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










Publication year range
1.
J Exp Biol ; 227(9)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38634224

ABSTRACT

In many species of animals, red carotenoid-based coloration is produced by metabolizing yellow dietary pigments, and this red ornamentation can be an honest signal of individual quality. However, the physiological basis for associations between organism function and the metabolism of red ornamental carotenoids from yellow dietary carotenoids remains uncertain. A recent hypothesis posits that carotenoid metabolism depends on mitochondrial performance, with diminished red coloration resulting from altered mitochondrial aerobic respiration. To test for an association between mitochondrial respiration and red carotenoids, we held wild-caught, molting male house finches in either small bird cages or large flight cages to create environmental challenges during the period when red ornamental coloration is produced. We predicted that small cages would present a less favorable environment than large flight cages and that captivity itself would decrease both mitochondrial performance and the abundance of red carotenoids compared with free-living birds. We found that captive-held birds circulated fewer red carotenoids, showed increased mitochondrial respiratory rates, and had lower complex II respiratory control ratios - a metric associated with mitochondrial efficiency - compared with free-living birds, though we did not detect a difference in the effects of small cages versus large cages. Among captive individuals, the birds that circulated the highest concentrations of red carotenoids had the highest mitochondrial respiratory control ratio for complex II substrate. These data support the hypothesis that the metabolism of red carotenoid pigments is linked to mitochondrial aerobic respiration in the house finch, but the mechanisms for this association remain to be established.


Subject(s)
Carotenoids , Finches , Mitochondria , Animals , Carotenoids/metabolism , Male , Finches/physiology , Finches/metabolism , Mitochondria/metabolism , Cell Respiration , Oxygen Consumption
2.
Metabolites ; 12(7)2022 Jul 16.
Article in English | MEDLINE | ID: mdl-35888782

ABSTRACT

Metabolic disease resulting from overnutrition is prevalent and rapidly increasing in incidence in modern society. Time restricted feeding (TRF) dietary regimens have recently shown promise in attenuating some of the negative metabolic effects associated with chronic nutrient stress. The purpose of this study is to utilize a multi-tissue metabolomics approach using nuclear magnetic resonance (NMR) spectroscopy to investigate TRF and sex-specific effects of high-fat diet in a diurnal Nile grass rat model. Animals followed a six-week dietary protocol on one of four diets: chow ad libitum, high-fat ad libitum (HF-AD), high-fat early TRF (HF-AM), or high-fat late TRF (HF-PM), and their liver, heart, and white adipose tissues were harvested at the end of the study and were analyzed by NMR. Time-domain complete reduction to amplitude-frequency table (CRAFT) was used to semi-automate and systematically quantify metabolites in liver, heart, and adipose tissues while minimizing operator bias. Metabolite profiling and statistical analysis revealed lipid remodeling in all three tissues and ectopic accumulation of cardiac and hepatic lipids for HF-AD feeding compared to a standard chow diet. Animals on TRF high-fat diet had lower lipid levels in the heart and liver compared to the ad libitum group; however, no significant differences were noted for adipose tissue. Regardless of diet, females exhibited greater amounts of hepatic lipids compared to males, while no consistent differences were shown in adipose and heart. In conclusion, this study demonstrates the feasibility of performing systematic and time-efficient multi-tissue NMR metabolomics to elucidate metabolites involved in the crosstalk between different metabolic tissues and provides a more holistic approach to better understand the etiology of metabolic disease and the effects of TRF on metabolic profiles.

3.
Biochim Biophys Acta Bioenerg ; 1863(7): 148582, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35667393

ABSTRACT

Naked mole-rats (NMR) and Damaraland mole-rats (DMR) exhibit extraordinary longevity for their body size, high tolerance to hypoxia and oxidative stress and high reproductive output; these collectively defy the concept that life-history traits should be negatively correlated. However, when life-history traits share similar underlying physiological mechanisms, these may be positively associated with each other. We propose that one such potential common mechanism might be the bioenergetic properties of mole-rats. Here, we aim to characterize the bioenergetic properties of two African mole-rats. We adopted a top-down perspective measuring the bioenergetic properties at the organismal, cellular, and molecular level in both species and the biological significance of these properties were compared with the same measures in Siberian hamsters and C57BL/6 mice, chosen for their similar body size to the mole-rat species. We found mole-rats shared several bioenergetic properties that differed from their comparison species, including low basal metabolic rates, a high dependence on glycolysis rather than on oxidative phosphorylation for ATP production, and low proton conductance across the mitochondrial inner membrane. These shared mole-rat features could be a result of evolutionary adaptation to tolerating variable oxygen atmospheres, in particular hypoxia, and may in turn be one of the molecular mechanisms underlying their extremely long lifespans.


Subject(s)
Mitochondria , Mole Rats , Animals , Hypoxia , Mice , Mice, Inbred C57BL , Mole Rats/physiology , Respiration
4.
Medicines (Basel) ; 9(2)2022 Feb 21.
Article in English | MEDLINE | ID: mdl-35200758

ABSTRACT

Lengthening the daily eating period contributes to the onset of obesity and metabolic syndrome. Dietary approaches, including energy restriction and time-restricted feeding, are promising methods to combat metabolic disorders. This study explored the effect of early and late time-restricted feeding (TRF) on weight and adiposity, food consumption, glycemic control, clock gene expression, and liver metabolite composition in diurnal Nile grass rats (NGRs). Adult male and female Nile grass rats were randomly assigned to one of three groups: (1) access to a 60% high-fat (HF) diet ad-libitum (HF-AD), (2) time-restricted access to the HF diet for the first 6 h of the 12 h light/active phase (HF-AM) or (3) the second 6 h of the 12 h light/active phase (HF-PM). Animals remained on their respective protocols for six weeks. TRF reduced total energy consumption and weight gain, and early TRF (HF-AM) reduced fasting blood glucose, restored Per1 expression, and reduced liver lipid levels. Although sex-dependent differences were observed for fat storage and lipid composition, TRF improved metabolic parameters in both male and female NGRs. In conclusion, this study demonstrated that early TRF protocol benefits weight management, improves lipid and glycemic control, and restores clock gene expression in NGRs.

5.
Nutrients ; 14(2)2022 Jan 12.
Article in English | MEDLINE | ID: mdl-35057482

ABSTRACT

As a redox-sensitive coenzyme, nicotinamide adenine dinucleotide (NAD+) plays a central role in cellular energy metabolism and homeostasis. Low NAD+ levels are linked to multiple disease states, including age-related diseases, such as metabolic and neurodegenerative diseases. Consequently, restoring/increasing NAD+ levels in vivo has emerged as an important intervention targeting age-related neurodegenerative diseases. One of the widely studied approaches to increase NAD+ levels in vivo is accomplished by using NAD+ precursors, such as nicotinamide mononucleotide (NMN). Oral administration of NMN has been shown to successfully increase NAD+ levels in a variety of tissues; however, it remains unclear whether NMN can cross the blood-brain barrier to increase brain NAD+ levels. This study evaluated the effects of oral NMN administration on NAD+ levels in C57/B6J mice brain tissues. Our results demonstrate that oral gavage of 400 mg/kg NMN successfully increases brain NAD+ levels in mice after 45 min. These findings provide evidence that NMN may be used as an intervention to increase NAD+ levels in the brain.


Subject(s)
Brain/drug effects , NAD/metabolism , Nicotinamide Mononucleotide/administration & dosage , Administration, Oral , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Humans , Mice , Mice, Inbred C57BL , Neurodegenerative Diseases/metabolism
6.
Sci Rep ; 9(1): 11883, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31417156

ABSTRACT

Post-translational regulation plays a central role in the circadian clock mechanism. However, nucleocytoplasmic translocation of core clock proteins, a key step in circadian timekeeping, is not fully understood. Earlier we found that the NRON scaffolding complex regulates nuclear translocation of NFAT and its signaling. Here, we show that components of the NRON complex also regulate the circadian clock. In peripheral cell clock models, genetic perturbation of the NRON complex affects PER and CRY protein nuclear translocation, dampens amplitude, and alters period length. Further, we show small molecules targeting the NFAT pathway alter nuclear translocation of PER and CRY proteins and impact circadian rhythms in peripheral cells and tissue explants of the master clock in the suprachiasmatic nucleus. Taken together, these studies highlight a key role for the NRON complex in regulating PER/CRY subcellular localization and circadian timekeeping.


Subject(s)
Cell Nucleus/metabolism , Circadian Clocks/physiology , Cryptochromes/metabolism , Period Circadian Proteins/metabolism , RNA, Long Noncoding/genetics , Animals , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Calcium Signaling , Cell Line , Circadian Rhythm/physiology , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Mice , Models, Biological , Protein Transport , RNA Interference , Signal Transduction
7.
PLoS Genet ; 14(5): e1007369, 2018 05.
Article in English | MEDLINE | ID: mdl-29750810

ABSTRACT

The circadian clock coordinates physiology and metabolism. mTOR (mammalian/mechanistic target of rapamycin) is a major intracellular sensor that integrates nutrient and energy status to regulate protein synthesis, metabolism, and cell growth. Previous studies have identified a key role for mTOR in regulating photic entrainment and synchrony of the central circadian clock in the suprachiasmatic nucleus (SCN). Given that mTOR activities exhibit robust circadian oscillations in a variety of tissues and cells including the SCN, here we continued to investigate the role of mTOR in orchestrating autonomous clock functions in central and peripheral circadian oscillators. Using a combination of genetic and pharmacological approaches we show that mTOR regulates intrinsic clock properties including period and amplitude. In peripheral clock models of hepatocytes and adipocytes, mTOR inhibition lengthens period and dampens amplitude, whereas mTOR activation shortens period and augments amplitude. Constitutive activation of mTOR in Tsc2-/-fibroblasts elevates levels of core clock proteins, including CRY1, BMAL1 and CLOCK. Serum stimulation induces CRY1 upregulation in fibroblasts in an mTOR-dependent but Bmal1- and Period-independent manner. Consistent with results from cellular clock models, mTOR perturbation also regulates period and amplitude in the ex vivo SCN and liver clocks. Further, mTOR heterozygous mice show lengthened circadian period of locomotor activity in both constant darkness and constant light. Together, these results support a significant role for mTOR in circadian timekeeping and in linking metabolic states to circadian clock functions.


Subject(s)
Circadian Clocks/genetics , Signal Transduction/genetics , Suprachiasmatic Nucleus/metabolism , TOR Serine-Threonine Kinases/genetics , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Cell Line , Cells, Cultured , Fibroblasts/metabolism , Hepatocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Suprachiasmatic Nucleus/cytology , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
8.
Methods Mol Biol ; 1755: 49-64, 2018.
Article in English | MEDLINE | ID: mdl-29671262

ABSTRACT

In mammals, many aspects of metabolic, physiological, and behavioral processes are regulated by endogenous circadian clocks. Oscillators of different tissue types share a common molecular mechanism at the cellular and molecular level which underlies the rhythmic expression of genes. Individual cells are the functional units for rhythm generation and cell-based clock models offer experimental tractability for discovery. Cellular clock models can be developed by introducing a noninvasive and readily detectable luciferase bioluminescence reporter as a rhythmic output, in which the promoter of a rhythmically expressed gene is fused with the firefly luciferase (Luc) gene. The bioluminescence expression in the cells is measured continuously over several days using a highly sensitive and automated recording device. As such, the data are of high temporal resolution and allow precise determination of key circadian parameters including period length, amplitude, damping rate, and phase. Miniaturization of the assays improves throughput for large scale screens. In our lab, we have expertise for constructing circadian reporters and developing reporter cell lines. Here, we describe the procedure for establishing a stable mouse hepatocyte reporter cell line. The procedure described here can be applied to various other cell types.


Subject(s)
Circadian Clocks/physiology , Genes, Reporter/genetics , Luciferases, Firefly/genetics , Luminescent Measurements/methods , Animals , Cell Line , Genetic Vectors/genetics , Hepatocytes , Lentivirus/genetics , Luciferases, Firefly/chemistry , Luminescent Measurements/instrumentation , Mice , Transfection/instrumentation , Transfection/methods
9.
Elife ; 72018 02 26.
Article in English | MEDLINE | ID: mdl-29481323

ABSTRACT

Diurnal oscillation of intracellular redox potential is known to couple metabolism with the circadian clock, yet the responsible mechanisms are not well understood. We show here that chemical activation of NRF2 modifies circadian gene expression and rhythmicity, with phenotypes similar to genetic NRF2 activation. Loss of Nrf2 function in mouse fibroblasts, hepatocytes and liver also altered circadian rhythms, suggesting that NRF2 stoichiometry and/or timing of expression are important to timekeeping in some cells. Consistent with this concept, activation of NRF2 at a circadian time corresponding to the peak generation of endogenous oxidative signals resulted in NRF2-dependent reinforcement of circadian amplitude. In hepatocytes, activated NRF2 bound specific enhancer regions of the core clock repressor gene Cry2, increased Cry2 expression and repressed CLOCK/BMAL1-regulated E-box transcription. Together these data indicate that NRF2 and clock comprise an interlocking loop that integrates cellular redox signals into tissue-specific circadian timekeeping.


Subject(s)
CLOCK Proteins/metabolism , Circadian Clocks , NF-E2-Related Factor 2/metabolism , Animals , Cell Line , Gene Expression Regulation , Mice , Oxidation-Reduction
10.
Nat Struct Mol Biol ; 22(6): 476-484, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25961797

ABSTRACT

The molecular circadian clock in mammals is generated from transcriptional activation by the bHLH-PAS transcription factor CLOCK-BMAL1 and subsequent repression by PERIOD and CRYPTOCHROME (CRY). The mechanism by which CRYs repress CLOCK-BMAL1 to close the negative feedback loop and generate 24-h timing is not known. Here we show that, in mouse fibroblasts, CRY1 competes for binding with coactivators to the intrinsically unstructured C-terminal transactivation domain (TAD) of BMAL1 to establish a functional switch between activation and repression of CLOCK-BMAL1. TAD mutations that alter affinities for co-regulators affect the balance of repression and activation to consequently change the intrinsic circadian period or eliminate cycling altogether. Our results suggest that CRY1 fulfills its role as an essential circadian repressor by sequestering the TAD from coactivators, and they highlight regulation of the BMAL1 TAD as a critical mechanism for establishing circadian timing.


Subject(s)
ARNTL Transcription Factors/metabolism , Circadian Clocks , Cryptochromes/metabolism , Animals , Cells, Cultured , Fibroblasts/physiology , Humans , Mice , Mice, Knockout
11.
Physiol Behav ; 138: 75-86, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25447482

ABSTRACT

Photic cues influence daily patterns of activity via two complementary mechanisms: (1) entraining the internal circadian clock and (2) directly increasing or decreasing activity, a phenomenon referred to as "masking". The direction of this masking response is dependent on the temporal niche an organism occupies, as nocturnal animals often decrease activity when exposed to light, while the opposite response is more likely to be seen in diurnal animals. Little is known about the neural mechanisms underlying these differences. Here, we examined the masking effects of light on behavior and the activation of several brain regions by that light, in diurnal Arvicanthis niloticus (Nile grass rats) and nocturnal Mus musculus (mice). Each species displayed the expected behavioral response to a 1h pulse of light presented 2h after lights-off, with the diurnal grass rats and nocturnal mice increasing and decreasing their activity, respectively. In grass rats light induced an increase in cFOS in all retinorecipient areas examined, which included the suprachiasmatic nucleus (SCN), the ventral subparaventricular zone (vSPZ), intergeniculate leaflet (IGL), lateral habenula (LH), olivary pretectal nucleus (OPT) and the dorsal lateral geniculate (DLG). In mice, light led to an increase in cFOS in one of these regions (SCN), no change in others (vSPZ, IGL and LH) and a decrease in two (OPT and DLG). In addition, light increased cFOS expression in three arousal-related brain regions (the lateral hypothalamus, dorsal raphe, and locus coeruleus) and in one sleep-promoting region (the ventrolateral preoptic area) in grass rats. In mice, light had no effect on cFOS in these four regions. Taken together, these results highlight several brain regions whose responses to light suggest that they may play a role in masking, and that the possibility that they contribute to species-specific patterns of behavioral responses to light should be explored in future.


Subject(s)
Brain/physiology , Circadian Rhythm/physiology , Light , Motor Activity/physiology , Murinae/physiology , Actigraphy , Animals , Female , Immunohistochemistry , Male , Photic Stimulation , Photomicrography , Proto-Oncogene Proteins c-fos/metabolism , Species Specificity
12.
PLoS Biol ; 12(4): e1001840, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24737000

ABSTRACT

Over the last decades, researchers have characterized a set of "clock genes" that drive daily rhythms in physiology and behavior. This arduous work has yielded results with far-reaching consequences in metabolic, psychiatric, and neoplastic disorders. Recent attempts to expand our understanding of circadian regulation have moved beyond the mutagenesis screens that identified the first clock components, employing higher throughput genomic and proteomic techniques. In order to further accelerate clock gene discovery, we utilized a computer-assisted approach to identify and prioritize candidate clock components. We used a simple form of probabilistic machine learning to integrate biologically relevant, genome-scale data and ranked genes on their similarity to known clock components. We then used a secondary experimental screen to characterize the top candidates. We found that several physically interact with known clock components in a mammalian two-hybrid screen and modulate in vitro cellular rhythms in an immortalized mouse fibroblast line (NIH 3T3). One candidate, Gene Model 129, interacts with BMAL1 and functionally represses the key driver of molecular rhythms, the BMAL1/CLOCK transcriptional complex. Given these results, we have renamed the gene CHRONO (computationally highlighted repressor of the network oscillator). Bi-molecular fluorescence complementation and co-immunoprecipitation demonstrate that CHRONO represses by abrogating the binding of BMAL1 to its transcriptional co-activator CBP. Most importantly, CHRONO knockout mice display a prolonged free-running circadian period similar to, or more drastic than, six other clock components. We conclude that CHRONO is a functional clock component providing a new layer of control on circadian molecular dynamics.


Subject(s)
ARNTL Transcription Factors/metabolism , Circadian Clocks/physiology , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Histone Deacetylases/metabolism , Repressor Proteins/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Artificial Intelligence , Cell Line , Circadian Clocks/genetics , Circadian Rhythm/genetics , Circadian Rhythm/physiology , Circadian Rhythm Signaling Peptides and Proteins/biosynthesis , Circadian Rhythm Signaling Peptides and Proteins/genetics , Cryptochromes/genetics , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Glucocorticoid/metabolism , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Sequence Alignment , Transcription, Genetic/genetics
13.
PLoS Genet ; 10(4): e1004244, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24699442

ABSTRACT

In animals, circadian rhythms in physiology and behavior result from coherent rhythmic interactions between clocks in the brain and those throughout the body. Despite the many tissue specific clocks, most understanding of the molecular core clock mechanism comes from studies of the suprachiasmatic nuclei (SCN) of the hypothalamus and a few other cell types. Here we report establishment and genetic characterization of three cell-autonomous mouse clock models: 3T3 fibroblasts, 3T3-L1 adipocytes, and MMH-D3 hepatocytes. Each model is genetically tractable and has an integrated luciferase reporter that allows for longitudinal luminescence recording of rhythmic clock gene expression using an inexpensive off-the-shelf microplate reader. To test these cellular models, we generated a library of short hairpin RNAs (shRNAs) against a panel of known clock genes and evaluated their impact on circadian rhythms. Knockdown of Bmal1, Clock, Cry1, and Cry2 each resulted in similar phenotypes in all three models, consistent with previous studies. However, we observed cell type-specific knockdown phenotypes for the Period and Rev-Erb families of clock genes. In particular, Per1 and Per2, which have strong behavioral effects in knockout mice, appear to play different roles in regulating period length and amplitude in these peripheral systems. Per3, which has relatively modest behavioral effects in knockout mice, substantially affects period length in the three cellular models and in dissociated SCN neurons. In summary, this study establishes new cell-autonomous clock models that are of particular relevance to metabolism and suitable for screening for clock modifiers, and reveals previously under-appreciated cell type-specific functions of clock genes.


Subject(s)
Adipocytes/metabolism , Circadian Clocks/genetics , Circadian Rhythm/genetics , Hepatocytes/metabolism , Animals , Cell Line , Luciferases/genetics , Mice , Mice, Knockout , NIH 3T3 Cells , Neurons/metabolism , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism
14.
J Vis Exp ; (67)2012 Sep 27.
Article in English | MEDLINE | ID: mdl-23052244

ABSTRACT

In mammals, many aspects of behavior and physiology such as sleep-wake cycles and liver metabolism are regulated by endogenous circadian clocks (reviewed). The circadian time-keeping system is a hierarchical multi-oscillator network, with the central clock located in the suprachiasmatic nucleus (SCN) synchronizing and coordinating extra-SCN and peripheral clocks elsewhere. Individual cells are the functional units for generation and maintenance of circadian rhythms, and these oscillators of different tissue types in the organism share a remarkably similar biochemical negative feedback mechanism. However, due to interactions at the neuronal network level in the SCN and through rhythmic, systemic cues at the organismal level, circadian rhythms at the organismal level are not necessarily cell-autonomous. Compared to traditional studies of locomotor activity in vivo and SCN explants ex vivo, cell-based in vitro assays allow for discovery of cell-autonomous circadian defects. Strategically, cell-based models are more experimentally tractable for phenotypic characterization and rapid discovery of basic clock mechanisms. Because circadian rhythms are dynamic, longitudinal measurements with high temporal resolution are needed to assess clock function. In recent years, real-time bioluminescence recording using firefly luciferase as a reporter has become a common technique for studying circadian rhythms in mammals, as it allows for examination of the persistence and dynamics of molecular rhythms. To monitor cell-autonomous circadian rhythms of gene expression, luciferase reporters can be introduced into cells via transient transfection or stable transduction. Here we describe a stable transduction protocol using lentivirus-mediated gene delivery. The lentiviral vector system is superior to traditional methods such as transient transfection and germline transmission because of its efficiency and versatility: it permits efficient delivery and stable integration into the host genome of both dividing and non-dividing cells. Once a reporter cell line is established, the dynamics of clock function can be examined through bioluminescence recording. We first describe the generation of P(Per2)-dLuc reporter lines, and then present data from this and other circadian reporters. In these assays, 3T3 mouse fibroblasts and U2OS human osteosarcoma cells are used as cellular models. We also discuss various ways of using these clock models in circadian studies. Methods described here can be applied to a great variety of cell types to study the cellular and molecular basis of circadian clocks, and may prove useful in tackling problems in other biological systems.


Subject(s)
Circadian Clocks/genetics , Gene Expression Profiling/methods , Luciferases/genetics , Luminescent Measurements/methods , 3T3 Cells , Animals , Cell Line, Tumor , HEK293 Cells , Humans , Lentivirus/genetics , Luciferases/biosynthesis , Luciferases/chemistry , Mice , Promoter Regions, Genetic
15.
Neurosci Lett ; 473(3): 220-3, 2010 Apr 12.
Article in English | MEDLINE | ID: mdl-20188794

ABSTRACT

The suprachiasmatic nucleus (SCN) of the hypothalamus is the central pacemaker that controls circadian rhythms in mammals. In diurnal grass rats (Arvicanthis niloticus), many functional aspects of the SCN are similar to those of nocturnal rodents, making it likely that the difference in the circadian system of diurnal and nocturnal animals lies downstream from the SCN. Rhythms in clock genes expression occur in several brain regions outside the SCN that may function as extra-SCN oscillators. In male grass rats PER1 is expressed in the oval nucleus of the bed nucleus of the stria terminalis (BNST-ov) and in the central and basolateral amygdala (CEA and BLA, respectively); several features of PER1 expression in these regions of the grass rat brain differ substantially from those of nocturnal species. Here we describe PER2 rhythms in the same three brain regions of the grass rat. In the BNST-ov and CEA PER2 expression peaked early in the light period Zeitgeber time (ZT) 2 and was low during the early night, which is the reverse of the pattern of nocturnal rodents. In the BLA, PER2 expression was relatively low for most of the 24-h cycle, but showed an acute elevation late in the light period (ZT10). This pattern is also different from that of nocturnal rodents that show elevated PER2 expression in the mid to late night and into the early day. These results are consistent with the hypothesis that diurnal behavior is associated with a phase change between the SCN and extra-SCN oscillators.


Subject(s)
Amygdala/metabolism , Circadian Rhythm , Period Circadian Proteins/biosynthesis , Septal Nuclei/metabolism , Animals , Male , Murinae
16.
Brain Behav Evol ; 75(1): 9-22, 2010.
Article in English | MEDLINE | ID: mdl-20134153

ABSTRACT

Evolutionary transitions between nocturnal and diurnal patterns of adaptation to the day-night cycle must have involved fundamental changes in the neural mechanisms that coordinate the daily patterning of activity, but little is known about how these mechanisms differ. One reason is that information on these systems in very closely related diurnal and nocturnal species is lacking. In this study, we characterize the suprachiasmatic nucleus (SCN), the primary brain structure involved in the generation and coordination of circadian rhythms, in two members of the genus Acomys with very different activity patterns, Acomys russatus (the golden spiny mouse, diurnal) and Acomys cahirinus (the common spiny mouse, nocturnal). Immunohistochemical techniques were used to label cell bodies containing vasoactive intestinal polypeptide (VIP), vasopressin (VP), gastrin-releasing peptide (GRP) and calbindin (CalB) in the SCN, as well as two sets of inputs to it, those containing serotonin (5-HT) and neuropeptide Y (NPY), respectively. All were present in the SCN of both species and no differences between them were seen. On the basis of neuronal phenotype, the SCN was organized into three basic regions that contained VIP-immunoreactive (-ir), CalB-ir and VP-ir cells, in the ventral, middle and dorsal SCN, respectively. In the rostral SCN, GRP-ir cells were in both the VIP and the CalB cell regions, and in the caudal area they were distributed across a portion of each of the other three regions. Fibers containing NPY-ir and serotonin (5-HT)-ir were most concentrated in the areas containing VIP-ir and CalB-ir cells, respectively. The details of the spatial relationships among the labeled cells and fibers seen here are discussed in relation to different approaches investigators have taken to characterize the SCN more generally.


Subject(s)
Circadian Rhythm/physiology , Neuropeptides/metabolism , Neurotransmitter Agents/metabolism , Photoperiod , Suprachiasmatic Nucleus/cytology , Suprachiasmatic Nucleus/physiology , Adaptation, Physiological/physiology , Animals , Calbindins , Female , Gastrin-Releasing Peptide/metabolism , Immunohistochemistry , Male , Mice , Neural Pathways/cytology , Neural Pathways/metabolism , Neurons/metabolism , Neuropeptide Y/metabolism , S100 Calcium Binding Protein G/metabolism , Serotonin/metabolism , Vasoactive Intestinal Peptide/metabolism , Vasopressins/metabolism
17.
Invest Ophthalmol Vis Sci ; 51(1): 327-34, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19710411

ABSTRACT

PURPOSE: To determine whether application of BDNF to the eye and brain provides a greater level of neuroprotection after optic nerve injury than treatment of the eye alone. METHODS: Retinal ganglion cell survival and pattern electroretinographic responses were compared in normal cat eyes and in eyes that received (1) a mild nerve crush and no treatment, (2) a single intravitreal injection of BDNF at the time of the nerve injury, or (3) intravitreal treatment combined with 1 to 2 weeks of continuous delivery of BDNF to the visual cortex, bilaterally. RESULTS: Relative to no treatment, administration of BDNF to the eye alone resulted in a significant increase in ganglion cell survival at both 1 and 2 weeks after nerve crush (1 week, 79% vs. 55%; 2 weeks, 60% vs. 31%). Combined treatment of the eye and visual cortex resulted in a modest additional increase (17%) in ganglion cell survival in the 1-week eyes, a further significant increase (55%) in the 2-week eyes, and ganglion cell survival levels for both that were comparable to normal (92%-93% survival). Pattern ERG responses for all the treated eyes were comparable to normal at 1 week after injury; however, at 2 weeks, only the responses of eyes receiving the combined BDNF treatment remained so. CONCLUSIONS: Although treatment of the eye alone with BDNF has a significant impact on ganglion cell survival after optic nerve injury, combined treatment of the eye and brain may represent an even more effective approach and should be considered in the development of future optic neuropathy-related neuroprotection strategies.


Subject(s)
Brain-Derived Neurotrophic Factor/administration & dosage , Neuroprotective Agents/administration & dosage , Retinal Degeneration/prevention & control , Retinal Ganglion Cells/drug effects , Visual Cortex/drug effects , Vitreous Body/drug effects , Animals , Cats , Cell Count , Cell Survival/drug effects , Disease Models, Animal , Electroretinography , Female , Injections , Male , Nerve Crush , Optic Nerve Injuries/physiopathology , Recombinant Proteins/administration & dosage , Retinal Degeneration/physiopathology , Retinal Ganglion Cells/physiology
18.
Neurosci Lett ; 441(1): 86-9, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18599213

ABSTRACT

In the diurnal rodent Arvicanthis niloticus (grass rats) the pattern of expression of the clock genes and their proteins in the suprachiasmatic nucleus (SCN) is very similar to that seen in nocturnal rodents. Rhythms in clock gene expression have been also documented in several forebrain regions outside the SCN in nocturnal Ratus norvegicus (lab rats). To investigate the neural basis for differences in the circadian systems of diurnal and nocturnal mammals, we examined PER1 expression in the oval nucleus of the bed nucleus of the stria terminalis (BNST-OV), and in the basolateral (BLA) and the central (CEA) amygdala of male grass rats kept in a 12:12 light/dark cycle. In the BNST-OV, peak levels of PER1 expression were seen early in the light phase of the cycle, 12h out of phase with what has been reported for nocturnal lab rats. In the BLA the pattern of PER1 expression featured sustained high levels during the day and low levels at night. PER1 expression in the CEA was also at its highest early in the light phase, but the effect of sampling time was not statistically significant (p<0.06). The results are consistent with the hypothesis that differences between nocturnal and diurnal species are due to differences in neural systems downstream from the SCN.


Subject(s)
Circadian Rhythm/physiology , Gene Expression Regulation/physiology , Nerve Tissue Proteins/metabolism , Septal Nuclei/metabolism , Animals , Arvicolinae , Cell Nucleus/metabolism , Male , Nerve Tissue Proteins/genetics , Septal Nuclei/cytology
19.
J Chem Neuroanat ; 33(3): 131-9, 2007 May.
Article in English | MEDLINE | ID: mdl-17368836

ABSTRACT

Diurnal and nocturnal animals differ with respect to the timing of a host of behavioral and physiological events including those associated with neuroendocrine functions, but the neural bases of these differences are poorly understood. In nocturnal species, rhythms in tyrosine hydroxylase-containing (TH+) neurons in the hypothalamus appear to be responsible for rhythms in prolactin secretion. Here we investigated TH+ cells in a diurnal rodent (Arvicanthis niloticus, the unstriped Nile grass rat), and comparing them with those of a nocturnal rodent (Rattus norvegicus, Sprague-Dawley rat). We also examined relationships between TH+ cells and fibers containing vasoactive intestinal polypeptide (VIP) that are thought to originate from cells in the suprachiasmatic nucleus (SCN), the site of the primary circadian clock in mammals. The distribution of TH+ neurons was very similar in the two species except for a population of cells in the basal forebrain that was only present in grass rats. Fibers containing VIP appeared to contact neuroendocrine TH+ cells in both species. These data suggest that, though there may be subtle species differences, temporal information is likely to be carried along the same direct pathways from the SCN to the TH+ neurons in day- and night-active species.


Subject(s)
Circadian Rhythm/physiology , Hypothalamus/cytology , Suprachiasmatic Nucleus/cytology , Tyrosine 3-Monooxygenase/metabolism , Vasoactive Intestinal Peptide/metabolism , Animals , Biomarkers/metabolism , Female , Hypothalamus/metabolism , Murinae , Nerve Fibers/metabolism , Neural Pathways , Neurons/metabolism , Neurosecretory Systems/cytology , Neurosecretory Systems/metabolism , Rats , Rats, Sprague-Dawley , Species Specificity , Suprachiasmatic Nucleus/metabolism
20.
Brain Res ; 1073-1074: 348-58, 2006 Feb 16.
Article in English | MEDLINE | ID: mdl-16430875

ABSTRACT

The suprachiasmatic nucleus (SCN) of the hypothalamus contains the primary circadian pacemaker in both diurnal and nocturnal mammals. The lower subparaventricular zone (LSPV) immediately dorsal to the SCN may also play an important role in the regulation of circadian rhythms. The SCN contains a multitude of oscillator cells that generate circadian rhythms through transcriptional/translational feedback loops involving a set of clock genes including per1 and per2. Little is known about the temporal and spatial features of the proteins encoded by these genes in day-active mammals. The first objective of this study was to characterize the expression of PER1 and PER2 in the SCN of a diurnal rodent, the unstriped Nile grass rat (Arvicanthis niloticus). The second objective was to evaluate the hypothesis that a molecular clock could exist in the LSPV, where endogenous rhythms in Fos expression are seen in grass rats but not in laboratory rats. Animals were kept on a 12:12 light/dark cycle and perfused at 4-h intervals, and their brains were processed for immunohistochemical detection of PER1 and PER2. Both proteins were seen in the SCN where they peaked early in the dark phase, providing further evidence that the differences between diurnal and nocturnal patterns of behavior emerge from mechanisms lying downstream from the pacemaker within the SCN. Rhythmic expression of PER1 and PER2 was also seen in the LSPV providing support for the hypothesis that this region might participate in circadian time keeping in the diurnal grass rat. In addition, rhythms were seen lateral to the LSPV and the SCN. Results of this study are discussed in light of similarities and differences in the circadian time-keeping systems of day- and night-active animals.


Subject(s)
Circadian Rhythm/physiology , Nuclear Proteins/metabolism , Suprachiasmatic Nucleus/metabolism , Transcription Factors/metabolism , Analysis of Variance , Animals , Brain Mapping , Cell Count/methods , Cell Cycle Proteins , Gene Expression Regulation/physiology , Immunohistochemistry/methods , Male , Neurons/metabolism , Period Circadian Proteins , Rats , Suprachiasmatic Nucleus/cytology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...