Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
3.
Bio Protoc ; 11(5): e3944, 2021 Mar 05.
Article in English | MEDLINE | ID: mdl-33796618

ABSTRACT

An endogenous circadian clock system enables organisms to adapt to time-of-day dependent environmental changes. In consequence, most physiological processes exhibit daily rhythms of, e.g., energy metabolism, immune function, sleep, or hormone production. Hypothalamic circadian clocks have been identified to play a particular role in coordinating many of these processes. Primary neuronal cultures are widely used as a physiologically relevant model to study molecular events within neurons. However, as circadian rhythms include dynamic molecular changes over longer timescales that vary between individual cells, longitudinal measurement methods are essential to investigate the regulation of circadian clocks of hypothalamic neurons. Here we provide a protocol for generating primary hypothalamic neuronal cultures expressing a circadian luciferase reporter. Such reporter cells can be used to longitudinally monitor cellular circadian rhythms at high temporal resolution by performing bioluminescence measurements.

4.
Mol Metab ; 42: 101070, 2020 12.
Article in English | MEDLINE | ID: mdl-32898712

ABSTRACT

The nucleus of the solitary tract (NTS) is emerging as a major site of action for the appetite-suppressive effects of leading pharmacotherapies currently investigated to treat obesity. However, our understanding of how NTS neurons regulate appetite remains incomplete. OBJECTIVES: In this study, we used NTS nutrient sensing as an entry point to characterize stimulus-defined neuronal ensembles engaged by the NTS to produce physiological satiety. METHODS: We combined histological analysis, neuroanatomical assessment using inducible viral tracing tools, and functional tests to characterize hindbrain-forebrain circuits engaged by NTS leucine sensing to suppress hunger. RESULTS: We found that NTS detection of leucine engages NTS prolactin-releasing peptide (PrRP) neurons to inhibit AgRP neurons via a population of leptin receptor-expressing neurons in the dorsomedial hypothalamus. This circuit is necessary for the anorectic response to NTS leucine, the appetite-suppressive effect of high-protein diets, and the long-term control of energy balance. CONCLUSIONS: These results extend the integrative capability of AgRP neurons to include brainstem nutrient sensing inputs.


Subject(s)
Appetite Regulation/physiology , Feeding Behavior/physiology , Solitary Nucleus/physiology , Agouti-Related Protein/metabolism , Animals , Appetite/physiology , Brain/metabolism , Energy Metabolism , Hypothalamus/metabolism , Leptin/metabolism , Leucine/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Obesity , Solitary Nucleus/metabolism
5.
Elife ; 92020 07 09.
Article in English | MEDLINE | ID: mdl-32644041

ABSTRACT

Endogenous circadian clocks have evolved to anticipate 24 hr rhythms in environmental demands. Recent studies suggest that circadian rhythm disruption is a major risk factor for the development of metabolic disorders in humans. Conversely, alterations in energy state can disrupt circadian rhythms of behavior and physiology, creating a vicious circle of metabolic dysfunction. How peripheral energy state affects diurnal food intake, however, is still poorly understood. We here show that the adipokine adiponectin (ADIPOQ) regulates diurnal feeding rhythms through clocks in energy regulatory centers of the mediobasal hypothalamus (MBH). Adipoq-deficient mice show increased rest phase food intake associated with disrupted transcript rhythms of clock and appetite-regulating genes in the MBH. ADIPOQ regulates MBH clocks via AdipoR1-mediated upregulation of the core clock gene Bmal1. BMAL1, in turn, controls expression of orexigenic neuropeptide expression in the MBH. Together, these data reveal a systemic metabolic circuit to regulate central circadian clocks and energy intake.


Subject(s)
Adiponectin/metabolism , Circadian Rhythm/physiology , Eating/physiology , Feedback, Physiological , Mice/physiology , Animals , Female , Male , Mice, Knockout
6.
Cell Metab ; 31(2): 301-312.e5, 2020 02 04.
Article in English | MEDLINE | ID: mdl-31955990

ABSTRACT

To understand hindbrain pathways involved in the control of food intake, we examined roles for calcitonin receptor (CALCR)-containing neurons in the NTS. Ablation of NTS Calcr abrogated the long-term suppression of food intake, but not aversive responses, by CALCR agonists. Similarly, activating CalcrNTS neurons decreased food intake and body weight but (unlike neighboring CckNTS cells) failed to promote aversion, revealing that CalcrNTS neurons mediate a non-aversive suppression of food intake. While both CalcrNTS and CckNTS neurons decreased feeding via projections to the PBN, CckNTS cells activated aversive CGRPPBN cells while CalcrNTS cells activated distinct non-CGRP PBN cells. Hence, CalcrNTS cells suppress feeding via non-aversive, non-CGRP PBN targets. Additionally, silencing CalcrNTS cells blunted food intake suppression by gut peptides and nutrients, increasing food intake and promoting obesity. Hence, CalcrNTS neurons define a hindbrain system that participates in physiological energy balance and suppresses food intake without activating aversive systems.


Subject(s)
Eating , Energy Metabolism , Neurons/metabolism , Receptors, Calcitonin/physiology , Solitary Nucleus/metabolism , Animals , Body Weight , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Solitary Nucleus/cytology
7.
Chronobiol Int ; 35(11): 1543-1552, 2018 10.
Article in English | MEDLINE | ID: mdl-29993301

ABSTRACT

Circadian clock-controlled 24-h oscillations in adipose tissues play an important role in the regulation of energy homeostasis, thus representing a potential drug target for prevention and therapy of metabolic diseases. For pharmacological screens, scalable adipose model systems are needed that largely recapitulate clock properties observed in vivo. In this study, we compared molecular circadian clock regulation in different ex vivo and in vitro models derived from murine adipose tissues. Explant cultures from three different adipose depots of PER2::LUC circadian reporter mice revealed stable and comparable rhythms of luminescence ex vivo. Likewise, primary pre- and mature adipocytes from these mice displayed stable luminescence rhythms, but with strong damping in mature adipocytes. Stable circadian periods were also observed using Bmal1-luc and Per2-luc reporters after lentiviral transduction of wild-type pre-adipocytes. SV40 immortalized adipocytes of murine brown, subcutaneous and epididymal adipose tissue origin showed rhythmic mRNA expression of the core clock genes Bmal1, Per2, Dbp and REV-erbα in pre- and mature adipocytes, with a maturation-associated increase in overall mRNA levels and amplitudes. A comparison of clock gene mRNA rhythm phases revealed specific changes between in vivo and ex vivo conditions. In summary, our data indicate that adipose culture systems to a large extent mimic in vivo tissue clock regulation. Thus, both explant and cell systems may be useful tools for large-scale screens for adipose clock regulating factors.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Circadian Clocks/physiology , Circadian Rhythm/physiology , ARNTL Transcription Factors/genetics , Adiposity/physiology , Animals , CLOCK Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , Period Circadian Proteins/genetics
8.
FASEB J ; 31(11): 4707-4719, 2017 11.
Article in English | MEDLINE | ID: mdl-28710114

ABSTRACT

Endogenous circadian clocks regulate 24-h rhythms of physiology and behavior. Circadian rhythm disruption (CRD) is suggested as a risk factor for inflammatory bowel disease. However, the underlying molecular mechanisms remain unknown. Intestinal biopsies from Per1/2 mutant and wild-type (WT) mice were investigated by electron microscopy, immunohistochemistry, and bromodeoxyuridine pulse-chase experiments. TNF-α was injected intraperitoneally, with or without necrostatin-1, into Per1/2 mice or rhythmic and externally desynchronized WT mice to study intestinal epithelial cell death. Experimental chronic colitis was induced by oral administration of dextran sodium sulfate. In vitro, caspase activity was assayed in Per1/2-specific small interfering RNA-transfected cells. Wee1 was overexpressed to study antiapoptosis and the cell cycle. Genetic ablation of circadian clock function or environmental CRD in mice increased susceptibility to severe intestinal inflammation and epithelial dysregulation, accompanied by excessive necroptotic cell death and a reduced number of secretory epithelial cells. Receptor-interacting serine/threonine-protein kinase (RIP)-3-mediated intestinal necroptosis was linked to increased mitotic cell cycle arrest via Per1/2-controlled Wee1, resulting in increased antiapoptosis via cellular inhibitor of apoptosis-2. Together, our data suggest that circadian rhythm stability is pivotal for the maintenance of mucosal barrier function. CRD increases intestinal necroptosis, thus rendering the gut epithelium more susceptible to inflammatory processes.-Pagel, R., Bär, F., Schröder, T., Sünderhauf, A., Künstner, A., Ibrahim, S. M., Autenrieth, S. E., Kalies, K., König, P., Tsang, A. H., Bettenworth, D., Divanovic, S., Lehnert, H., Fellermann, K., Oster, H., Derer, S., Sina, C. Circadian rhythm disruption impairs tissue homeostasis and exacerbates chronic inflammation in the intestine.


Subject(s)
Circadian Rhythm , Homeostasis , Inflammatory Bowel Diseases/metabolism , Animals , Caspases/genetics , Caspases/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Imidazoles/pharmacology , Indoles/pharmacology , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Mice , Mice, Mutant Strains , Mutation , Necrosis , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha/adverse effects , Tumor Necrosis Factor-alpha/pharmacology
9.
Mol Metab ; 6(6): 512-523, 2017 06.
Article in English | MEDLINE | ID: mdl-28580282

ABSTRACT

OBJECTIVE: Circadian Clock gene mutant mice show dampened 24-h feeding rhythms and an increased sensitivity to high-fat diet (HFD) feeding. Restricting HFD access to the dark phase counteracts its obesogenic effect in wild-type mice. The extent to which altered feeding rhythms are causative for the obesogenic phenotype of Clock mutant mice, however, remains unknown. METHODS: Metabolic parameters of wild-type (WT) and ClockΔ19 mutant mice (MT) were investigated under ad libitum and nighttime restricted HFD feeding. Liver circadian clock function was partially rescued by hydrodynamic tail vein delivery of WT-Clock DNA vectors in mutant mice and transcriptional, metabolic, endocrine and behavioral rhythms studied. RESULTS: Nighttime-restricted feeding restored food intake, but not body weight regulation in MT mice under HFD, suggesting Clock-dependent metabolic dysregulation downstream of circadian appetite control. Liver-directed Clock gene therapy partially restored liver circadian oscillator function and transcriptome regulation without affecting centrally controlled circadian behaviors. Under HFD, MT mice with partially restored liver clock function (MT-LR) showed normalized body weight gain, rescued 24-h food intake rhythms, and WT-like energy expenditure. This was associated with decreased nighttime leptin and daytime ghrelin levels, reduced hepatic lipid accumulation, and improved glucose tolerance. Transcriptome analysis revealed that hepatic Clock rescue in MT mice affected a range of metabolic pathways. CONCLUSION: Liver Clock gene therapy improves resistance against HFD-induced metabolic impairments in mice with circadian clock disruption. Restoring or stabilizing liver clock function might be a promising target for therapeutic interventions in obesity and metabolic disorders.


Subject(s)
CLOCK Proteins/genetics , Diet, High-Fat/adverse effects , Genetic Therapy , Hyperphagia/therapy , Liver/metabolism , Obesity/prevention & control , Animals , CLOCK Proteins/metabolism , Feeding Behavior , Hyperphagia/complications , Male , Mice , Mice, Inbred C57BL , Mutation , Obesity/etiology
10.
Article in English | MEDLINE | ID: mdl-28243224

ABSTRACT

The circadian timing system consists on a distributed network of cellular clocks that together coordinate 24-h rhythms of physiology and behavior. Clock function and metabolism are tightly coupled, from the cellular to the organismal level. Genetic and non-genetic approaches in rodents have been employed to study circadian clock function in the living organism. Due to the ubiquitous expression of clock genes and the intricate interaction between the circadian system and energy metabolism, genetic approaches targeting specific tissue clocks have been used to assess their contribution in systemic metabolic processes. However, special requirements regarding specificity and efficiency have to be met to allow for valid conclusions from such studies. In this review, we provide a brief summary of different approaches developed for dissecting tissue clock function in the metabolic context in rodents, compare their strengths and weaknesses, and suggest new strategies in assessing tissue clock output and the consequences of circadian clock disruption in vivo.

11.
Compr Physiol ; 7(2): 383-427, 2017 03 16.
Article in English | MEDLINE | ID: mdl-28333377

ABSTRACT

The different types of adipose tissues fulfill a wide range of biological functions-from energy storage to hormone secretion and thermogenesis-many of which show pronounced variations over the course of the day. Such 24-h rhythms in physiology and behavior are coordinated by endogenous circadian clocks found in all tissues and cells, including adipocytes. At the molecular level, these clocks are based on interlocked transcriptional-translational feedback loops comprised of a set of clock genes/proteins. Tissue-specific clock-controlled transcriptional programs translate time-of-day information into physiologically relevant signals. In adipose tissues, clock gene control has been documented for adipocyte proliferation and differentiation, lipid metabolism as well as endocrine function and other adipose oscillations are under control of systemic signals tied to endocrine, neuronal, or behavioral rhythms. Circadian rhythm disruption, for example, by night shift work or through genetic alterations, is associated with changes in adipocyte metabolism and hormone secretion. At the same time, adipose metabolic state feeds back to central and peripheral clocks, adjusting behavioral and physiological rhythms. In this overview article, we summarize our current knowledge about the crosstalk between circadian clocks and energy metabolism with a focus on adipose physiology. © 2017 American Physiological Society. Compr Physiol 7:383-427, 2017.


Subject(s)
Adipose Tissue/physiology , Circadian Rhythm/physiology , Adipocytes/cytology , Adipocytes/physiology , Adipogenesis/physiology , Adipokines/metabolism , Adipose Tissue/anatomy & histology , Adipose Tissue/cytology , Animals , Body Temperature Regulation/physiology , Cell Differentiation/physiology , Chronobiology Disorders/complications , Chronobiology Disorders/metabolism , Circadian Clocks/physiology , Humans , Lipid Metabolism/physiology , Lipogenesis/physiology
12.
Immunity ; 46(1): 120-132, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28087238

ABSTRACT

Lymphocytes circulate through lymph nodes (LN) in search for antigen in what is believed to be a continuous process. Here, we show that lymphocyte migration through lymph nodes and lymph occurred in a non-continuous, circadian manner. Lymphocyte homing to lymph nodes peaked at night onset, with cells leaving the tissue during the day. This resulted in strong oscillations in lymphocyte cellularity in lymph nodes and efferent lymphatic fluid. Using lineage-specific genetic ablation of circadian clock function, we demonstrated this to be dependent on rhythmic expression of promigratory factors on lymphocytes. Dendritic cell numbers peaked in phase with lymphocytes, with diurnal oscillations being present in disease severity after immunization to induce experimental autoimmune encephalomyelitis (EAE). These rhythms were abolished by genetic disruption of T cell clocks, demonstrating a circadian regulation of lymphocyte migration through lymph nodes with time-of-day of immunization being critical for adaptive immune responses weeks later.


Subject(s)
Adaptive Immunity/immunology , Chemotaxis, Leukocyte/immunology , Circadian Clocks/immunology , Immunologic Surveillance/immunology , Lymphocytes/immunology , Adoptive Transfer , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Flow Cytometry , Fluorescent Antibody Technique , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Real-Time Polymerase Chain Reaction
13.
Endocrinology ; 157(11): 4222-4233, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27690690

ABSTRACT

The circadian rhythm of glucocorticoids affects diverse physiological systems, including stress responses and the coordination of rhythmic functions in peripheral and central tissues. Circadian clocks are considered to be important coordinators of glucocorticoid release and loss of the core clock component Brain and muscle Arnt-like protein-1 leads to ablation of behavioral and physiological rhythms, hypocortisolism, impaired ACTH, and behavioral stress responses. Transplantation and conditional clock gene knock-down studies in mice suggest an important role of local adrenocortical clock function in this context. Here, we present a Cre-loxP-mediated conditional knockout of Bmal1 in the steroidogenic cells of the adrenal cortex in mice. Mutant animals show a loss of molecular clock gene activity rhythms in this tissue with subsequent disruption of rhythmic steroidogenic gene expression. However, despite this loss of normal clock rhythmicity in the adrenal cortex, behavioral and physiological rhythms and acute stress responses persist in mutant mice. These findings reveal a dissociation of transcriptional and endocrine rhythm regulation in the adrenal cortex, arguing for a less pivotal function of the local clock machinery in the regulation of circadian and acute glucocorticoid outputs.


Subject(s)
ARNTL Transcription Factors/deficiency , Adrenal Cortex/metabolism , Circadian Rhythm/physiology , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Adrenal Cortex Hormones/metabolism , Animals , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Corticosterone/metabolism , Genotype , Glucocorticoids/metabolism , Male , Mice , Mice, Knockout , Mice, Mutant Strains , Physical Conditioning, Animal
14.
J Endocrinol ; 230(1): R1-R11, 2016 07.
Article in English | MEDLINE | ID: mdl-27106109

ABSTRACT

Endogenous circadian clocks regulate 24-h rhythms of behavior and physiology to align with external time. The endocrine system serves as a major clock output to regulate various biological processes. Recent findings suggest that some of the rhythmic hormones can also provide feedback to the circadian system at various levels, thus contributing to maintaining the robustness of endogenous rhythmicity. This delicate balance of clock-hormone interaction is vulnerable to modern lifestyle factors such as shiftwork or high-calorie diets, altering physiological set points. In this review, we summarize the current knowledge on the communication between the circadian timing and endocrine systems, with a focus on adrenal glucocorticoids and metabolic peptide hormones. We explore the potential role of hormones as systemic feedback signals to adjust clock function and their relevance for the maintenance of physiological and metabolic circadian homeostasis.


Subject(s)
Circadian Clocks/physiology , Circadian Rhythm/physiology , Endocrine System/metabolism , Animals , Glucocorticoids/metabolism , Humans , Hypothalamo-Hypophyseal System/physiology , Insulin/metabolism , Leptin/metabolism , Pituitary-Adrenal System/physiology
15.
Elife ; 4: e06253, 2015 Mar 30.
Article in English | MEDLINE | ID: mdl-25821984

ABSTRACT

Circadian clocks coordinate 24-hr rhythms of behavior and physiology. In mammals, a master clock residing in the suprachiasmatic nucleus (SCN) is reset by the light-dark cycle, while timed food intake is a potent synchronizer of peripheral clocks such as the liver. Alterations in food intake rhythms can uncouple peripheral clocks from the SCN, resulting in internal desynchrony, which promotes obesity and metabolic disorders. Pancreas-derived hormones such as insulin and glucagon have been implicated in signaling mealtime to peripheral clocks. In this study, we identify a novel, more direct pathway of food-driven liver clock resetting involving oxyntomodulin (OXM). In mice, food intake stimulates OXM secretion from the gut, which resets liver transcription rhythms via induction of the core clock genes Per1 and 2. Inhibition of OXM signaling blocks food-mediated resetting of hepatocyte clocks. These data reveal a direct link between gastric filling with food and circadian rhythm phasing in metabolic tissues.


Subject(s)
Circadian Clocks/drug effects , Circadian Rhythm/drug effects , Liver/drug effects , Oxyntomodulin/pharmacology , Period Circadian Proteins/genetics , Animals , Circadian Clocks/genetics , Circadian Rhythm/genetics , Eating/drug effects , Eating/physiology , Fasting , Gene Expression Regulation , Insulin/biosynthesis , Insulin/metabolism , Insulin Secretion , Intestinal Mucosa/metabolism , Intestines/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Microtomy , Oxyntomodulin/biosynthesis , Oxyntomodulin/genetics , Oxyntomodulin/metabolism , Period Circadian Proteins/metabolism , Photoperiod , Signal Transduction , Suprachiasmatic Nucleus/drug effects , Suprachiasmatic Nucleus/physiology , Tissue Culture Techniques
16.
Horm Mol Biol Clin Investig ; 19(2): 103-15, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25390019

ABSTRACT

Endogenous circadian clocks facilitate the adaptation of physiology and behavior to recurring environmental changes brought about by the Earth's rotation around its axis. Adipose tissues harbor intrinsic circadian oscillators based on interlocked transcriptional-translational feedback loops built from a set of clock genes that regulate important aspects of lipid metabolism and adipose endocrine function. These adipocyte clocks are reset via neuronal and endocrine pathways originating from a master circadian pacemaker residing in the hypothalamic suprachiasmatic nucleus. One important mediator of circadian output is the stress hormone cortisol, which, at the same time, is one of the major regulators of adipose physiology. In this review we summarize recent findings on the interaction between circadian and stress systems in the regulation of adipose physiology and discuss the implications of this crosstalk for the development of metabolic disorders associated with circadian disruption and/or chronic stress, for example in shift workers.


Subject(s)
Adipose Tissue/physiology , Circadian Clocks , Stress, Physiological , Animals , Humans , Hydrocortisone/metabolism , Hypothalamo-Hypophyseal System/physiopathology , Metabolic Diseases/metabolism , Metabolic Diseases/physiopathology , Pituitary-Adrenal System/physiopathology
17.
FASEB J ; 28(11): 4950-60, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25063847

ABSTRACT

The mammalian circadian timing system consists of a master pacemaker in the suprachiasmatic nucleus (SCN), which is thought to synchronize peripheral clocks in various organs with each other and with external time. Our knowledge about the role of the SCN clock is based mainly on SCN lesion and transplantation studies. We have now directly deleted the SCN clock using the Cre/LoxP system and investigated how this affects synchronization of peripheral rhythms. Impaired locomotor activity and arrhythmic clock gene expression in the SCN confirm that the SCN clockwork was efficiently abolished in our mouse model. Nonetheless, under light-dark (LD) conditions, peripheral clocks remained rhythmic and synchronized to the LD cycle, and phase relationships between peripheral clocks were sustained. Adaptation to a shifted LD cycle was accelerated in SCN clock-deficient mice. Moreover, under zeitgeber-free conditions, rhythmicity of the peripheral clock gene expression was initially dampened, and after several days peripheral clocks were desynchronized. These findings suggest that the SCN clock is dispensable for the synchronization of peripheral clocks to the LD cycle. A model describing an SCN clock-independent pathway that synchronizes peripheral clocks with the LD cycle is discussed.


Subject(s)
CLOCK Proteins/metabolism , Circadian Rhythm/genetics , Photoperiod , Suprachiasmatic Nucleus/metabolism , Animals , Circadian Clocks/genetics , Circadian Clocks/physiology , Circadian Rhythm/physiology , Gene Expression/physiology , Mice , Motor Activity/genetics
18.
J Mol Endocrinol ; 52(1): R1-16, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23997239

ABSTRACT

In most species, endogenous circadian clocks regulate 24-h rhythms of behavior and physiology. Clock disruption has been associated with decreased cognitive performance and increased propensity to develop obesity, diabetes, and cancer. Many hormonal factors show robust diurnal secretion rhythms, some of which are involved in mediating clock output from the brain to peripheral tissues. In this review, we describe the mechanisms of clock-hormone interaction in mammals, the contribution of different tissue oscillators to hormonal regulation, and how changes in circadian timing impinge on endocrine signalling and downstream processes. We further summarize recent findings suggesting that hormonal signals may feed back on circadian regulation and how this crosstalk interferes with physiological and metabolic homeostasis.


Subject(s)
Circadian Rhythm/physiology , Endocrine System/physiology , Adrenal Glands/physiology , Animals , Circadian Clocks/physiology , Hormones/physiology , Humans , Pineal Gland/physiology , Suprachiasmatic Nucleus/physiology
19.
Am J Physiol Endocrinol Metab ; 304(10): E1053-63, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23531614

ABSTRACT

Perturbation of circadian rhythmicity in mammals, either by environmental influences such as shiftwork or by genetic manipulation, has been associated with metabolic disturbance and the development of obesity and diabetes. Circadian clocks are based on transcriptional/translational feedback loops, comprising positive and negative components. Whereas the metabolic effects of deletion of the positive arm of the clock gene machinery, as in Clock- or Bmal1-deficient mice, have been well characterized, inactivation of Period genes (Per1-3) as components of the negative arm have more complex, sometimes contradictory effects on energy homeostasis. The CRYPTOCHROMEs are critical interaction partners of PERs, and simultaneous deletion of Cry1 and -2 results in behavioral and molecular circadian arrhythmicity. We show that, when challenged with a high-fat diet, Cry1/2(-/-) mice rapidly gain weight and surpass that of wild-type mice, despite displaying hypophagia. Transcript analysis of white adipose tissue reveals upregulated expression of lipogenic genes, many of which are insulin targets. High-fat diet-induced hyperinsulinemia, as a result of potentiated insulin secretion, coupled with selective insulin sensitivity in adipose tissue of Cry1/2(-/-) mice, correlates with increased lipid uptake. Collectively, these data indicate that Cry deficiency results in an increased vulnerability to high-fat diet-induced obesity that might be mediated by increased insulin secretion and lipid storage in adipose tissues.


Subject(s)
Adipose Tissue, White/metabolism , Circadian Rhythm/physiology , Cryptochromes/physiology , Hyperinsulinism/metabolism , Insulin Resistance/physiology , Animals , Blood Glucose/metabolism , Calorimetry, Indirect/methods , Circadian Rhythm/genetics , Cryptochromes/genetics , Diet, High-Fat , Histocytochemistry , Hyperinsulinism/etiology , Hyperinsulinism/genetics , Insulin/blood , Insulin Resistance/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/chemistry , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Up-Regulation/physiology
20.
J Biol Rhythms ; 27(6): 478-89, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23223373

ABSTRACT

In mammals, the molecular circadian clockwork is comprised of interlocked transcriptional-translational feedback loops (TTLs). Three Period (Per1-3) and 2 Dec (Dec1/2) genes interact in regulating the activity of the transcriptional activators CLOCK/NPAS2 and BMAL1. While deletion of Per1 and Per2 in mice results in behavioral arrhythmicity, Dec deletion has less dramatic effects on activity rhythms, affecting primarily phase of entrainment and free-running period. In intact animals, clock gene mutant phenotypes are often masked due to intercellular coupling mechanisms that stabilize cellular rhythms. Therefore, to study Per/Dec genetic interaction at the cellular level, we isolated fibroblasts from different tissues of Per1, Per2, and Dec2 single and double mutant mice. We show that in the cellular TTL, Pers and Dec2 act in a principally synergistic way, but tissue-specific differences in this interaction are seen. A rescue of rhythmicity in Per2 mutant cells after additional deletion of Dec2 was observed, indicating that in the absence of Per2, DEC2 destabilizes TTL function. Rhythm power in Per1/Dec2 and Per2/Dec2 double mutants was strongly reduced, suggesting that interaction of Dec2 with both Per genes is important for stabilizing clock period. Contrary to what was observed for behavior, nonsynergistic effects of Dec2 and Per1/2 mutations were observed on cellular clock phase regulation that do not correlate with period effects. Our data reveal cell type-specific interactions of Per1/2 and Dec2 in the regulation of period, phase, and rhythm sustainment, emphasizing the differential organization of the mammalian clock machinery in different tissues.


Subject(s)
Circadian Rhythm , Fibroblasts/physiology , Period Circadian Proteins/metabolism , Transcription Factors/metabolism , Animals , Cells, Cultured , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation , Genes, Reporter , Male , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...