared SCN and cortical amino acid levels between wild-type and Bmal1-deficient animals, either in the whole body or specifically in the liver. Interestingly, lack of Bmal1 in the whole body led to a significant increase in most amino acids in the SCN but not in the cerebral cortex. In contrast, deletion of Bmal1 in the liver mostly affected cortical amino acid levels during the subjective day. This study demonstrates that laser capture microdissection can be used for the isolation of microscopic brain structures for metabolomic purposes and reveals interactions between liver and SCN amino acid metabolism.


Subject(s)
Amino Acids/metabolism , Cerebral Cortex/metabolism , Circadian Rhythm , Laser Capture Microdissection , Suprachiasmatic Nucleus/metabolism , ARNTL Transcription Factors/physiology , Animals , Chromatography, Liquid , Liver/metabolism , Male , Mass Spectrometry , Mice
16.
J Biol Rhythms ; 32(2): 101-108, 2017 04.
Article in English | MEDLINE | ID: mdl-28470123

ABSTRACT

Among nonphotic stimulants, a classic cholinergic agonist, carbachol, is known to have a strong and unique phase-resetting effect on the circadian clock: Intracerebroventricular carbachol treatment causes phase delays during the subjective early night and phase advances in the subjective late night, but the effects of this drug on the suprachiasmatic nucleus (SCN) in vivo and in vitro are still controversial. In the present study, we succeeded in reproducing the biphasic phase-shifting effect of carbachol on clock gene expression in organotypic SCN slices prepared from mice carrying a Per1-promoter fused luciferase gene ( Per1-luc). Since this biphasic effect of carbachol in Per1-luc SCN was prevented by atropine but not by mecamylamine, we concluded that these phase shifts were muscarinic receptor-dependent. Next, we analyzed the expression of muscarinic receptors in the SCN by in situ hybridization and found that M3 and M4 subtypes were expressed in SCN cells. These signals appeared neonatally and reached adult levels at postnatal day 10. Together, these findings suggest that carbachol has a phase-dependent phase-shifting effect on the SCN clock through muscarinic receptor subtypes expressed in the SCN.


Subject(s)
Carbachol/pharmacology , Cholinergic Agonists/pharmacology , Circadian Rhythm/drug effects , Period Circadian Proteins/genetics , Suprachiasmatic Nucleus/drug effects , Suprachiasmatic Nucleus/physiology , Transcription, Genetic , Animals , Animals, Newborn , Atropine/pharmacology , Circadian Clocks/drug effects , Gene Expression , Luciferases/genetics , Mecamylamine/pharmacology , Mice , Motor Activity , Muscarinic Antagonists/pharmacology , Nicotinic Antagonists/pharmacology , Organ Culture Techniques , Promoter Regions, Genetic , Receptors, Muscarinic/genetics , Receptors, Muscarinic/metabolism
17.
J Clin Endocrinol Metab ; 99(2): E257-62, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24423300

ABSTRACT

CONTEXT: Therapeutic management of primary aldosteronism requires accurate differentiation between aldosterone-producing adenoma (APA) and idiopathic hyperaldosteronism (IHA). However, little is known about the molecular features that delineate the difference between APA and IHA. Two different isoforms of 3ß-hydroxysteroid dehydrogenase (HSD3B1 and HSD3B2) are thought to be expressed in the human adrenal gland, but the lack of isoform-specific antibody has so far hampered mapping of these isoforms in APA and IHA. OBJECTIVES: The aim of our study is to develop and characterize isoform-specific monoclonal antibodies against HSD3B1 and HSD3B2. Using these antibodies, we determined for the first time the immunolocalization of HSD3B1 and HSD3B2 in normal human adrenal cortex as well as in adrenal specimens from APA and IHA. RESULTS: Immunohistochemical analysis with isoform-specific antibodies revealed zone-specific expression of HSD3B1 and HSD3B2 in the adrenal cortex. HSD3B1 immunoreactivities were essentially confined to the zona glomerulosa (ZG), in which aldosterone is produced. In contrast, HSD3B2 was not confined to the ZG but was found across the zona fasciculata, which is where cortisol is produced. Moreover, immunohistopathological analysis of primary aldosteronism revealed a previously uncharacterized difference between APA and IHA. Notably, hyperplasia of ZG seen for IHA was accompanied by a robust expression of ZG isoform HSD3B1. In contrast, tumor cells in APA were not immunopositive to HSD3B1. Rather, a strong and dominant expression of HSD3B2 characterized APA. Moreover, perhaps due to compensatory responses to excess aldosterone, APA had an adjacent ZG whose immunoreactivities to HSD3B1 and HSD3B2 were profoundly reduced. CONCLUSIONS: Isoform-specific monoclonal antibodies against HSD3B1 and HSD3B2 may be of great value for immunohistochemical differentiation between APA and IHA.


Subject(s)
3-Hydroxysteroid Dehydrogenases/immunology , Adrenal Cortex/metabolism , Hyperaldosteronism/immunology , Adenoma/metabolism , Adenoma/pathology , Adrenal Cortex/pathology , Adrenal Cortex Neoplasms/metabolism , Adrenal Cortex Neoplasms/pathology , Antibodies, Monoclonal/metabolism , Humans , Hyperaldosteronism/classification , Hyperaldosteronism/metabolism , Zona Glomerulosa/metabolism , Zona Glomerulosa/pathology
18.
Cell ; 155(4): 793-806, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24209618

ABSTRACT

The eukaryotic biological clock involves a negative transcription-translation feedback loop in which clock genes regulate their own transcription and that of output genes of metabolic significance. While around 10% of the liver transcriptome is rhythmic, only about a fifth is driven by de novo transcription, indicating mRNA processing is a major circadian component. Here, we report that inhibition of transmethylation reactions elongates the circadian period. RNA sequencing then reveals methylation inhibition causes widespread changes in the transcription of the RNA processing machinery, associated with m(6)A-RNA methylation. We identify m(6)A sites on many clock gene transcripts and show that specific inhibition of m(6)A methylation by silencing of the m(6)A methylase Mettl3 is sufficient to elicit circadian period elongation and RNA processing delay. Analysis of the circadian nucleocytoplasmic distribution of clock genes Per2 and Arntl then revealed an uncoupling between steady-state pre-mRNA and cytoplasmic mRNA rhythms when m(6)A methylation is inhibited.


Subject(s)
Circadian Clocks , Methyltransferases/metabolism , RNA Processing, Post-Transcriptional , RNA/metabolism , Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Cell Line , Cell Line, Tumor , Gene Knockdown Techniques , Humans , Methylation/drug effects , Methyltransferases/genetics , Period Circadian Proteins/metabolism , Tubercidin/pharmacology
19.
Science ; 342(6154): 85-90, 2013 Oct 04.
Article in English | MEDLINE | ID: mdl-24092737

ABSTRACT

Jet-lag symptoms arise from temporal misalignment between the internal circadian clock and external solar time. We found that circadian rhythms of behavior (locomotor activity), clock gene expression, and body temperature immediately reentrained to phase-shifted light-dark cycles in mice lacking vasopressin receptors V1a and V1b (V1a(-/-)V1b(-/-)). Nevertheless, the behavior of V1a(-/-)V1b(-/-) mice was still coupled to the internal clock, which oscillated normally under standard conditions. Experiments with suprachiasmatic nucleus (SCN) slices in culture suggested that interneuronal communication mediated by V1a and V1b confers on the SCN an intrinsic resistance to external perturbation. Pharmacological blockade of V1a and V1b in the SCN of wild-type mice resulted in accelerated recovery from jet lag, which highlights the potential of vasopressin signaling as a therapeutic target for management of circadian rhythm misalignment, such as jet lag and shift work.


Subject(s)
Jet Lag Syndrome/genetics , Receptors, Vasopressin/genetics , Animals , Antidiuretic Hormone Receptor Antagonists , Body Temperature/genetics , CLOCK Proteins/genetics , Cell Communication/drug effects , Cell Communication/genetics , Cells, Cultured , Circadian Rhythm/genetics , Gene Expression Regulation , Jet Lag Syndrome/physiopathology , Mice , Mice, Knockout , Motor Activity/genetics , Suprachiasmatic Nucleus/physiopathology
20.
Proc Natl Acad Sci U S A ; 110(23): 9547-52, 2013 Jun 04.
Article in English | MEDLINE | ID: mdl-23690615

ABSTRACT

The suprachiasmatic nucleus (SCN) coordinates circadian rhythms that adapt the individual to solar time. SCN pacemaking revolves around feedback loops in which expression of Period (Per) and Cryptochrome (Cry) genes is periodically suppressed by their protein products. Specifically, PER/CRY complexes act at E-box sequences in Per and Cry to inhibit their transactivation by CLOCK/BMAL1 heterodimers. To function effectively, these closed intracellular loops need to be synchronized between SCN cells and to the light/dark cycle. For Per expression, this is mediated by neuropeptidergic and glutamatergic extracellular cues acting via cAMP/calcium-responsive elements (CREs) in Per genes. Cry genes, however, carry no CREs, and how CRY-dependent SCN pacemaking is synchronized remains unclear. Furthermore, whereas reporter lines are available to explore Per circadian expression in real time, no Cry equivalent exists. We therefore created a mouse, B6.Cg-Tg(Cry1-luc)01Ld, carrying a transgene (mCry1-luc) consisting of mCry1 elements containing an E-box and E'-box driving firefly luciferase. mCry1-luc organotypic SCN slices exhibited stable circadian bioluminescence rhythms with appropriate phase, period, profile, and spatial organization. In SCN lacking vasoactive intestinal peptide or its receptor, mCry1 expression was damped and desynchronized between cells. Despite the absence of CREs, mCry1-luc expression was nevertheless (indirectly) sensitive to manipulation of cAMP-dependent signaling. In mPer1/2-null SCN, mCry1-luc bioluminescence was arrhythmic and no longer suppressed by elevation of cAMP. Finally, an SCN graft procedure showed that PER-independent as well as PER-dependent mechanisms could sustain circadian expression of mCry1. The mCry1-luc mouse therefore reports circadian mCry1 expression and its interactions with vasoactive intestinal peptide, cAMP, and PER at the heart of the SCN pacemaker.


Subject(s)
Circadian Rhythm/physiology , Cryptochromes/metabolism , Feedback, Physiological/physiology , Period Circadian Proteins/metabolism , Suprachiasmatic Nucleus/physiology , Animals , Cyclic AMP/metabolism , DNA Primers/genetics , Luciferases , Mice , Mice, Inbred C57BL , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL
...