Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Peptides ; 176: 171213, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38604379

ABSTRACT

Glucagon is best known for its contribution to glucose regulation through activation of the glucagon receptor (GCGR), primarily located in the liver. However, glucagon's impact on other organs may also contribute to its potent effects in health and disease. Given that glucagon-based medicine is entering the arena of anti-obesity drugs, elucidating extrahepatic actions of glucagon are of increased importance. It has been reported that glucagon may stimulate secretion of arginine-vasopressin (AVP)/copeptin, growth hormone (GH) and adrenocorticotrophic hormone (ACTH) from the pituitary gland. Nevertheless, the mechanisms and whether GCGR is present in human pituitary are unknown. In this study we found that intravenous administration of 0.2 mg glucagon to 14 healthy subjects was not associated with increases in plasma concentrations of copeptin, GH, ACTH or cortisol over a 120-min period. GCGR immunoreactivity was present in the anterior pituitary but not in cells containing GH or ACTH. Collectively, glucagon may not directly stimulate secretion of GH, ACTH or AVP/copeptin in humans but may instead be involved in yet unidentified pituitary functions.


Subject(s)
Adrenocorticotropic Hormone , Glucagon , Glycopeptides , Humans , Glycopeptides/metabolism , Glucagon/metabolism , Glucagon/blood , Adrenocorticotropic Hormone/blood , Adrenocorticotropic Hormone/metabolism , Male , Adult , Female , Pituitary Gland/metabolism , Pituitary Gland/drug effects , Hydrocortisone/blood , Receptors, Glucagon/metabolism , Human Growth Hormone/metabolism , Growth Hormone/metabolism , Growth Hormone/blood , Middle Aged
2.
Int J Mol Sci ; 24(20)2023 Oct 12.
Article in English | MEDLINE | ID: mdl-37894782

ABSTRACT

Pituitary adenylate cyclase-activating polypeptide (PACAP) is a naturally occurring neuropeptide found in both the central and peripheral nervous systems of vertebrates. Recent studies have revealed the presence of PACAP and its corresponding receptors, namely, the pituitary adenylate cyclase-activating polypeptide type I receptor (PAC1R), vasoactive intestinal peptide receptor 1 (VIPR1), and vasoactive intestinal peptide receptor 2 (VIPR2), in various structures implicated in migraine pathophysiology, including sensory trigeminal neurons. Human studies have demonstrated that when infused, PACAP can cause dilation of cranial vessels and result in delayed migraine-like attacks. In light of this, we present a novel ELISA assay that has been validated for quantifying PACAP in tissue extracts and human plasma. Using two well characterized antibodies specifically targeting PACAP, we successfully developed a sandwich ELISA assay, capable of detecting and accurately quantifying PACAP without any cross-reactivity to closely related peptides. The quantification range was between 5.2 pmol/L and 400 pmol/L. The recovery in plasma ranged from 98.2% to 100%. The increasing evidence pointing to the crucial role of PACAP in migraine pathophysiology necessitates the availability of tools capable of detecting changes in the circulatory levels of PACAP and its potential application as a reliable biomarker.


Subject(s)
Migraine Disorders , Pituitary Adenylate Cyclase-Activating Polypeptide , Animals , Humans , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Vasoactive Intestinal Polypeptide, Type I , Receptors, Vasoactive Intestinal Peptide, Type II , Mammals , Enzyme-Linked Immunosorbent Assay , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Vasoactive Intestinal Peptide
3.
Front Physiol ; 14: 1321007, 2023.
Article in English | MEDLINE | ID: mdl-38317846

ABSTRACT

Light-sensitive neurons are located in the ventral and central core of the suprachiasmatic nucleus (SCN), whereas stably oscillating clock neurons are found mainly in the dorsal shell. Signals between the SCN core and shell are believed to play an important role in light entrainment. Core neurons express vasoactive intestinal polypeptide (VIP), gastrin-releasing peptide (GRP), and Neuroglobin (Ngb), whereas the shell neurons express vasopressin (AVP), prokineticin 2, and the VIP type 2 (VPAC2) receptor. In rodents, light has a phase-shifting capacity at night, which induces rapid and transient expression of the EGR1 and FOS in the SCN. Methods: The present study used immunohistochemical staining of FOS, EGR1, and phenotypical markers of SCN neurons (VIP, AVP, Ngb) to identify subtypes/populations of light-responsive neurons at early night. Results: Double immunohistochemistry and cell counting were used to evaluate the number of SCN neurons expressing FOS and EGR1 in the SCN. The number of neurons expressing either EGR1 or FOS was higher than the total number of neurons co-storing EGR1 and FOS. Of the total number of light-responsive cells, 42% expressed only EGR1, 43% expressed only FOS, and 15% expressed both EGR1 and FOS. Light-responsive VIP neurons represented only 31% of all VIP neurons, and EGR1 represents the largest group of light-responsive VIP neurons (18%). VIP neurons expressing only FOS represented 1% of the total light-responsive VIP neurons. 81% of the Ngb neurons in the mouse SCN were light-responsive, and of these neurons expressing only EGR1 after light stimulation represented 44%, whereas 24% expressed FOS. Although most light-responsive neurons are found in the core of the SCN, 29% of the AVP neurons in the shell were light-responsive, of which 8% expressed EGR1, 10% expressed FOS, and 11% co-expressed both EGR1 and FOS after light stimulation. Discussion: Our analysis revealed cell-specific differences in light responsiveness between different peptidergic and Ngb-expressing neurons in different compartments of the mouse SCN, indicating that light activates diverse neuronal networks in the SCN, some of which participate in photoentrainment.

4.
Sci Adv ; 8(14): eabc9061, 2022 Apr 08.
Article in English | MEDLINE | ID: mdl-35394844

ABSTRACT

The circadian clock controls many aspects of physiology, but it remains undescribed whether extracellular vesicles (EVs), including exosomes, involved in cell-cell communications between tissues are regulated in a circadian pattern. We demonstrate a 24-hour rhythmic abundance of individual proteins in small EVs using liquid chromatography-mass spectrometry in circadian-synchronized tendon fibroblasts. Furthermore, the release of small EVs enriched in RNA binding proteins was temporally separated from those enriched in cytoskeletal and matrix proteins, which peaked during the end of the light phase. Last, we targeted the protein sorting mechanism in the exosome biogenesis pathway and established (by knockdown of circadian-regulated flotillin-1) that matrix metalloproteinase 14 abundance in tendon fibroblast small EVs is under flotillin-1 regulation. In conclusion, we have identified proteomic time signatures for small EVs released by tendon fibroblasts, which supports the view that the circadian clock regulates protein cargo in EVs involved in cell-cell cross-talk.

5.
Front Endocrinol (Lausanne) ; 12: 737581, 2021.
Article in English | MEDLINE | ID: mdl-34539582

ABSTRACT

VIP/VPAC2-receptor signaling is crucial for functioning of the circadian clock in the suprachiasmatic nucleus (SCN) since the lack results in disrupted synchrony between SCN cells and altered locomotor activity, body temperature, hormone secretion and heart rhythm. Endocrine glands, including the thyroid, show daily oscillations in clock gene expression and hormone secretion, and SCN projections target neurosecretory hypothalamic thyroid-stimulating hormone (TSH)-releasing hormone cells. The aim of the study was to gain knowledge of mechanisms important for regulation of the thyroid clock by evaluating the impact of VIP/VPAC2-receptor signaling. Quantifications of mRNAs of three clock genes (Per1, Per2 and Bmal1) in thyroids of wild type (WT) and VPAC2-receptor deficient mice were done by qPCR. Tissues were taken every 4th h during 24-h 12:12 light-dark (LD) and constant darkness (DD) periods, both genders were used. PER1 immunoreactivity was visualized on sections of both WT and VPAC2 lacking mice during a LD cycle. Finally, TSH and the thyroid hormone T4 levels were measured in the sera by commercial ELISAs. During LD, rhythmic expression of all three mRNA was found in both the WT and knockout animals. In VPAC2-receptor knockout animals, the amplitudes were approximately halved compared to the ones in the WT mice. In the WT, Per1 mRNA peaked around "sunset", Per2 mRNA followed with approximately 2 h, while Bmal1 mRNA was in antiphase with Per1. In the VPAC2 knockout mice, the phases of the mRNAs were advanced approximately 5 h compared to the WT. During DD, the phases of all the mRNAs were identical to the ones found during LD in both groups of mice. PER1 immunoreactivity was delayed compared to its mRNA and peaked during the night in follicular cells of both the thyroid and parathyroid glands in the WT animals. In WT animals, TSH was high around the transition to darkness compared to light-on, while T4 did not change during the 24 h cycle. In conclusion, sustained and identical rhythms (phases and amplitudes) of three clock genes were found in VPAC2 deficient mice during LD and DD suggesting high degree of independence of the thyroid clock from the master SCN clock.


Subject(s)
Circadian Clocks/physiology , Receptors, Vasoactive Intestinal Peptide, Type II/genetics , Thyroid Gland/metabolism , Animals , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Circadian Rhythm/physiology , Female , Male , Mice , Mice, Knockout , Receptors, Vasoactive Intestinal Peptide, Type II/metabolism , Thyrotropin, beta Subunit/blood
6.
PLoS One ; 15(5): e0232748, 2020.
Article in English | MEDLINE | ID: mdl-32379800

ABSTRACT

The brain's biological clock is located in the suprachiasmatic nucleus (SCN) of the hypothalamus and generates circadian rhythms in physiology and behavior. The circadian clock needs daily adjustment by light to stay synchronized (entrained) with the astronomical 24 h light/dark cycle. Light entrainment occurs via melanopsin expressing retinal ganglion cells (mRGCs) and two neurotransmitters of the retinohypothalamic tract (RHT), PACAP and glutamate, which transmit light information to the SCN neurons. In SCN neurons, light signaling involves the immediate-early genes Fos, Egr1 and the clock genes Per1 and Per2. In this study, we used PACAP deficient mice to evaluate PACAP's role in light induced gene expression of EGR1 in SCN neurons during early (ZT17) and late (ZT23) subjective night at high (300 lux) and low (10 lux) white light exposure. We found significantly lower levels of both EGR1 mRNA and protein in the SCN in PACAP deficient mice compared to wild type mice at early subjective night (ZT17) exposed to low but not high light intensity. No difference was found between the two genotypes at late night (ZT23) at neither light intensities. In conclusion, light mediated EGR1 induction in SCN neurons at early night at low light intensities is dependent of PACAP signaling. A role of PACAP in shaping synaptic plasticity during light stimulation at night is discussed.


Subject(s)
Early Growth Response Protein 1/genetics , Gene Expression Regulation , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Suprachiasmatic Nucleus/metabolism , Animals , Circadian Rhythm , Female , Light , Male , Mice , Photoperiod , Suprachiasmatic Nucleus/cytology
7.
Eur J Neurosci ; 50(7): 3115-3132, 2019 10.
Article in English | MEDLINE | ID: mdl-31211910

ABSTRACT

Neurons of the hypothalamic suprachiasmatic nucleus (SCN) express clock genes, which regulate their own transcription and generate daily output signals driving circadian rhythmic behavior and physiology. The neuropeptide VIP and its specific receptor, the VPAC2 receptor, are important for synchronization of clock neurons. In the present study, we characterized PER1 and PER2 expressing neurons in wild-type and VPAC2-deficient mice. We found evidence for distinct spatiotemporal circadian oscillation in the expression of the PER genes in two separate clusters of SCN neurons. In wild-type mice corresponding to the SCN shell and ventral core, high expression of PER was found at lights-off most likely representing an evening clock (E-clock). In another smaller cluster of neurons located in the central core of the SCN, PER expression peaks in antiphase at lights-on and could represent a morning clock (M-clock). BMAL1 immunoreactivity was found to be expressed in antiphase to PER in M and E neurons, respectively. PER was found in 98% of neurons expressing vasopressin (AVP) and in 92% of VIP neurons. The chemotype of M neurons was not identified. M but not E cells were responsive to long but not short photoperiods. The expression of the VPAC2 receptor was found in both M and E cells, and VPAC2-deficient mice displayed markedly blunted PER expression in both cell clusters of the SCN. Conclusion: These observations support the existence of M and E clocks involved in circadian and seasonal adaptation, which seem dependent on intact VIP/VPAC2 signaling in the SCN.


Subject(s)
Circadian Rhythm/physiology , Neurons/metabolism , Period Circadian Proteins/metabolism , Receptors, Vasoactive Intestinal Peptide, Type II/metabolism , Suprachiasmatic Nucleus/metabolism , Animals , Female , Male , Mice, Knockout , Receptors, Vasoactive Intestinal Peptide, Type II/genetics , Signal Transduction
8.
Exp Eye Res ; 169: 134-140, 2018 04.
Article in English | MEDLINE | ID: mdl-29428294

ABSTRACT

The intraocular pressure of mice displays a daily rhythmicity being highest during the dark period. The present study was performed to elucidate the role of the circadian clock and light in the diurnal and the circadian variations in intraocular pressure in mice, by using animals with disrupted clock function (VPAC2 receptor knockout mice) or impaired light information to the clock (PACAP knockout mice). In wildtype mice, intraocular pressure measured under light/dark conditions showed a statistically significant 24 h sinusoidal rhythm with nadir during the light phase and peak during the dark phase. After transfer of the wildtype mice into constant darkness, the intraocular pressure increased, but the rhythmic changes in intraocular pressure continued with a pattern identical to that obtained during the light/dark cycle. The intraocular pressure in VPAC2 receptor deficient mice during light/dark conditions also showed a sinusoidal pattern with significant changes as a function of a 24 h cycle. However, transfer of the VPAC2 receptor knockout mice into constant darkness completely abolished the rhythmic changes in intraocular pressure. The intraocular pressure in PACAP deficient mice oscillated significantly during both 24 h light and darkness and during constant darkness. During LD conditions, the amplitude of PACAP deficient was significantly lower compared to wildtype mice, resulting in higher daytime and lower nighttime values. In conclusion, by studying the VPAC2 receptor knockout mouse which lacks circadian control and the PACAP knockout mouse which displays impaired light signaling, we provided evidence that the daily intraocular pressure rhythms are primarily generated by the circadian master clock and to a lesser extent by environmental light and darkness.


Subject(s)
Circadian Clocks/physiology , Circadian Rhythm/physiology , Intraocular Pressure/physiology , Light , Pituitary Adenylate Cyclase-Activating Polypeptide/deficiency , Receptors, Vasoactive Intestinal Peptide, Type II/deficiency , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Pituitary Adenylate Cyclase-Activating Polypeptide/physiology , Receptors, Vasoactive Intestinal Peptide, Type II/physiology , Tonometry, Ocular
9.
J Mol Neurosci ; 64(1): 9-19, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29138967

ABSTRACT

Early growth response transcription factor 1 (EGR1) is expressed in the suprachiasmatic nucleus (SCN) after light stimulation. We used EGR1-deficient mice to address the role of EGR1 in the clock function and light-induced resetting of the clock. The diurnal rhythms of expression of the clock genes BMAL1 and PER1 in the SCN were evaluated by semi-quantitative in situ hybridization. We found no difference in the expression of PER1 mRNA between wildtype and EGR1-deficient mice; however, the daily rhythm of BMAL1 mRNA was completely abolished in the EGR1-deficient mice. In addition, we evaluated the circadian running wheel activity, telemetric locomotor activity, and core body temperature of the mice. Loss of EGR1 neither altered light-induced phase shifts at subjective night nor affected negative masking. Overall, circadian light entrainment was found in EGR1-deficient mice but they displayed a reduced locomotor activity and an altered temperature regulation compared to wild type mice. When placed in running wheels, a subpopulation of EGR1-deficient mice displayed a more disrupted activity rhythm with no measurable endogenous period length (tau). In conclusion, the present study provides the first evidence that the circadian clock in the SCN is disturbed in mice deficient of EGR1.


Subject(s)
ARNTL Transcription Factors/metabolism , Body Temperature , Circadian Rhythm , Early Growth Response Protein 1/genetics , Locomotion , ARNTL Transcription Factors/genetics , Animals , Early Growth Response Protein 1/deficiency , Early Growth Response Protein 1/metabolism , Female , Male , Mice , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism
10.
PLoS One ; 12(11): e0188166, 2017.
Article in English | MEDLINE | ID: mdl-29155851

ABSTRACT

The two sister peptides, pituitary adenylate cyclase activating polypeptide (PACAP) and vasoactive intestinal polypeptide (VIP) and their receptors, the PAC1 -and the VPAC2 receptors, are involved in regulation of the circadian timing system. PACAP as a neurotransmitter in the retinohypothalamic tract (RHT) and VIP as a neurotransmitter, involved in synchronization of SCN neurons. Behavior and physiology in VPAC2 deficient mice are strongly regulated by light most likely as a result of masking. Consequently, we used VPAC2 and PAC1/VPAC2 double mutant mice in comparison with PAC1 receptor deficient mice to further elucidate the role of PACAP in the light mediated regulation of behavior and physiology of the circadian system. We compared circadian rhythms in mice equipped with running wheels or implanted radio-transmitter measuring core body temperature kept in a full photoperiod ((FPP)(12:12 h light dark-cycles (LD)) and skeleton photo periods (SPP) at high and low light intensity. Furthermore, we examined the expression of PAC1- and VPAC2 receptors in the SCN of the different genotypes in combination with visualization of PACAP and VIP and determined whether compensatory changes in peptide and/or receptor expression in the reciprocal knockouts (KO) (PAC1 and VPAC2) had occurred. Our data demonstrate that in although being closely related at both ligand and receptor structure/sequence, PACAP/PAC1 receptor signaling are independent of VIP/VPAC2 receptor signaling and vice versa. Furthermore, lack of either of the receptors does not result in compensatory changes at neither the physiological or anatomical level. PACAP/PAC1 signaling is important for light regulated behavior, VIP/VPAC2signaling for stable clock function and both signaling pathways may play a role in shaping diurnality versus nocturnality.


Subject(s)
Circadian Rhythm/physiology , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Receptors, Vasoactive Intestinal Peptide, Type II/genetics , Vasoactive Intestinal Peptide/genetics , Amino Acid Sequence , Animals , Body Temperature/physiology , Female , Gene Expression Regulation , Light , Light Signal Transduction , Male , Mice , Mice, Knockout , Photoperiod , Physical Conditioning, Animal , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/deficiency , Receptors, Vasoactive Intestinal Peptide, Type II/deficiency , Running , Vasoactive Intestinal Peptide/metabolism
11.
Mitochondrion ; 36: 77-84, 2017 09.
Article in English | MEDLINE | ID: mdl-28412540

ABSTRACT

Melanopsin retinal ganglion cells (mRGCs) are intrinsically photosensitive RGCs deputed to non-image forming functions of the eye such as synchronization of circadian rhythms to light-dark cycle. These cells are characterized by unique electrophysiological, anatomical and biochemical properties and are usually more resistant than conventional RGCs to different insults, such as axotomy and different paradigms of stress. We also demonstrated that these cells are relatively spared compared to conventional RGCs in mitochondrial optic neuropathies (Leber's hereditary optic neuropathy and Dominant Optic Atrophy). However, these cells are affected in other neurodegenerative conditions, such as glaucoma and Alzheimer's disease. We here review the current evidences that may underlie this dichotomy. We also present our unpublished data on cell experiments demonstrating that melanopsin itself does not explain the robustness of these cells and some preliminary data on immunohistochemical assessment of mitochondria in mRGCs.


Subject(s)
Gene Expression , Mitochondrial Diseases/pathology , Optic Nerve Diseases/pathology , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/physiology , Rod Opsins/biosynthesis , Stress, Physiological , Humans
12.
Neuropeptides ; 60: 83-89, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27745706

ABSTRACT

The neuropeptide PACAP is expressed throughout the central and peripheral nervous system where it modulates diverse physiological functions including neuropeptide gene expression. We here report that in human neuroblastoma NB-1 cells PACAP transiently induces its own expression. Maximal PACAP mRNA expression was found after stimulation with PACAP for 3h. PACAP auto-regulation was found to be mediated by activation of PACAP specific PAC1Rs as PACAP had >100-fold higher efficacy than VIP, and the PAC1R selective agonist Maxadilan potently induced PACAP gene expression. Experiments with pharmacological kinase inhibitors revealed that both PKA and novel but not conventional PKC isozymes were involved in the PACAP auto-regulation. Inhibition of MAPK/ERK kinase (MEK) also impeded the induction, and we found that PKA, novel PKC and ERK acted in parallel and were thus not part of the same pathways. The expression of the transcription factor EGR1 previously ascribed as target of PACAP signalling was found to be transiently induced by PACAP and pharmacological inhibition of either PKC or MEK1/2 abolished PACAP mediated EGR1 induction. In contrast, inhibition of PKA mediated increased PACAP mediated EGR1 induction. Experiments using siRNA against EGR1 to lower the expression did however not affect the PACAP auto-regulation indicating that this immediate early gene product is not part of PACAP auto-regulation in NB-1 cells. We here reveal that in NB-1 neuroblastoma cells, PACAP induces its own expression by activation of PAC1R, and that the signalling is different from the PAC1R signalling mediating induction of VIP in the same cells. PACAP auto-regulation depends on parallel activation of PKA, novel PKC isoforms, and ERK, while EGR1 does not seem to be part of the PACAP auto-regulation.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Homeostasis/physiology , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Protein Isoforms/metabolism , Protein Kinase C/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Brain Neoplasms/metabolism , Cell Line, Tumor , Homeostasis/drug effects , Humans , Neuroblastoma/metabolism , Neurons/drug effects , Neurons/metabolism , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects
13.
PLoS One ; 11(1): e0146981, 2016.
Article in English | MEDLINE | ID: mdl-26757053

ABSTRACT

Light signals from intrinsically photosensitive retinal ganglion cells (ipRGCs) entrain the circadian clock and regulate negative masking. Two neurotransmitters, glutamate and Pituitary Adenylate Cyclase Activating Polypeptide (PACAP), found in the ipRGCs transmit light signals to the brain via glutamate receptors and the specific PACAP type 1 (PAC1) receptor. Light entrainment occurs during the twilight zones and has little effect on clock phase during daytime. When nocturnal animals have access to food only for a few hours during the resting phase at daytime, they adapt behavior to the restricted feeding (RF) paradigm and show food anticipatory activity (FAA). A recent study in mice and rats demonstrating that light regulates FAA prompted us to investigate the role of PACAP/PAC1 signaling in the light mediated regulation of FAA. PAC1 receptor knock out (PAC1-/-) and wild type (PAC1+/+) mice placed in running wheels were examined in a full photoperiod (FPP) of 12:12 h light/dark (LD) and a skeleton photoperiod (SPP) 1:11:1:11 h L:DD:L:DD at 300 and 10 lux light intensity. Both PAC1-/- mice and PAC1+/+ littermates entrained to FPP and SPP at both light intensities. However, when placed in RF with access to food for 4-5 h during the subjective day, a significant change in behavior was observed in PAC1-/- mice compared to PAC1+/+ mice. While PAC1-/- mice showed similar FAA as PAC1+/+ animals in FPP at 300 lux, PAC1-/- mice demonstrated an advanced onset of FAA with a nearly 3-fold increase in amplitude compared to PAC1+/+ mice when placed in SPP at 300 lux. The same pattern of FAA was observed at 10 lux during both FPP and SPP. The present study indicates a role of PACAP/PAC1 signaling during light regulated FAA. Most likely, PACAP found in ipRGCs mediating non-image forming light information to the brain is involved.


Subject(s)
Circadian Rhythm , Feeding Behavior , Pituitary Adenylate Cyclase-Activating Polypeptide/physiology , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Animals , Behavior, Animal , Brain/physiology , Female , Glutamic Acid/chemistry , Light , Male , Mice , Mice, Knockout , Motor Activity/physiology , Photoperiod , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/physiology , Retinal Ganglion Cells/cytology , Signal Transduction , Suprachiasmatic Nucleus/metabolism
14.
J Biol Chem ; 289(51): 35482-93, 2014 Dec 19.
Article in English | MEDLINE | ID: mdl-25378407

ABSTRACT

The G protein-coupled light-sensitive receptor melanopsin is involved in non-image-forming light responses including circadian timing. The predicted secondary structure of melanopsin indicates a long cytoplasmic tail with many potential phosphorylation sites. Using bioinformatics, we identified a number of amino acids with a high probability of being phosphorylated. We generated antibodies against melanopsin phosphorylated at Ser-381 and Ser-398, respectively. The antibody specificity was verified by immunoblotting and immunohistochemical staining of HEK-293 cells expressing rat melanopsin mutated in Ser-381 or Ser-398. Using the antibody recognizing phospho-Ser-381 melanopsin, we demonstrated by immunoblotting and immunohistochemical staining in HEK-293 cells expressing rat melanopsin that the receptor is phosphorylated in this position during the dark and dephosphorylated when light is turned on. On the contrary, we found that melanopsin at Ser-398 was unphosphorylated in the dark and became phosphorylated after light stimulation. The light-induced changes in phosphorylation at both Ser-381 and Ser-398 were rapid and lasted throughout the 4-h experimental period. Furthermore, phosphorylation at Ser-381 and Ser-398 was independent of each other. The changes in phosphorylation were confirmed in vivo by immunohistochemical staining of rat retinas during light and dark. We further demonstrated that mutation of Ser-381 and Ser-398 in melanopsin-expressing HEK-293 cells affected the light-induced Ca(2+) response, which was significantly reduced as compared with wild type. Examining the light-evoked Ca(2+) response in a melanopsin Ser-381 plus Ser-398 double mutant provided evidence that the phosphorylation events were independent.


Subject(s)
Calcium/metabolism , Retinal Ganglion Cells/metabolism , Rod Opsins/metabolism , Serine/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Calcium Signaling/radiation effects , Darkness , Eye/metabolism , Eye/radiation effects , HEK293 Cells , Humans , Immunohistochemistry , Light , Male , Microscopy, Confocal , Molecular Sequence Data , Mutation , Phosphorylation/radiation effects , Protein Structure, Secondary , Rats, Wistar , Retinal Ganglion Cells/radiation effects , Rod Opsins/chemistry , Rod Opsins/genetics , Serine/genetics
15.
Photochem Photobiol ; 90(5): 1069-76, 2014.
Article in English | MEDLINE | ID: mdl-24909488

ABSTRACT

We established a cell line (HEK-hMel) expressing melanopsin in a tetracycline dependent manner to elucidate new aspects of melanopsin's light response. Different light stimuli were evaluated using FOS expression as response parameter. Immunoblotting was used to evaluate expression of melanopsin and FOS and qPCR to quantify FOS mRNA responses. The magnitude of the FOS response was found to correlate with the amount of melanopsin expressed by the cells, and a transient FOS mRNA induction followed by FOS protein still elevated after 24 h of illumination was revealed. Exposing the cells to darkness after light resulted in reduction of the response compared to exposure to light solely showing dependency on continuous light. Increasing irradiances of blue light (480 nm) up to 10(11) quanta cm(-2)  s(-1) elicited steep increases in FOS mRNA, while increases between 10(12) and 5 × 10(13) quanta  cm(-2)  s(-1) resulted in equally high FOS expression. The HEK-hMel cells were used to characterize facets of melanopsin's light-induced FOS response not approachable in vivo. Novel information such as dependency of the FOS response on both melanopsin amount and light intensity in addition to a detailed time-course of both FOS mRNA and protein were revealed.


Subject(s)
Proto-Oncogene Proteins c-fos/genetics , RNA, Messenger/genetics , Rod Opsins/genetics , Dose-Response Relationship, Radiation , Gene Expression Regulation , HEK293 Cells , Humans , Light , Photochemical Processes , Photoperiod , Plasmids/chemistry , Plasmids/metabolism , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/metabolism , Radiation Dosage , Rod Opsins/metabolism , Signal Transduction , Transformation, Genetic
16.
Exp Eye Res ; 106: 55-63, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23187103

ABSTRACT

Melanopsin is expressed in a subpopulation of retinal ganglion cells rendering these cells intrinsically photosensitive (ipRGCs). The ipRGCs are the primary RGCs mediating light entrainment of the circadian clock and control of the pupillary light reflex, light regulated melatonin secretion and negative masking behaviour. Previous studies have demonstrated that melanopsin expression in albino rats is regulated by light and darkness. The present study was undertaken to study the influence of light and darkness during the circadian day and after extended periods of constant light and darkness on melanopsin expression in the pigmented retina of the Brown Norwegian rat (Rattus norvegicus). The diurnal and circadian expressions were examined in retinal extracts from rats euthanized every 4 h during a 24 h light/dark (LD) and a 24 h dark cycle (DD) using quantitative real-time PCR and Western blotting. To study whether light regulates melanopsin expression, rats were sacrificed after being placed in either constant light (LL) or darkness for 3 or 21 d. Flat mount retinas from animals kept during either LL or DD were also examined by immunohistochemistry. Melanopsin mRNA expression displayed a significant rhythmic change during the LD cycle with peak expression around dusk and nadir at dawn. Melanopsin protein also changed over the LD cycle with peak expression at the end of the night and nadir at dusk. Rhythmic expression of melanopsin mRNA but not melanopsin protein was found in constant darkness. After 3 or 21 d in either LL or DD melanopsin mRNA expression was unaltered. Melanopsin protein was at the same high level after 3 and 21 d in DD, whereas a significant decrease was found after prolonging the light period for 3 or 21 d. The change in melanopsin protein was primarily due to change in immunoreactivity in the dendritic processes. In conclusion we found that light and darkness are important for regulation of melanopsin protein expression whereas input from a retinal networks regulates melanopsin mRNA expression. It is likely to speculate that altered level of melanopsin is one way in which the retina adapts to environmental light and darkness conditions ensuring optimal light sensitivity for the transmission to the brain.


Subject(s)
Gene Expression Regulation/physiology , RNA, Messenger/metabolism , Retina/metabolism , Rod Opsins/genetics , Animals , Blotting, Western , Circadian Rhythm/physiology , Dark Adaptation , Immunohistochemistry , Light , Male , Rats , Rats, Inbred BN , Real-Time Polymerase Chain Reaction , Retina/radiation effects , Rod Opsins/metabolism
17.
Cell Tissue Res ; 350(3): 539-48, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22940729

ABSTRACT

Daily oscillations of clock genes have recently been demonstrated in the ovaries of several species. Clock gene knockout or mutant mice demonstrate a variety of reproductive defects. Accumulating evidence suggests that these rhythms act to synchronise the expression of specific ovarian genes to hypothalamo-pituitary signals and that they are regulated by one or both of the gonadotropins. The aim of this study has been to examine the spatio-temporal expression of the clock genes Per1 and Bmal1 during gonadotropin-independent and gonadotropin-dependent follicle development in the rat ovary. We have examined the ovaries of prepubertal rats, of prepubertal rats stimulated with equine chorionic gonadotropin (eCG)/human chorionic gonadotropin (hCG) and of hypophysectomised adult animals. Using quantitative reverse transcription with the polymerase chain reaction, in situ hybridisation histochemistry and immunohistochemistry, we have demonstrated that the expression of the two clock genes is low and arrhythmic in ovarian cells during early gonadotropin-independent follicle development in prepubertal animals and in hypophysectomised animals. We have also demonstrated that the expression of the clock genes becomes rhythmic following eCG stimulation in the theca interna cells and the secondary interstitial cells and that, following additional hCG stimulation, the expression of the clock genes also becomes rhythmic in the granulosa cells of preovulatory follicles. These findings link the initiation of clock gene rhythms in the rat ovary to the luteinising hormone receptor and suggest a functional link to androgen and progesterone production. In hypophysectomised animals, rhythmic clock gene expression is also observed in the corpora lutea and in secondary interstitial cells demonstrating that, in these compartments, entrainment of clock gene rhythms is gonadotropin-independent.


Subject(s)
ARNTL Transcription Factors/genetics , Gonadotropins/pharmacology , Ovarian Follicle/drug effects , Ovarian Follicle/physiology , Period Circadian Proteins/genetics , ARNTL Transcription Factors/biosynthesis , ARNTL Transcription Factors/metabolism , Animals , Circadian Rhythm/genetics , Circadian Rhythm/physiology , Female , Gene Expression Regulation, Developmental/drug effects , Horses , Humans , Hypophysectomy , Ovarian Follicle/growth & development , Ovarian Follicle/metabolism , Period Circadian Proteins/biosynthesis , Period Circadian Proteins/metabolism , Rats , Rats, Wistar
18.
J Mol Neurosci ; 48(3): 584-96, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22622901

ABSTRACT

The circadian time-keeping system consists of clocks in the suprachiasmatic nucleus (SCN) and in peripheral organs including an adrenal clock linked to the rhythmic corticosteroid production by regulating steroidogenic acute regulatory protein (StAR). Clock cells contain an autonomous molecular oscillator based on a group of clock genes and their protein products. Mice lacking the VPAC2 receptor display disrupted circadian rhythm of physiology and behaviour, and therefore, we using real-time RT-PCR quantified (1) the mRNAs for the clock genes Per1 and Bmal1 in the adrenal gland and SCN, (2) the adrenal Star mRNA and (3) the serum corticosterone concentration both during a light/dark (L/D) cycle and at constant darkness in wild type (WT) and VPAC2 receptor-deficient mice (VPAC2-KO). We also examined if PER1 and StAR were co-localised in the adrenal steroidogenic cells. Per1 and Bmal1 mRNA showed a 24-h rhythmic expression in the adrenal of WT mice under L/D and dark conditions. During a L/D cycle, the adrenal clock gene rhythm in VPAC2-KO mice was phase-advanced by approximately 6 h compared to WT mice and became arrhythmic in constant darkness. A significant 24-h rhythmic variation in the adrenal Star mRNA expression and circulating corticosterone concentration was similarly phase-advanced during the L/D cycle. The loss of adrenal clock gene rhythm in the VPAC2 receptor knockout mice after transfer into constant darkness was accompanied by disappearance of rhythmicity in Star mRNA expression and serum corticosterone concentration. Double immunohistochemistry showed that the PER1 protein and StAR were co-localised in the same steroidogenic cells. Circulating corticosterone plays a role in the circadian timing system and the misaligned corticosterone rhythm in the VPAC2 receptor knockout mice could be involved in their abnormal rhythms of physiology.


Subject(s)
ARNTL Transcription Factors/genetics , Adrenal Cortex/metabolism , Circadian Rhythm/physiology , Corticosterone/blood , Gene Expression Regulation/physiology , Period Circadian Proteins/genetics , Phosphoproteins/genetics , Receptors, Vasoactive Intestinal Peptide, Type II/deficiency , Suprachiasmatic Nucleus/metabolism , ARNTL Transcription Factors/biosynthesis , Adrenal Medulla/metabolism , Animals , Circadian Rhythm/genetics , Circadian Rhythm/radiation effects , Corticosterone/metabolism , Darkness , Gene Expression Regulation/radiation effects , In Situ Hybridization , Light , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Period Circadian Proteins/biosynthesis , Phosphoproteins/biosynthesis , RNA, Messenger , Real-Time Polymerase Chain Reaction , Receptors, Vasoactive Intestinal Peptide, Type II/physiology
19.
PLoS One ; 7(4): e34462, 2012.
Article in English | MEDLINE | ID: mdl-22496809

ABSTRACT

Neuroglobin (Ngb), a neuron-specific oxygen-binding globin with an unknown function, has been proposed to play a key role in neuronal survival. We have previously shown Ngb to be highly expressed in the rat suprachiasmatic nucleus (SCN). The present study addresses the effect of Ngb deficiency on circadian behavior. Ngb-deficient and wild-type (wt) mice were placed in running wheels and their activity rhythms, endogenous period and response to light stimuli were investigated. The effect of Ngb deficiency on the expression of Period1 (Per1) and the immediate early gene Fos was determined after light stimulation at night and the neurochemical phenotype of Ngb expressing neurons in wt mice was characterized. Loss of Ngb function had no effect on overall circadian entrainment, but resulted in a significantly larger phase delay of circadian rhythm upon light stimulation at early night. A light-induced increase in Per1, but not Fos, gene expression was observed in Ngb-deficient mice. Ngb expressing neurons which co-stored Gastrin Releasing Peptide (GRP) and were innervated from the eye and the geniculo-hypothalamic tract expressed FOS after light stimulation. No PER1 expression was observed in Ngb-positive neurons. The present study demonstrates for the first time that the genetic elimination of Ngb does not affect core clock function but evokes an increased behavioural response to light concomitant with increased Per1 gene expression in the SCN at early night.


Subject(s)
Circadian Rhythm/physiology , Globins/physiology , Light , Nerve Tissue Proteins/physiology , Period Circadian Proteins/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Suprachiasmatic Nucleus/metabolism , Animals , Behavior, Animal , Blotting, Western , Female , Fluorescent Antibody Technique , Immunoenzyme Techniques , Immunoprecipitation , Male , Mice , Mice, Inbred C57BL , Neuroglobin , Neurons/cytology , Neurons/metabolism , Period Circadian Proteins/genetics , Proto-Oncogene Proteins c-fos/genetics , RNA, Messenger/genetics , Rats , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Suprachiasmatic Nucleus/cytology , Synapses/physiology
20.
J Neurochem ; 112(3): 797-806, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19943848

ABSTRACT

The photopigment melanopsin is expressed in a subtype of mammalian ganglion cells in the retina that project to the circadian clock in the hypothalamic suprachiasmatic nucleus to mediate non-visual light information. Melanopsin renders these retinal ganglion cells intrinsically photosensitive and the cells respond to light by a membrane depolarization and induction of the immediate early response gene Fos. Previous studies showed that the light activated melanopsin-induced signaling, the phototransduction, leading to depolarization of the membrane resembles the invertebrate opsins, which involves a Galpha(q/11) coupled phospholipase C activation. However, the signaling proteins mediating melanopsin-induced Fos expression are unresolved. In this study, we examined the phototransduction leading to Fos expression in melanopsin-transfected PC12 cells. A pivotal role of the extracellular signal-regulated protein kinase 1/2 (ERK1/2) was found as pharmacological blockage of this kinase suppressed the light-induced Fos expression. Illumination increased the inositol phosphate turnover and induced phosphorylation of ERK1/2 and p38 but not the c-Jun N-terminal kinase. The Galpha(q/11) protein inhibitor YM254890 attenuated these intracellular light responses. Our data strongly indicate that Galpha(q/11)-mediated ERK1/2 activation is essential for expression of Fos upon illumination of melanopsin-expressing PC12 cells.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/drug effects , Light , Oncogene Proteins v-fos/metabolism , Rod Opsins/metabolism , Animals , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/genetics , Gene Expression Regulation/physiology , Inositol Phosphates/metabolism , Oncogene Proteins v-fos/genetics , PC12 Cells/drug effects , PC12 Cells/physiology , PC12 Cells/radiation effects , Phosphorylation/drug effects , Phosphorylation/radiation effects , Rats , Rod Opsins/genetics , Statistics, Nonparametric , Transfection/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...