Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Res Sq ; 2023 Mar 17.
Article in English | MEDLINE | ID: mdl-36993397

ABSTRACT

There is growing interest in developing artificial lighting that stimulates intrinsically photosensitive retinal ganglion cells (ipRGCs) to entrain circadian rhythms to improve mood, sleep, and health. Efforts have focused on stimulating the intrinsic photopigment, melanopsin; however, recently, specialized color vision circuits have been elucidated in the primate retina that transmit blue-yellow cone-opponent signals to ipRGCs. We designed a light that stimulates color-opponent inputs to ipRGCs by temporally alternating short and longer wavelength components that strongly modulate short-wavelength sensitive (S) cones. Two-hour exposure to this S-cone modulating light produced an average circadian phase advance of one hour and twenty minutes in 6 subjects (mean age = 30 years) compared to no phase advance for the subjects after exposure to a 500-lux white light equated for melanopsin effectiveness. These results are promising for developing artificial lighting that is highly effective in controlling circadian rhythms by invisibly modulating cone-opponent circuits.

3.
Front Physiol ; 13: 934591, 2022.
Article in English | MEDLINE | ID: mdl-35957988

ABSTRACT

Mammals maintain their internal body temperature within a physiologically optimal range. This involves the regulation of core body temperature in response to changing environmental temperatures and a natural circadian oscillation of internal temperatures. The preoptic area (POA) of the hypothalamus coordinates body temperature by responding to both external temperature cues and internal brain temperature. Here we describe an autonomous circadian clock system in the murine ventromedial POA (VMPO) in close proximity to cells which express the atypical violet-light sensitive opsin, Opn5. We analyzed the light-sensitivity and thermal-sensitivity of the VMPO circadian clocks ex vivo. The phase of the VMPO circadian oscillations was not influenced by light. However, the VMPO clocks were reset by temperature changes within the physiological internal temperature range. This thermal-sensitivity of the VMPO circadian clock did not require functional Opn5 expression or a functional circadian clock within the Opn5-expressing cells. The presence of temperature-sensitive circadian clocks in the VMPO provides an advancement in the understanding of mechanisms involved in the dynamic regulation of core body temperature.

4.
Semin Cell Dev Biol ; 126: 87-96, 2022 06.
Article in English | MEDLINE | ID: mdl-33810978

ABSTRACT

Most organisms contain self-sustained circadian clocks. These clocks can be synchronized by environmental stimuli, but can also oscillate indefinitely in isolation. In mammals this is true at the molecular level for the majority of cell types that have been examined. A core set of "clock genes" form a transcriptional/translational feedback loop (TTFL) which repeats with a period of approximately 24 h. The exact mechanism of the TTFL differs slightly in various cell types, but all involve similar family members of the core cohort of clock genes. The clock has many outputs which are unique for different tissues. Cells in diverse tissues will convert the timing signals provided by the TTFL into uniquely orchestrated transcriptional oscillations of many clock-controlled genes and cellular processes.


Subject(s)
Circadian Clocks , Circadian Rhythm , Animals , Circadian Clocks/genetics , Circadian Rhythm/genetics , Humans , Mammals/genetics , Protein Processing, Post-Translational
6.
J Biol Rhythms ; 36(5): 470-482, 2021 10.
Article in English | MEDLINE | ID: mdl-34323103

ABSTRACT

The presence of an endogenous circadian clock within most mammalian cells is associated with the amazing observation that within a given tissue, these clocks are largely in synchrony with each other. Different tissues use a variety of systemic or environmental cues to precisely coordinate the phase of these clocks. The cornea is a unique tissue in that it is largely isolated from the direct blood supply that most tissues experience, it is transparent to visible light, and it is exposed directly to environmental light and temperature. Melatonin is a hormone that has been implicated in regulation of the cornea's circadian clocks. Here, we analyze the ability of rhythmic melatonin to entrain corneas ex vivo, and analyze the phase of corneal circadian clocks in vivo both in light: dark cycles and in constant darkness. We find that the presence of a retina from a melatonin-proficient mouse strain, C3Sn, can photoentrain the circadian clocks of a co-cultured mouse cornea, but a retina from a melatonin-deficient strain, C57Bl/6, cannot. Furthermore, pharmacologic blockade of melatonin or use of a retina with advanced retinal degeneration, Pde6brd1, blocks the photoentraining effect. Corneal circadian clocks in vivo adopt an advanced phase in C3Sn mice compared with C57Bl/6, but the circadian clocks in the liver are unaffected. This observation is not attributable to a shorter endogenous period of the cornea or behavior between the strains. Some transcripts of circadian genes in the corneas of C3Sn mice also show an advanced phase of expression in a light: dark cycle, while the transcript of Per2 exhibits a light-dependent transient induction at the onset of darkness. We conclude that melatonin acts as a phase modifying factor in a rhythmic manner for the circadian clocks of the cornea.


Subject(s)
Circadian Clocks , Melatonin , Animals , Circadian Clocks/genetics , Circadian Rhythm , Cornea , Light , Melatonin/pharmacology , Mice , Mice, Inbred C57BL
7.
J Biol Rhythms ; 36(2): 109-126, 2021 04.
Article in English | MEDLINE | ID: mdl-33765865

ABSTRACT

Animals have evolved light-sensitive G protein-coupled receptors, known as opsins, to detect coherent and ambient light for visual and nonvisual functions. These opsins have evolved to satisfy the particular lighting niches of the organisms that express them. While many unique patterns of evolution have been identified in mammals for rod and cone opsins, far less is known about the atypical mammalian opsins. Using genomic data from over 400 mammalian species from 22 orders, unique patterns of evolution for each mammalian opsins were identified, including photoisomerases, RGR-opsin (RGR) and peropsin (RRH), as well as atypical opsins, encephalopsin (OPN3), melanopsin (OPN4), and neuropsin (OPN5). The results demonstrate that OPN5 and rhodopsin show extreme conservation across all mammalian lineages. The cone opsins, SWS1 and LWS, and the nonvisual opsins, OPN3 and RRH, demonstrate a moderate degree of sequence conservation relative to other opsins, with some instances of lineage-specific gene loss. Finally, the photoisomerase, RGR, and the best-studied atypical opsin, OPN4, have high sequence diversity within mammals. These conservation patterns are maintained in human populations. Importantly, all mammalian opsins retain key amino acid residues important for conjugation to retinal-based chromophores, permitting light sensitivity. These patterns of evolution are discussed along with known functions of each atypical opsin, such as in circadian or metabolic physiology, to provide insight into the observed patterns of evolutionary constraint.


Subject(s)
Evolution, Molecular , Mammals/metabolism , Opsins/metabolism , Opsins/radiation effects , Animals , Circadian Rhythm/radiation effects , Conserved Sequence , Humans , Mice , Opsins/chemistry , Opsins/genetics , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/radiation effects , Retina/metabolism , Retina/radiation effects , Rhodopsin/chemistry , Rhodopsin/genetics , Rhodopsin/metabolism , Rhodopsin/radiation effects
8.
Am J Respir Cell Mol Biol ; 64(1): 59-68, 2021 01.
Article in English | MEDLINE | ID: mdl-33058732

ABSTRACT

Recently, we characterized blue light-mediated relaxation (photorelaxation) of airway smooth muscle (ASM) and implicated the involvement of opsin 3 (OPN3), an atypical opsin. In the present study, we characterized the cellular signaling mechanisms of photorelaxation. We confirmed the functional role of OPN3 in blue light photorelaxation using trachea from OPN3 null mice (maximal relaxation 52 ± 13% compared with wild-type mice 90 ± 4.3%, P < 0.05). We then demonstrated colocalization of OPN3 and Gαs using co-IP and proximity ligation assays in primary human ASM cells, which was further supported by an increase in cAMP in mouse trachea treated with blue light compared with dark controls (23 ± 3.6 vs. 14 ± 2.6 pmol cAMP/ring, P < 0.05). Downstream PKA (protein kinase A) involvement was shown by inhibiting photorelaxation using Rp-cAMPS (P < 0.0001). Moreover, we observed converging mechanisms of desensitization by chronic ß2-agonist exposure in mouse trachea and correlated this finding with colocalization of OPN3 and GRK2 (G protein receptor kinase) in primary human ASM cells. Finally, an overexpression model of OPN1LW (a red light photoreceptor in the same opsin family) in human ASM cells showed an increase in intracellular cAMP levels following red light exposure compared with nontransfected cells (48 ± 13 vs. 13 ± 2.1 pmol cAMP/mg protein, P < 0.01), suggesting a conserved photorelaxation mechanism for wavelengths of light that are more tissue penetrant. Together, these results demonstrate that blue light photorelaxation in ASM is mediated by the OPN3 receptor interacting with Gαs, which increases cAMP levels, activating PKA and modulated by GRK2.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/metabolism , Muscle Relaxation/physiology , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/metabolism , Rod Opsins/metabolism , Trachea/metabolism , Animals , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Opsins/metabolism , Signal Transduction/physiology
9.
Biotechniques ; 70(1): 49-53, 2021 01.
Article in English | MEDLINE | ID: mdl-33307798

ABSTRACT

In molecular biology laboratories, many tasks require fine motor control and high acuity vision. For example, lab technicians with visual impairment experience difficulty loading samples into the small wells of a horizontal agarose gel. We have developed a 3D-printable gel loading system which allows technicians with low-contrast vision to load gels correctly. It includes a casting tray, a bridge, and a modified comb. The system provides a high-contrast visual field to improve visibility, and the bridge allows pipette tips to be inserted at the correct location and only to the correct depth. The necessary computer files for printing this device are freely available to increase the accessibility of molecular biology laboratories to people with visual impairment.


Subject(s)
Electrophoresis/instrumentation , Printing, Three-Dimensional , Vision Disorders , Humans
10.
Nature ; 585(7825): 420-425, 2020 09.
Article in English | MEDLINE | ID: mdl-32879486

ABSTRACT

The opsin family of G-protein-coupled receptors are used as light detectors in animals. Opsin 5 (also known as neuropsin or OPN5) is a highly conserved opsin that is sensitive to visible violet light1,2. In mice, OPN5 is a known photoreceptor in the retina3 and skin4 but is also expressed in the hypothalamic preoptic area (POA)5. Here we describe a light-sensing pathway in which POA neurons that express Opn5 regulate thermogenesis in brown adipose tissue (BAT). We show that Opn5 is expressed in glutamatergic warm-sensing POA neurons that receive synaptic input from several thermoregulatory nuclei. We further show that Opn5 POA neurons project to BAT and decrease its activity under chemogenetic stimulation. Opn5-null mice show overactive BAT, increased body temperature, and exaggerated thermogenesis when cold-challenged. Moreover, violet photostimulation during cold exposure acutely suppresses BAT temperature in wild-type mice but not in Opn5-null mice. Direct measurements of intracellular cAMP ex vivo show that Opn5 POA neurons increase cAMP when stimulated with violet light. This analysis thus identifies a violet light-sensitive deep brain photoreceptor that normally suppresses BAT thermogenesis.


Subject(s)
Color , Light , Membrane Proteins/metabolism , Neurons/metabolism , Neurons/radiation effects , Opsins/metabolism , Preoptic Area/cytology , Thermogenesis/radiation effects , Adipose Tissue, Brown/innervation , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/radiation effects , Animals , Body Temperature , Cold Temperature , Cyclic AMP/metabolism , Female , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Opsins/deficiency , Opsins/genetics , Thermogenesis/genetics
11.
Invest Ophthalmol Vis Sci ; 61(6): 37, 2020 06 03.
Article in English | MEDLINE | ID: mdl-32543667

ABSTRACT

Purpose: Autonomous molecular circadian clocks are present in the majority of mammalian tissues. These clocks are synchronized to phases appropriate for their physiologic role by internal systemic cues, external environmental cues, or both. The circadian clocks of the in vivo mouse cornea synchronize to the phase of the brain's master clock primarily through systemic cues, but ex vivo corneal clocks entrain to environmental light cycles. We evaluated the underlying mechanisms of this difference. Methods: Molecular circadian clocks of mouse corneas were evaluated in vivo and ex vivo for response to environmental light. The presence of opsins and effect of genetic deletion of opsins were evaluated for influence on circadian photoresponses. Opn5-expressing cells were identified using Opn5Cre;Ai14 mice and RT-PCR, and they were characterized using immunocytochemistry. Results: Molecular circadian clocks of the cornea remain in phase with behavioral circadian locomotor rhythms in vivo but are photoentrainable in tissue culture. After full-thickness incision or epithelial debridement, expression of the opsin photopigment Opn5 is induced in the cornea in a subset of preexisting epithelial cells adjacent to the wound site. This induction coincides with conferral of direct, short-wavelength light sensitivity to the circadian clocks throughout the cornea. Conclusions: Corneal circadian rhythms become photosensitive after wounding. Opn5 gene function (but not Opn3 or Opn4 function) is necessary for induced photosensitivity. These results demonstrate that opsin-dependent direct light sensitivity can be facultatively induced in the murine cornea.


Subject(s)
Circadian Rhythm/physiology , Cornea/metabolism , Corneal Injuries/genetics , Gene Expression Regulation , Membrane Proteins/genetics , Opsins/genetics , RNA/genetics , Rod Opsins/metabolism , Animals , Cornea/pathology , Corneal Injuries/metabolism , Corneal Injuries/physiopathology , Disease Models, Animal , Membrane Proteins/biosynthesis , Mice , Mice, Inbred C57BL , Opsins/biosynthesis , Photoperiod
12.
Cell Rep ; 30(3): 672-686.e8, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31968245

ABSTRACT

Almost all life forms can detect and decode light information for adaptive advantage. Examples include the visual system, in which photoreceptor signals are processed into virtual images, and the circadian system, in which light entrains a physiological clock. Here we describe a light response pathway in mice that employs encephalopsin (OPN3, a 480 nm, blue-light-responsive opsin) to regulate the function of adipocytes. Germline null and adipocyte-specific conditional null mice show a light- and Opn3-dependent deficit in thermogenesis and become hypothermic upon cold exposure. We show that stimulating mouse adipocytes with blue light enhances the lipolysis response and, in particular, phosphorylation of hormone-sensitive lipase. This response is Opn3 dependent. These data establish a key mechanism in which light-dependent, local regulation of the lipolysis response in white adipocytes regulates energy metabolism.


Subject(s)
Adipocytes, Brown/metabolism , Adipocytes, Brown/radiation effects , Adipocytes, White/metabolism , Adipocytes, White/radiation effects , Light , Rod Opsins/metabolism , Thermogenesis/radiation effects , Animals , Cold Temperature , Energy Metabolism/radiation effects , Gene Expression Profiling , Lipolysis/radiation effects , Mice, Inbred C57BL , Phenotype , Photons , Thermogenesis/genetics
13.
Curr Biol ; 29(20): 3478-3487.e4, 2019 10 21.
Article in English | MEDLINE | ID: mdl-31607531

ABSTRACT

Nearly all mammalian tissues have functional, autonomous circadian clocks, which free-run with non-24 h periods and must be synchronized (entrained) to the 24 h day. This entrainment mechanism is thought to be hierarchical, with photic input to the retina entraining the master circadian clock in the suprachiasmatic nuclei (SCN) and the SCN in turn synchronizing peripheral tissues via endocrine mechanisms. Here, we assess the function of a population of melanocyte precursor cells in hair and vibrissal follicles that express the photopigment neuropsin (OPN5). Organotypic cultures of murine outer ear and vibrissal skin entrain to a light-dark cycle ex vivo, requiring cis-retinal chromophore and Opn5 gene function. Short-wavelength light strongly phase shifts skin circadian rhythms ex vivo via an Opn5-dependent mechanism. In vivo, the normal amplitude of Period mRNA expression in outer ear skin is dependent on both the light-dark cycle and Opn5 function. In Opn4-/-; Pde6brd1/rd1 mice that cannot behaviorally entrain to light-dark cycles, the phase of skin-clock gene expression remains synchronized to the light-dark cycle, even as other peripheral clocks remain phase-locked to the free-running behavioral rhythm. Taken together, these results demonstrate the presence of a direct photic circadian entrainment pathway and direct light-response elements for clock genes in murine skin, similar to pathways previously described for invertebrates and certain non-mammalian vertebrates.


Subject(s)
Circadian Clocks/genetics , Circadian Rhythm/physiology , Light , Membrane Proteins/genetics , Opsins/genetics , Photoperiod , Animals , Circadian Clocks/radiation effects , Circadian Rhythm/radiation effects , Female , Male , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Opsins/metabolism , Random Allocation , Skin
14.
Nat Cell Biol ; 21(4): 420-429, 2019 04.
Article in English | MEDLINE | ID: mdl-30936473

ABSTRACT

During mouse postnatal eye development, the embryonic hyaloid vascular network regresses from the vitreous as an adaption for high-acuity vision. This process occurs with precisely controlled timing. Here, we show that opsin 5 (OPN5; also known as neuropsin)-dependent retinal light responses regulate vascular development in the postnatal eye. In Opn5-null mice, hyaloid vessels regress precociously. We demonstrate that 380-nm light stimulation via OPN5 and VGAT (the vesicular GABA/glycine transporter) in retinal ganglion cells enhances the activity of inner retinal DAT (also known as SLC6A3; a dopamine reuptake transporter) and thus suppresses vitreal dopamine. In turn, dopamine acts directly on hyaloid vascular endothelial cells to suppress the activity of vascular endothelial growth factor receptor 2 (VEGFR2) and promote hyaloid vessel regression. With OPN5 loss of function, the vitreous dopamine level is elevated and results in premature hyaloid regression. These investigations identify violet light as a developmental timing cue that, via an OPN5-dopamine pathway, regulates optic axis clearance in preparation for visual function.


Subject(s)
Dopamine/metabolism , Eye/blood supply , Light , Membrane Proteins/metabolism , Opsins/metabolism , Animals , Dopamine Plasma Membrane Transport Proteins/antagonists & inhibitors , Dopamine Plasma Membrane Transport Proteins/chemistry , Dopamine Plasma Membrane Transport Proteins/metabolism , Endothelium, Vascular/metabolism , Eye/enzymology , Eye/growth & development , Eye/metabolism , Membrane Proteins/genetics , Mice , Mice, Knockout , Opsins/genetics , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/radiation effects , Threonine/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/physiology , Vitreous Body/metabolism
15.
Invest Ophthalmol Vis Sci ; 59(12): 4856-4870, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30347082

ABSTRACT

Vision is a highly rhythmic function adapted to the extensive changes in light intensity occurring over the 24-hour day. This adaptation relies on rhythms in cellular and molecular processes, which are orchestrated by a network of circadian clocks located within the retina and in the eye, synchronized to the day/night cycle and which, together, fine-tune detection and processing of light information over the 24-hour period and ensure retinal homeostasis. Systematic or high throughput studies revealed a series of genes rhythmically expressed in the retina, pointing at specific functions or pathways under circadian control. Conversely, knockout studies demonstrated that the circadian clock regulates retinal processing of light information. In addition, recent data revealed that it also plays a role in development as well as in aging of the retina. Regarding synchronization by the light/dark cycle, the retina displays the unique property of bringing together light sensitivity, clock machinery, and a wide range of rhythmic outputs. Melatonin and dopamine play a particular role in this system, being both outputs and inputs for clocks. The retinal cellular complexity suggests that mechanisms of regulation by light are diverse and intricate. In the context of the whole eye, the retina looks like a major determinant of phase resetting for other tissues such as the retinal pigmented epithelium or cornea. Understanding the pathways linking the cell-specific molecular machineries to their cognate outputs will be one of the major challenges for the future.


Subject(s)
Adaptation, Ocular/physiology , Circadian Clocks/physiology , Circadian Rhythm/physiology , Retina/physiology , Animals , CLOCK Proteins/genetics , Dopamine/metabolism , Gene Expression , Humans , Melatonin/metabolism , Ocular Physiological Phenomena
16.
Vis Neurosci ; 35: E004, 2018 01.
Article in English | MEDLINE | ID: mdl-29905117

ABSTRACT

A unique class of intrinsically photosensitive retinal ganglion cells in mammalian retinae has been recently discovered and characterized. These neurons can generate visual signals in the absence of inputs from rods and cones, the conventional photoreceptors in the visual system. These light sensitive ganglion cells (mRGCs) express the non-rod, non-cone photopigment melanopsin and play well documented roles in modulating pupil responses to light, photoentrainment of circadian rhythms, mood, sleep and other adaptive light functions. While most research efforts in mammals have focused on mRGCs in retina, recent studies reveal that melanopsin is expressed in non-retinal tissues. For example, light-evoked melanopsin activation in extra retinal tissue regulates pupil constriction in the iris and vasodilation in the vasculature of the heart and tail. As another example of nonretinal melanopsin expression we report here the previously unrecognized localization of this photopigment in nerve fibers within the cornea. Surprisingly, we were unable to detect light responses in the melanopsin-expressing corneal fibers in spite of our histological evidence based on genetically driven markers and antibody staining. We tested further for melanopsin localization in cell bodies of the trigeminal ganglia (TG), the principal nuclei of the peripheral nervous system that project sensory fibers to the cornea, and found expression of melanopsin mRNA in a subset of TG neurons. However, neither electrophysiological recordings nor calcium imaging revealed any light responsiveness in the melanopsin positive TG neurons. Given that we found no light-evoked activation of melanopsin-expressing fibers in cornea or in cell bodies in the TG, we propose that melanopsin protein might serve other sensory functions in the cornea. One justification for this idea is that melanopsin expressed in Drosophila photoreceptors can serve as a temperature sensor.


Subject(s)
Cornea/metabolism , Gene Expression Regulation/physiology , Rod Opsins/genetics , Trigeminal Ganglion/metabolism , Animals , Cell Body/metabolism , Cells, Cultured , Dependovirus/genetics , Electrophysiology , Female , Guinea Pigs , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nerve Fibers/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Rod Opsins/metabolism , Transfection
17.
Proc Natl Acad Sci U S A ; 114(42): E8855-E8864, 2017 10 17.
Article in English | MEDLINE | ID: mdl-28973913

ABSTRACT

We previously created two PER2::LUCIFERASE (PER2::LUC) circadian reporter knockin mice that differ only in the Per2 3'-UTR region: Per2::Luc, which retains the endogenous Per2 3'-UTR and Per2::LucSV, where the endogenous Per2 3'-UTR was replaced by an SV40 late poly(A) signal. To delineate the in vivo functions of Per2 3'-UTR, we analyzed circadian rhythms of Per2::LucSV mice. Interestingly, Per2::LucSV mice displayed more than threefold stronger amplitude in bioluminescence rhythms than Per2::Luc mice, and also exhibited lengthened free-running periods (∼24.0 h), greater phase delays following light pulse, and enhanced temperature compensation relative to Per2::Luc Analysis of the Per2 3'-UTR sequence revealed that miR-24, and to a lesser degree miR-30, suppressed PER2 protein translation, and the reversal of this inhibition in Per2::LucSV augmented PER2::LUC protein level and oscillatory amplitude. Interestingly, Bmal1 mRNA and protein oscillatory amplitude as well as CRY1 protein oscillation were increased in Per2::LucSV mice, suggesting rhythmic overexpression of PER2 enhances expression of Per2 and other core clock genes. Together, these studies provide important mechanistic insights into the regulatory roles of Per2 3'-UTR, miR-24, and PER2 in Per2 expression and core clock function.


Subject(s)
Circadian Rhythm/physiology , MicroRNAs/genetics , Period Circadian Proteins/genetics , 3' Untranslated Regions , Animals , Circadian Clocks/genetics , Gene Expression Regulation , Gene Knock-In Techniques , Luciferases/genetics , Mice, Inbred C57BL , Mice, Transgenic , Period Circadian Proteins/metabolism , Protein Biosynthesis , Temperature
18.
PLoS One ; 12(9): e0184790, 2017.
Article in English | MEDLINE | ID: mdl-28934261

ABSTRACT

PURPOSE: Intraocular pressure (IOP) is known to have a strong circadian rhythm, yet how light/dark cycles entrain this rhythm is unknown. The purpose of this study was to assess whether, like the retina, the mammalian ciliary body and IOP clocks have an intrinsic ability to entrain to light/dark cycles. METHODS: Iris-ciliary body complexes were obtained from period2:luciferase (PER2::LUC) mice and cultured to measure bioluminescence rhythmicity. Pairs of the iris-ciliary body complex were exposed to antiphasic 9:15 h light/dark cycle in vitro. After 4 days of exposure to light/dark cycles, bioluminescence was recorded to establish their circadian phases. In addition, pairs of the iris-ciliary body complex co-cultured with the retinas or corneas of wild-type mice were also investigated. The IOP circadian changes of free-running Opn4-/-;rd1/rd1 mice whose behavior was antiphasic to wild-type were measured by a rebound tonometry, and compared with wild-type mice. Opn3, Opn4, and Opn5 mRNA expression in the iris-ciliary body were analyzed using RT-PCR. RESULTS: The iris/ciliary body complex expressed Opn3, Opn4, and Opn5 mRNA; however, unlike in retina and cornea, neither the iris-CB complex nor the co-cultured complex was directly entrained by light-dark cycle in vitro. The diurnal IOP change of Opn4-/-;rd1/rd1 mice showed an antiphasic pattern to wild-type mice and their rhythms followed the whole-animal behavioral rhythm. CONCLUSIONS: Despite expressing mRNA for several non-visual opsins, circadian rhythms of the iris-ciliary body complex of mice do not entrain directly to light-dark cycles ex vivo. Unlike retina, the iris/ciliary body clocks of blind mice remain synchronized to the organismal behavioral rhythm rather than local light-dark cycles. These results suggest that IOP rhythm entrainment is mediated by a systemic rather than local signal in mice.


Subject(s)
Circadian Rhythm/physiology , Intraocular Pressure/physiology , Photoperiod , Animals , Ciliary Body/physiology , Coculture Techniques , Cornea/physiology , Female , Gene Expression Regulation/physiology , Male , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Opsins/metabolism , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , Photic Stimulation , RNA, Messenger/metabolism , Retina/physiology , Rod Opsins/genetics , Rod Opsins/metabolism
19.
Curr Biol ; 27(1): 128-136, 2017 Jan 09.
Article in English | MEDLINE | ID: mdl-28017605

ABSTRACT

The suprachiasmatic nucleus (SCN) is the central circadian clock in mammals. It is entrained by light but resistant to temperature shifts that entrain peripheral clocks [1-5]. The SCN expresses many functionally important neuropeptides, including vasoactive intestinal peptide (VIP), which drives light entrainment, synchrony, and amplitude of SCN cellular clocks and organizes circadian behavior [5-16]. The transcription factor LHX1 drives SCN Vip expression, and cellular desynchrony in Lhx1-deficient SCN largely results from Vip loss [17, 18]. LHX1 regulates many genes other than Vip, yet activity rhythms in Lhx1-deficient mice are similar to Vip-/- mice under light-dark cycles and only somewhat worse in constant conditions. We suspected that LHX1 targets other than Vip have circadian functions overlooked in previous studies. In this study, we compared circadian sleep and temperature rhythms of Lhx1- and Vip-deficient mice and found loss of acute light control of sleep in Lhx1 but not Vip mutants. We also found loss of circadian resistance to fever in Lhx1 but not Vip mice, which was partially recapitulated by heat application to cultured Lhx1-deficient SCN. Having identified VIP-independent functions of LHX1, we mapped the VIP-independent transcriptional network downstream of LHX1 and a largely separable VIP-dependent transcriptional network. The VIP-independent network does not affect core clock amplitude and synchrony, unlike the VIP-dependent network. These studies identify Lhx1 as the first gene required for temperature resistance of the SCN clockworks and demonstrate that acute light control of sleep is routed through the SCN and its immediate output regions.


Subject(s)
Circadian Clocks , Gene Regulatory Networks , LIM-Homeodomain Proteins/physiology , Sleep , Transcription Factors/physiology , Vasoactive Intestinal Peptide/physiology , Wakefulness , Animals , Circadian Rhythm , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Hot Temperature , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Photoperiod , Signal Transduction , Suprachiasmatic Nucleus/cytology , Suprachiasmatic Nucleus/metabolism
20.
Neuron ; 90(5): 909-11, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27253443

ABSTRACT

In this issue of Neuron, Mure et al. (2016) demonstrate that two mechanisms-phosphorylation of a C-terminal intracellular region, and mechanism involving the whole of the C terminus-oppositely shape the kinetics and sensitivity of the nonvisual photoreceptor melanopsin.


Subject(s)
Retinal Ganglion Cells , Rod Opsins , Humans , Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL
...