1
|
Constable PA, Skuse DH, Thompson DA, Lee IO. Brief report: effects of methylphenidate on the light adapted electroretinogram. Doc Ophthalmol 2024:10.1007/s10633-024-10000-3. [PMID: 39674982 DOI: 10.1007/s10633-024-10000-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 12/06/2024] [Indexed: 12/17/2024]
Abstract
PURPOSE To explore changes in the electroretinogram (ERG) following methylphenidate use in attention-deficit/hyperactivity disorder (ADHD). METHODS Light adapted ERGs were recorded in five individuals (3 male and 2 female, age range 13.6-21.8 years) with a diagnosis of ADHD. Six flash strengths ranging from 71 to 446 Td.s were qualitatively evaluated following a minimum of 24 h without any medication and from 2 to 6 h following the individuals' standard slow-release (XL) methylphenidate dose that ranged from 18 to 60 mg. RESULTS Of the six flash strengths, the 178 Td.s strength revealed changes in four of the five participants with a median 27.4% increase in b-wave amplitude. For three individuals there was an increase in the a-wave amplitude and for two of the same individuals there was also a noticeable pronouncement of the oscillatory potentials. The a-wave amplitude showed a greatest median increase at the 446 Td.s flash strength of 25.8%. One individual - on the highest dose (60 mg) exhibited no morphologically distinct changes in the ERG. No differences in the time to peaks of the a- and b-wave were observed for any individual. CONCLUSION The a- and b-wave amplitudes of the light adapted ERG could provide insights into the effect of methylphenidate in ADHD.
Collapse
Affiliation(s)
- Paul A Constable
- College of Nursing and Health Sciences, Caring Futures Institute, Flinders University, Adelaide, Australia.
| | - David H Skuse
- Behavioural and Brain Sciences Unit, Population Policy and Practice Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Dorothy A Thompson
- The Tony Kriss Visual Electrophysiology Unit, Clinical and Academic Department of Ophthalmology, Great Ormond Street Hospital for Children NHS Trust, London, UK
- UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Irene O Lee
- Behavioural and Brain Sciences Unit, Population Policy and Practice Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| |
Collapse
|
2
|
Xie J, Goodbourn PT, Bui BV, Jusuf PR. Establishment and comprehensive characterization of a novel dark-reared zebrafish model for myopia studies. Exp Eye Res 2024; 246:110009. [PMID: 39067805 DOI: 10.1016/j.exer.2024.110009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 07/09/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Myopia is predicted to impact approximately 5 billion people by 2050, necessitating mechanistic understanding of its development. Myopia results from dysregulated genetic mechanisms of emmetropization, caused by over-exposure to aberrant visual environments; however, these genetic mechanisms remain unclear. Recent human genome-wide association studies have identified a range of novel myopia-risk genes. To facilitate large-scale in vivo mechanistic examination of gene-environment interactions, this study aims to establish a myopia model platform that allows efficient environmental and genetic manipulations. We established an environmental zebrafish myopia model by dark-rearing. Ocular biometrics including relative ocular refraction were quantified using optical coherence tomography images. Spatial vision was assessed using optomotor response (OMR). Retinal function was analyzed via electroretinography (ERG). Myopia-associated molecular contents or distributions were examined using RT-qPCR or immunohistochemistry. Our model produces robust phenotypic changes, showing myopia after 2 weeks of dark-rearing, which were recoverable within 2 weeks after returning animals to normal lighting. 2-week dark-reared zebrafish have reduced spatial-frequency tuning function. ERG showed reduced photoreceptor and bipolar cell function (a- and b-waves) after only 2 days of dark-rearing, which worsened after 2 weeks of dark-rearing. We also found dark-rearing-induced changes to expression of myopia-risk genes, including egr1, vegfaa, vegfab, rbp3, gjd2a and gjd2b, inner retinal distribution of EFEMP1, TIMP2 and MMP2, as well as transiently reduced PSD95 density in the inner plexiform layer. Coupled with the gene editing tools available for zebrafish, our environmental myopia model provides an excellent platform for large-scale investigation of gene-environment interactions in myopia development.
Collapse
Affiliation(s)
- Jiaheng Xie
- School of Biosciences, The University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Patrick T Goodbourn
- Melbourne School of Psychological Sciences, The University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Bang V Bui
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, 3010, Victoria, Australia.
| | - Patricia Regina Jusuf
- School of Biosciences, The University of Melbourne, Parkville, 3010, Victoria, Australia.
| |
Collapse
|
3
|
Bergum N, Berezin CT, Vigh J. Dopamine enhances GABA A receptor-mediated current amplitude in a subset of intrinsically photosensitive retinal ganglion cells. J Neurophysiol 2024; 132:501-513. [PMID: 38958282 PMCID: PMC11427049 DOI: 10.1152/jn.00457.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 06/05/2024] [Accepted: 06/26/2024] [Indexed: 07/04/2024] Open
Abstract
Neuromodulation in the retina is crucial for effective processing of retinal signal at different levels of illuminance. Intrinsically photosensitive retinal ganglion cells (ipRGCs), the neurons that drive nonimage-forming visual functions, express a variety of neuromodulatory receptors that tune intrinsic excitability as well as synaptic inputs. Past research has examined actions of neuromodulators on light responsiveness of ipRGCs, but less is known about how neuromodulation affects synaptic currents in ipRGCs. To better understand how neuromodulators affect synaptic processing in ipRGC, we examine actions of opioid and dopamine agonists have on inhibitory synaptic currents in ipRGCs. Although µ-opioid receptor (MOR) activation had no effect on γ-aminobutyric acid (GABA) currents, dopamine [via the D1-type dopamine receptor (D1R)]) amplified GABAergic currents in a subset of ipRGCs. Furthermore, this D1R-mediated facilitation of the GABA conductance in ipRGCs was mediated by a cAMP/PKA-dependent mechanism. Taken together, these findings reinforce the idea that dopamine's modulatory role in retinal adaptation affects both nonimage-forming and image-forming visual functions.NEW & NOTEWORTHY Neuromodulators such as dopamine are important regulators of retinal function. Here, we demonstrate that dopamine increases inhibitory inputs to intrinsically photosensitive retinal ganglion cells (ipRGCs), in addition to its previously established effect on intrinsic light responsiveness. This indicates that dopamine, in addition to its ability to intrinsically modulate ipRGC activity, can also affect synaptic inputs to ipRGCs, thereby tuning retina circuits involved in nonimage-forming visual functions.
Collapse
Affiliation(s)
- Nikolas Bergum
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States
| | - Casey-Tyler Berezin
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins,Colorado, United States
| | - Jozsef Vigh
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins,Colorado, United States
| |
Collapse
|
4
|
Lau MYH, Gadiwalla S, Jones S, Galliano E. Different electrophysiological profiles of genetically labelled dopaminergic neurons in the mouse midbrain and olfactory bulb. Eur J Neurosci 2024; 59:1480-1499. [PMID: 38169095 DOI: 10.1111/ejn.16239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/06/2023] [Accepted: 12/09/2023] [Indexed: 01/05/2024]
Abstract
Dopaminergic (DA) neurons play pivotal roles in diverse brain functions, spanning movement, reward processing and sensory perception. DA neurons are most abundant in the midbrain (Substantia Nigra pars compacta [SNC] and Ventral Tegmental Area [VTA]) and the olfactory bulb (OB) in the forebrain. Interestingly, a subtype of OB DA neurons is capable of regenerating throughout life, while a second class is exclusively born during embryonic development. Compelling evidence in SNC and VTA also indicates substantial heterogeneity in terms of morphology, connectivity and function. To further investigate this heterogeneity and directly compare form and function of midbrain and forebrain bulbar DA neurons, we performed immunohistochemistry and whole-cell patch-clamp recordings in ex vivo brain slices from juvenile DAT-tdTomato mice. After confirming the penetrance and specificity of the dopamine transporter (DAT) Cre line, we compared soma shape, passive membrane properties, voltage sags and action potential (AP) firing across midbrain and forebrain bulbar DA subtypes. We found that each DA subgroup within midbrain and OB was highly heterogeneous, and that DA neurons across the two brain areas are also substantially different. These findings complement previous work in rats as well as gene expression and in vivo datasets, further questioning the existence of a single "dopaminergic" neuronal phenotype.
Collapse
Affiliation(s)
- Maggy Yu Hei Lau
- Department of Physiology, Development and Neuroscience, University of Cambridge, UK
| | - Sana Gadiwalla
- Department of Physiology, Development and Neuroscience, University of Cambridge, UK
| | - Susan Jones
- Department of Physiology, Development and Neuroscience, University of Cambridge, UK
| | - Elisa Galliano
- Department of Physiology, Development and Neuroscience, University of Cambridge, UK
| |
Collapse
|
5
|
Elmers J, Colzato LS, Ziemssen F, Ziemssen T, Beste C. Optical coherence tomography as a potential surrogate marker of dopaminergic modulation across the life span. Ageing Res Rev 2024; 96:102280. [PMID: 38518921 DOI: 10.1016/j.arr.2024.102280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 02/02/2024] [Accepted: 03/18/2024] [Indexed: 03/24/2024]
Abstract
The retina has been considered a "window to the brain" and shares similar innervation by the dopaminergic system with the cortex in terms of an unequal distribution of D1 and D2 receptors. Here, we provide a comprehensive overview that Optical Coherence Tomography (OCT), a non-invasive imaging technique, which provides an "in vivo" representation of the retina, shows promise to be used as a surrogate marker of dopaminergic neuromodulation in cognition. Overall, most evidence supports reduced retinal thickness in individuals with dopaminergic dysregulation (e.g., patients with Parkinson's Disease, non-demented older adults) and with poor cognitive functioning. By using the theoretical framework of metacontrol, we derive hypotheses that retinal thinning associated to decreased dopamine (DA) levels affecting D1 families, might lead to a decrease in the signal-to-noise ratio (SNR) affecting cognitive persistence (depending on D1-modulated DA activity) but not cognitive flexibility (depending on D2-modulated DA activity). We argue that the use of OCT parameters might not only be an insightful for cognitive neuroscience research, but also a potentially effective tool for individualized medicine with a focus on cognition. As our society progressively ages in the forthcoming years and decades, the preservation of cognitive abilities and promoting healthy aging will hold of crucial significance. OCT has the potential to function as a swift, non-invasive, and economical method for promptly recognizing individuals with a heightened vulnerability to cognitive deterioration throughout all stages of life.
Collapse
Affiliation(s)
- Julia Elmers
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Germany; Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus, TU Dresden, Germany
| | - Lorenza S Colzato
- Cognitive Psychology, Faculty of Psychology, Shandong Normal University, Jinan, China
| | - Focke Ziemssen
- Ophthalmological Clinic, University Clinic Leipzig, Germany
| | - Tjalf Ziemssen
- Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus, TU Dresden, Germany
| | - Christian Beste
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Germany; Cognitive Psychology, Faculty of Psychology, Shandong Normal University, Jinan, China.
| |
Collapse
|
6
|
Ruan Y, Buonfiglio F, Gericke A. Adrenoceptors in the Eye - Physiological and Pathophysiological Relevance. Handb Exp Pharmacol 2024; 285:453-505. [PMID: 38082203 DOI: 10.1007/164_2023_702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
The autonomic nervous system plays a crucial role in the innervation of the eye. Consequently, it comes as no surprise that catecholamines and their corresponding receptors have been extensively studied and characterized in numerous ocular structures, including the cornea, conjunctiva, lacrimal gland, trabecular meshwork, uvea, and retina. These investigations have unveiled substantial clinical implications, particularly in the context of treating glaucoma, a progressive neurodegenerative disorder responsible for irreversible vision loss on a global scale. The primary therapeutic approaches for glaucoma frequently involve the modulation of α1-, α2-, and β-adrenoceptors, making them pivotal targets. In this chapter, we offer a comprehensive overview of the expression, distribution, and functional roles of adrenoceptors within various components of the eye and its associated structures. Additionally, we delve into the pivotal role of adrenoceptors in the pathophysiology of glaucoma. Furthermore, we provide a concise historical perspective on adrenoceptor research, examine the distinct contributions of individual adrenoceptor subtypes to the treatment of various ocular conditions, and propose potential future avenues of exploration in this field.
Collapse
Affiliation(s)
- Yue Ruan
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Francesco Buonfiglio
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Adrian Gericke
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
7
|
Bergum N, Berezin CT, Vigh J. Dopamine enhances GABA A receptor-mediated current amplitude in a subset of intrinsically photosensitive retinal ganglion cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.11.571141. [PMID: 38168350 PMCID: PMC10760026 DOI: 10.1101/2023.12.11.571141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Neuromodulation in the retina is crucial for effective processing of retinal signal at different levels of illuminance. Intrinsically photosensitive retinal ganglion cells (ipRGCs), the neurons that drive non-image forming visual functions, express a variety of neuromodulatory receptors that tune intrinsic excitability as well as synaptic inputs. Past research has examined actions of neuromodulators on light responsiveness of ipRGCs, but less is known about how neuromodulation affects synaptic currents in ipRGCs. To better understand how neuromodulators affect synaptic processing in ipRGC, we examine actions of opioid and dopamine agonists have on inhibitory synaptic currents in ipRGCs. Although μ-opioid receptor (MOR) activation had no effect on γ-aminobutyric acid (GABA) currents, dopamine (via the D1R) amplified GABAergic currents in a subset of ipRGCs. Furthermore, this D1R-mediated facilitation of the GABA conductance in ipRGCs was mediated by a cAMP/PKA-dependent mechanism. Taken together, these findings reinforce the idea that dopamine's modulatory role in retinal adaptation affects both non-image forming as well as image forming visual functions.
Collapse
Affiliation(s)
- Nikolas Bergum
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Casey-Tyler Berezin
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO, USA
| | - Jozsef Vigh
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
8
|
Guidolin D, Tortorella C, Marcoli M, Cervetto C, De Caro R, Maura G, Agnati LF. Modulation of Neuron and Astrocyte Dopamine Receptors via Receptor-Receptor Interactions. Pharmaceuticals (Basel) 2023; 16:1427. [PMID: 37895898 PMCID: PMC10610355 DOI: 10.3390/ph16101427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 09/29/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
Dopamine neurotransmission plays critical roles in regulating complex cognitive and behavioral processes including reward, motivation, reinforcement learning, and movement. Dopamine receptors are classified into five subtypes, widely distributed across the brain, including regions responsible for motor functions and specific areas related to cognitive and emotional functions. Dopamine also acts on astrocytes, which express dopamine receptors as well. The discovery of direct receptor-receptor interactions, leading to the formation of multimeric receptor complexes at the cell membrane and providing the cell decoding apparatus with flexible dynamics in terms of recognition and signal transduction, has expanded the knowledge of the G-protein-coupled receptor-mediated signaling processes. The purpose of this review article is to provide an overview of currently identified receptor complexes containing dopamine receptors and of their modulatory action on dopamine-mediated signaling between neurons and between neurons and astrocytes. Pharmacological possibilities offered by targeting receptor complexes in terms of addressing neuropsychiatric disorders associated with altered dopamine signaling will also be briefly discussed.
Collapse
Affiliation(s)
- Diego Guidolin
- Department of Neuroscience, University of Padova, 35122 Padova, Italy; (C.T.); (R.D.C.)
| | - Cinzia Tortorella
- Department of Neuroscience, University of Padova, 35122 Padova, Italy; (C.T.); (R.D.C.)
| | - Manuela Marcoli
- Department of Pharmacy, University of Genova, 16126 Genova, Italy; (M.M.); (C.C.); (G.M.)
| | - Chiara Cervetto
- Department of Pharmacy, University of Genova, 16126 Genova, Italy; (M.M.); (C.C.); (G.M.)
| | - Raffaele De Caro
- Department of Neuroscience, University of Padova, 35122 Padova, Italy; (C.T.); (R.D.C.)
| | - Guido Maura
- Department of Pharmacy, University of Genova, 16126 Genova, Italy; (M.M.); (C.C.); (G.M.)
| | - Luigi F. Agnati
- Department of Biomedical, Metabolic Sciences and Neuroscience, University of Modena and Reggio Emilia, 41121 Modena, Italy;
| |
Collapse
|
9
|
Liu D, Fujihara K, Yanagawa Y, Mushiake H, Ohshiro T. Gad1 knock-out rats exhibit abundant spike-wave discharges in EEG, exacerbated with valproate treatment. Front Neurol 2023; 14:1243301. [PMID: 37830095 PMCID: PMC10566305 DOI: 10.3389/fneur.2023.1243301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/29/2023] [Indexed: 10/14/2023] Open
Abstract
Objective To elucidate the functional role of gamma-aminobutyric acid (GABA)-ergic inhibition in suppressing epileptic brain activities such as spike-wave discharge (SWD), we recorded electroencephalogram (EEG) in knockout rats for Glutamate decarboxylase 1 (Gad1), which encodes one of the two GABA-synthesizing enzymes in mammals. We also examined how anti-epileptic drug valproate (VPA) acts on the SWDs present in Gad1 rats and affects GABA synthesis in the reticular thalamic nucleus (RTN), which is known to play an essential role in suppressing SWD. Methods Chronic EEG recordings were performed in freely moving control rats and homozygous knockout Gad1 (-/-) rats. Buzzer tones (82 dB) were delivered to the rats during EEG monitoring to test whether acoustic stimulation could interrupt ongoing SWDs. VPA was administered orally to the rats, and the change in the number of SWDs was examined. The distribution of GABA in the RTN was examined immunohistochemically. Results SWDs were abundant in EEG from Gad1 (-/-) rats as young as 2 months old. Although SWDs were universally detected in older rats irrespective of their Gad1 genotype, SWD symptom was most severe in Gad1 (-/-) rats. Acoustic stimulation readily interrupted ongoing SWDs irrespective of the Gad1 genotype, whereas SWDs were more resistant to interruption in Gad1 (-/-) rats. VPA treatment alleviated SWD symptoms in control rats, however, counterintuitively exacerbated the symptoms in Gad1 (-/-) rats. The immunohistochemistry results indicated that GABA immunoreactivity was significantly reduced in the somata of RTN neurons in Gad1 (-/-) rats but not in their axons targeting the thalamus. VPA treatment greatly increased GABA immunoreactivity in the RTN neurons of Gad1 (-/-) rats, which is likely due to the intact GAD2, another GAD isozyme, in these neurons. Discussion Our results revealed two opposing roles of GABA in SWD generation: suppression and enhancement of SWD. To account for these contradictory roles, we propose a model in which GABA produced by GAD1 in the RTN neuronal somata is released extrasynaptically and mediates intra-RTN inhibition.
Collapse
Affiliation(s)
- Dongyu Liu
- Department of Physiology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Kazuyuki Fujihara
- Department of Psychiatry and Neuroscience, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Yuchio Yanagawa
- Department of Genetic and Behavioral Neuroscience, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Hajime Mushiake
- Department of Physiology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Tomokazu Ohshiro
- Department of Physiology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| |
Collapse
|
10
|
Beltrán-Matas P, Hartveit E, Veruki ML. Functional properties of GABA A receptors of AII amacrine cells of the rat retina. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1134765. [PMID: 38983040 PMCID: PMC11182327 DOI: 10.3389/fopht.2023.1134765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/08/2023] [Indexed: 07/11/2024]
Abstract
Amacrine cells are a highly diverse group of inhibitory retinal interneurons that sculpt the responses of bipolar cells, ganglion cells, and other amacrine cells. They integrate excitatory inputs from bipolar cells and inhibitory inputs from other amacrine cells, but for most amacrine cells, little is known about the specificity and functional properties of their inhibitory inputs. Here, we have investigated GABAA receptors of the AII amacrine, a critical neuron in the rod pathway microcircuit, using patch-clamp recording in rat retinal slices. Puffer application of GABA evoked robust responses, but, surprisingly, spontaneous GABAA receptor-mediated postsynaptic currents were not observed, neither under control conditions nor following application of high-K+ solution to facilitate release. To investigate the biophysical and pharmacological properties of GABAA receptors in AIIs, we therefore used nucleated patches and a fast application system. Both brief and long pulses of GABA (3 mM) evoked GABAA receptor-mediated currents with slow, multi-exponential decay kinetics. The average weighted time constant (τw) of deactivation was ~163 ms. Desensitization was even slower, with τw ~330 ms. Non-stationary noise analysis of patch responses and directly observed channel gating yielded a single-channel conductance of ~23 pS. Pharmacological investigation suggested the presence of α2 and/or α3 subunits, as well as the γ2 subunit. Such subunit combinations are typical of GABAA receptors with slow kinetics. If synaptic GABAA receptors of AII amacrines have similar functional properties, the slow deactivation and desensitization kinetics will facilitate temporal summation of GABAergic inputs, allowing effective summation and synaptic integration to occur even for relatively low frequencies of inhibitory inputs.
Collapse
Affiliation(s)
| | - Espen Hartveit
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | |
Collapse
|
11
|
β-Adrenoreceptors as Therapeutic Targets for Ocular Tumors and Other Eye Diseases-Historical Aspects and Nowadays Understanding. Int J Mol Sci 2023; 24:ijms24054698. [PMID: 36902129 PMCID: PMC10003534 DOI: 10.3390/ijms24054698] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/21/2023] [Accepted: 02/24/2023] [Indexed: 03/05/2023] Open
Abstract
β-adrenoreceptors (ARs) are members of the superfamily of G-protein-coupled receptors (GPCRs), and are activated by catecholamines, such as epinephrine and norepinephrine. Three subtypes of β-ARs (β1, β2, and β3) have been identified with different distributions among ocular tissues. Importantly, β-ARs are an established target in the treatment of glaucoma. Moreover, β-adrenergic signaling has been associated with the development and progression of various tumor types. Hence, β-ARs are a potential therapeutic target for ocular neoplasms, such as ocular hemangioma and uveal melanoma. This review aims to discuss the expression and function of individual β-AR subtypes in ocular structures, as well as their role in the treatment of ocular diseases, including ocular tumors.
Collapse
|
12
|
Liu H, Schaeffel F, Yang Z, Feldkaemper MP. GABAB Receptor Activation Affects Eye Growth in Chickens with Visually Induced Refractive Errors. Biomolecules 2023; 13:biom13030434. [PMID: 36979369 PMCID: PMC10046083 DOI: 10.3390/biom13030434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 02/08/2023] [Accepted: 02/13/2023] [Indexed: 03/02/2023] Open
Abstract
This study aims to explore the role of GABAB receptors in the development of deprivation myopia (DM), lens-induced myopia (LIM) and lens-induced hyperopia (LIH). Chicks were intravitreally injected with 25 µg baclofen (GABABR agonist) in one eye and saline into the fellow eye. Choroidal thickness (ChT) was measured via OCT before and 2, 4, 6, 8, 24 h after injection. ChT decreased strongly at 6 and 8 h after baclofen injection and returned back to baseline level after 24 h. Moreover, chicks were monocularly treated with translucent diffusers, −7D or +7D lenses and randomly assigned to baclofen or saline treatment. DM chicks were injected daily into both eyes, while LIM and LIH chicks were monocularly injected into the lens-wearing eyes, for 4 days. Refractive error, axial length and ChT were measured before and after treatment. Dopamine and its metabolites were analyzed via HPLC. Baclofen significantly reduced the myopic shift and eye growth in DM and LIM eyes. However, it did not change ChT compared to respective saline-injected eyes. On the other hand, baclofen inhibited the hyperopic shift and choroidal thickening in LIH eyes. All the baclofen-injected eyes showed significantly lower vitreal DOPAC content. Since GABA is an inhibitory ubiquitous neurotransmitter, interfering with its signaling affects spatial retinal processing and therefore refractive error development with both diffusers and lenses.
Collapse
Affiliation(s)
- Hong Liu
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, 72076 Tuebingen, Germany
- Aier Institute of Optometry and Vision Science, Aier Eye Hospital Group, Changsha 410000, China
| | - Frank Schaeffel
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, 72076 Tuebingen, Germany
- Myopia Research Group, Institute of Molecular and Clinical Ophthalmology Basel (IOB), 4031 Basel, Switzerland
| | - Zhikuan Yang
- Aier Institute of Optometry and Vision Science, Aier Eye Hospital Group, Changsha 410000, China
- Hunan Province Optometry Engineering and Technology Research Center, Changsha 410000, China
- Hunan Province International Cooperation Base for Optometry Science and Technology, Changsha 410000, China
- Correspondence: (Z.Y.); (M.P.F.)
| | - Marita Pauline Feldkaemper
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, 72076 Tuebingen, Germany
- Correspondence: (Z.Y.); (M.P.F.)
| |
Collapse
|
13
|
Raja S, Milosavljevic N, Allen AE, Cameron MA. Burning the candle at both ends: Intraretinal signaling of intrinsically photosensitive retinal ganglion cells. Front Cell Neurosci 2023; 16:1095787. [PMID: 36687522 PMCID: PMC9853061 DOI: 10.3389/fncel.2022.1095787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 12/13/2022] [Indexed: 01/09/2023] Open
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs) are photoreceptors located in the ganglion cell layer. They project to brain regions involved in predominately non-image-forming functions including entrainment of circadian rhythms, control of the pupil light reflex, and modulation of mood and behavior. In addition to possessing intrinsic photosensitivity via the photopigment melanopsin, these cells receive inputs originating in rods and cones. While most research in the last two decades has focused on the downstream influence of ipRGC signaling, recent studies have shown that ipRGCs also act retrogradely within the retina itself as intraretinal signaling neurons. In this article, we review studies examining intraretinal and, in addition, intraocular signaling pathways of ipRGCs. Through these pathways, ipRGCs regulate inner and outer retinal circuitry through both chemical and electrical synapses, modulate the outputs of ganglion cells (both ipRGCs and non-ipRGCs), and influence arrangement of the correct retinal circuitry and vasculature during development. These data suggest that ipRGC function plays a significant role in the processing of image-forming vision at its earliest stage, positioning these photoreceptors to exert a vital role in perceptual vision. This research will have important implications for lighting design to optimize the best chromatic lighting environments for humans, both in adults and potentially even during fetal and postnatal development. Further studies into these unique ipRGC signaling pathways could also lead to a better understanding of the development of ocular dysfunctions such as myopia.
Collapse
Affiliation(s)
- Sushmitha Raja
- School of Medicine, Western Sydney University, Sydney, NSW, Australia
| | - Nina Milosavljevic
- Division of Neuroscience, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Annette E. Allen
- Division of Neuroscience, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Morven A. Cameron
- School of Medicine, Western Sydney University, Sydney, NSW, Australia,*Correspondence: Morven A. Cameron,
| |
Collapse
|
14
|
Wilmet B, Callebert J, Duvoisin R, Goulet R, Tourain C, Michiels C, Frederiksen H, Schaeffel F, Marre O, Sahel JA, Audo I, Picaud S, Zeitz C. Mice Lacking Gpr179 with Complete Congenital Stationary Night Blindness Are a Good Model for Myopia. Int J Mol Sci 2022; 24:ijms24010219. [PMID: 36613663 PMCID: PMC9820543 DOI: 10.3390/ijms24010219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 12/25/2022] Open
Abstract
Mutations in GPR179 are one of the most common causes of autosomal recessive complete congenital stationary night blindness (cCSNB). This retinal disease is characterized in patients by impaired dim and night vision, associated with other ocular symptoms, including high myopia. cCSNB is caused by a complete loss of signal transmission from photoreceptors to ON-bipolar cells. In this study, we hypothesized that the lack of Gpr179 and the subsequent impaired ON-pathway could lead to myopic features in a mouse model of cCSNB. Using ultra performance liquid chromatography, we show that adult Gpr179-/- mice have a significant decrease in both retinal dopamine and 3,4-dihydroxyphenylacetic acid, compared to Gpr179+/+ mice. This alteration of the dopaminergic system is thought to be correlated with an increased susceptibility to lens-induced myopia but does not affect the natural refractive development. Altogether, our data added a novel myopia model, which could be used to identify therapeutic interventions.
Collapse
Affiliation(s)
- Baptiste Wilmet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
- Correspondence: (B.W.); (C.Z.); Tel.: +33-1-53-46-25-26 (B.W.); +33-1-53-46-25-40 (C.Z.)
| | - Jacques Callebert
- Service of Biochemistry and Molecular Biology, INSERM U942, Hospital Lariboisière, AP-HP, 75010 Paris, France
| | - Robert Duvoisin
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
| | - Ruben Goulet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Christophe Tourain
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
- Wavefront-Engineering Microscopy Group, Neurophotonics Laboratory, CNRS UMR8250, Paris Descartes University, 75270 Paris, France
| | - Christelle Michiels
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Helen Frederiksen
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Frank Schaeffel
- Institute of Molecular and Clinical Ophthalmology Basel (IOB), 4056 Basel, Switzerland
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, 72076 Tuebingen, Germany
- Zeiss Vision Lab, Ophthalmic Research Institute, University of Tuebingen, 72076 Tuebingen, Germany
| | - Olivier Marre
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - José Alain Sahel
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
- Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Centre Hospitalier National d’Ophtalmologie des Quinze-Vingts, INSERM-DGOS CIC 1423, 75012 Paris, France
- Fondation Ophtalmologique Adolphe de Rothschild, 75019 Paris, France
- Académie des Sciences, Institut de France, 75006 Paris, France
| | - Isabelle Audo
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
- Centre Hospitalier National d’Ophtalmologie des Quinze-Vingts, INSERM-DGOS CIC 1423, 75012 Paris, France
| | - Serge Picaud
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Christina Zeitz
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
- Correspondence: (B.W.); (C.Z.); Tel.: +33-1-53-46-25-26 (B.W.); +33-1-53-46-25-40 (C.Z.)
| |
Collapse
|
15
|
Huang W, Xu Q, Liu F, Su J, Xiao D, Tang L, Hao ZZ, Liu R, Xiang K, Bi Y, Miao Z, Liu X, Liu Y, Liu S. Identification of TPBG-Expressing Amacrine Cells in DAT-tdTomato Mouse. Invest Ophthalmol Vis Sci 2022; 63:13. [PMID: 35551574 PMCID: PMC9123489 DOI: 10.1167/iovs.63.5.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/25/2022] [Indexed: 11/29/2022] Open
Abstract
Purpose Neurons are the bricks of the neuronal system and experimental access to certain neuron subtypes will be of great help to decipher neuronal circuits. Here, we identified trophoblast glycoprotein (TPBG)-expressing GABAergic amacrine cells (ACs) that were selectively labeled in DAT-tdTomato transgenic mice. Methods Retina and brain sections were prepared for immunostaining with antibodies against various biomarkers. Patch-sequencing was performed to obtain the transcriptomes of tdTomato-positive cells in DAT-tdTomato mice. Whole-cell recordings were conducted to identify responses to light stimulation. Results Tyrosine hydroxylase immunoreactive cells were colocalized with tdTomato-positive cells in substantia nigra pars compacta, but not in the retina. Transcriptomes collected from tdTomato-positive cells in retinas via Patch-sequencing exhibited the expression of marker genes of ACs (Pax6 and Slc32a1) and marker genes of GABAergic neurons (Gad1, Gad2, and Slc6a1). Immunostaining with antibodies against relevant proteins (GAD67, GAD65, and GABA) also confirmed transcriptomic results. Furthermore, tdTomato-positive cells in retinas selectively expressed Tpbg, a marker gene for distinct clusters molecularly defined, which was proved with TPBG immunoreactivity in fluorescently labeled cells. Finally, tdTomato-positive cells recorded showed ON-OFF responses to light stimulation. Conclusions Ectopic expression occurs in the retina but not in the substantia nigra pars compacta in the DAT-tdTomato mouse, and fluorescently labeled cells in the retina are TPBG-expressing GABAergic ACs. This type of transgenic mice has been proved as an ideal tool to achieve efficient labeling of a distinct subset of ACs that selectively express Tpbg.
Collapse
Affiliation(s)
- Wanjing Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Qiang Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Feng Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jing Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Dongchang Xiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Lei Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Zhao-Zhe Hao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Ruifeng Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Kangjian Xiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yalan Bi
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge CB10 1SD, United Kingdom
| | - Zhichao Miao
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge CB10 1SD, United Kingdom
- Translational Research Institute of Brain and Brain-Like Intelligence and Department of Anesthesiology, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Xialin Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yizhi Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
- Research Unit of Ocular Development and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Sheng Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Guangzhou, China
| |
Collapse
|
16
|
De-Miguel FF. The Thermodynamically Expensive Contribution of Three Calcium Sources to Somatic Release of Serotonin. Int J Mol Sci 2022; 23:1495. [PMID: 35163419 PMCID: PMC8836226 DOI: 10.3390/ijms23031495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/28/2021] [Accepted: 12/30/2021] [Indexed: 02/01/2023] Open
Abstract
The soma, dendrites and axon of neurons may display calcium-dependent release of transmitters and peptides. Such release is named extrasynaptic for occurring in absence of synaptic structures. This review describes the cooperative actions of three calcium sources on somatic exocytosis. Emphasis is given to the somatic release of serotonin by the classical leech Retzius neuron, which has allowed detailed studies on the fine steps from excitation to exocytosis. Trains of action potentials induce transmembrane calcium entry through L-type channels. For action potential frequencies above 5 Hz, summation of calcium transients on individual action potentials activates the second calcium source: ryanodine receptors produce calcium-induced calcium release. The resulting calcium tsunami activates mitochondrial ATP synthesis to fuel transport of vesicles to the plasma membrane. Serotonin that is released maintains a large-scale exocytosis by activating the third calcium source: serotonin autoreceptors coupled to phospholipase C promote IP3 production. Activated IP3 receptors in peripheral endoplasmic reticulum release calcium that promotes vesicle fusion. The Swiss-clock workings of the machinery for somatic exocytosis has a striking disadvantage. The essential calcium-releasing endoplasmic reticulum near the plasma membrane hinders the vesicle transport, drastically reducing the thermodynamic efficiency of the ATP expenses and elevating the energy cost of release.
Collapse
Affiliation(s)
- Francisco F De-Miguel
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
17
|
Liu Y, Wang Y, Xiao Y, Li X, Ruan S, Luo X, Wan X, Wang F, Sun X. Retinal degeneration in mice lacking the cyclic nucleotide-gated channel subunit CNGA1. FASEB J 2021; 35:e21859. [PMID: 34418172 DOI: 10.1096/fj.202101004r] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/28/2021] [Accepted: 08/02/2021] [Indexed: 12/16/2022]
Abstract
Cyclic nucleotide-gated (CNG) channels are important mediators in the transduction pathways of rod and cone photoreceptors. Native CNG channels are heterotetramers composed of homologous A and B subunits. Biallelic mutations in CNGA1 or CNGB1 genes result in autosomal recessive retinitis pigmentosa (RP). To investigate the pathogenic mechanism of CNG channel-associated retinal degeneration, we developed a mouse model of CNGA1 knock-out using CRISPR/Cas9 technology. We observed progressive retinal thinning and a concomitant functional deficit in vivo as typical phenotypes for RP. Immunofluorescence and TUNEL staining showed progressive degeneration in rods and cones. Moreover, microglial activation and oxidative stress damage occurred in parallel. RNA-sequencing analysis of the retinae suggested down-regulated synaptic transmission and phototransduction as early as 9 days postnatal, possibly inducing later photoreceptor degeneration. In addition, the down-regulated PI3K-AKT-mTOR pathway indicated upregulation of autophagic process, and chaperone-mediated autophagy was further shown to coincide with the time course of photoreceptor death. Taken together, our studies add to a growing body of research exploring the mechanisms of photoreceptor death during RP progression and provide a novel CNGA1 knockout mouse model for potential development of therapies.
Collapse
Affiliation(s)
- Yang Liu
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yafang Wang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yushu Xiao
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaomeng Li
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shang Ruan
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xueting Luo
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
| | - Xiaoling Wan
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
| | - Fenghua Wang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.,National Clinical Research Center for Eye Diseases, Shanghai, China.,Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Xiaodong Sun
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.,National Clinical Research Center for Eye Diseases, Shanghai, China.,Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| |
Collapse
|
18
|
De-Miguel FF, Leon-Pinzon C, Torres-Platas SG, Del-Pozo V, Hernández-Mendoza GA, Aguirre-Olivas D, Méndez B, Moore S, Sánchez-Sugía C, García-Aguilera MA, Martínez-Valencia A, Ramírez-Santiago G, Rubí JM. Extrasynaptic Communication. Front Mol Neurosci 2021; 14:638858. [PMID: 33994942 PMCID: PMC8119753 DOI: 10.3389/fnmol.2021.638858] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 04/01/2021] [Indexed: 12/18/2022] Open
Abstract
Streams of action potentials or long depolarizations evoke a massive exocytosis of transmitters and peptides from the surface of dendrites, axons and cell bodies of different neuron types. Such mode of exocytosis is known as extrasynaptic for occurring without utilization of synaptic structures. Most transmitters and all peptides can be released extrasynaptically. Neurons may discharge their contents with relative independence from the axon, soma and dendrites. Extrasynaptic exocytosis takes fractions of a second in varicosities or minutes in the soma or dendrites, but its effects last from seconds to hours. Unlike synaptic exocytosis, which is well localized, extrasynaptic exocytosis is diffuse and affects neuronal circuits, glia and blood vessels. Molecules that are liberated may reach extrasynaptic receptors microns away. The coupling between excitation and exocytosis follows a multistep mechanism, different from that at synapses, but similar to that for the release of hormones. The steps from excitation to exocytosis have been studied step by step for the vital transmitter serotonin in leech Retzius neurons. The events leading to serotonin exocytosis occur similarly for the release of other transmitters and peptides in central and peripheral neurons. Extrasynaptic exocytosis occurs commonly onto glial cells, which react by releasing the same or other transmitters. In the last section, we discuss how illumination of the retina evokes extrasynaptic release of dopamine and ATP. Dopamine contributes to light-adaptation; ATP activates glia, which mediates an increase in blood flow and oxygenation. A proper understanding of the workings of the nervous system requires the understanding of extrasynaptic communication.
Collapse
Affiliation(s)
- Francisco F De-Miguel
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, México City, Mexico.,Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, México City, Mexico
| | - Carolina Leon-Pinzon
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, México City, Mexico
| | - Susana G Torres-Platas
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, México City, Mexico
| | - Vanessa Del-Pozo
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, México City, Mexico
| | | | - Dilia Aguirre-Olivas
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, México City, Mexico
| | - Bruno Méndez
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, México City, Mexico
| | - Sharlen Moore
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, México City, Mexico
| | - Celeste Sánchez-Sugía
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, México City, Mexico
| | | | | | | | - J Miguel Rubí
- Facultat de Fisica, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
19
|
The Role of Adrenoceptors in the Retina. Cells 2020; 9:cells9122594. [PMID: 33287335 PMCID: PMC7761662 DOI: 10.3390/cells9122594] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/29/2020] [Accepted: 12/01/2020] [Indexed: 01/16/2023] Open
Abstract
The retina is a part of the central nervous system, a thin multilayer with neuronal lamination, responsible for detecting, preprocessing, and sending visual information to the brain. Many retinal diseases are characterized by hemodynamic perturbations and neurodegeneration leading to vision loss and reduced quality of life. Since catecholamines and respective bindings sites have been characterized in the retina, we systematically reviewed the literature with regard to retinal expression, distribution and function of alpha1 (α1)-, alpha2 (α2)-, and beta (β)-adrenoceptors (ARs). Moreover, we discuss the role of the individual adrenoceptors as targets for the treatment of retinal diseases.
Collapse
|
20
|
Hirano AA, Vuong HE, Kornmann HL, Schietroma C, Stella SL, Barnes S, Brecha NC. Vesicular Release of GABA by Mammalian Horizontal Cells Mediates Inhibitory Output to Photoreceptors. Front Cell Neurosci 2020; 14:600777. [PMID: 33335476 PMCID: PMC7735995 DOI: 10.3389/fncel.2020.600777] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/04/2020] [Indexed: 12/14/2022] Open
Abstract
Feedback inhibition by horizontal cells regulates rod and cone photoreceptor calcium channels that control their release of the neurotransmitter glutamate. This inhibition contributes to synaptic gain control and the formation of the center-surround antagonistic receptive fields passed on to all downstream neurons, which is important for contrast sensitivity and color opponency in vision. In contrast to the plasmalemmal GABA transporter found in non-mammalian horizontal cells, there is evidence that the mechanism by which mammalian horizontal cells inhibit photoreceptors involves the vesicular release of the inhibitory neurotransmitter GABA. Historically, inconsistent findings of GABA and its biosynthetic enzyme, L-glutamate decarboxylase (GAD) in horizontal cells, and the apparent lack of surround response block by GABAergic agents diminished support for GABA's role in feedback inhibition. However, the immunolocalization of the vesicular GABA transporter (VGAT) in the dendritic and axonal endings of horizontal cells that innervate photoreceptor terminals suggested GABA was released via vesicular exocytosis. To test the idea that GABA is released from vesicles, we localized GABA and GAD, multiple SNARE complex proteins, synaptic vesicle proteins, and Cav channels that mediate exocytosis to horizontal cell dendritic tips and axonal terminals. To address the perceived relative paucity of synaptic vesicles in horizontal cell endings, we used conical electron tomography on mouse and guinea pig retinas that revealed small, clear-core vesicles, along with a few clathrin-coated vesicles and endosomes in horizontal cell processes within photoreceptor terminals. Some small-diameter vesicles were adjacent to the plasma membrane and plasma membrane specializations. To assess vesicular release, a functional assay involving incubation of retinal slices in luminal VGAT-C antibodies demonstrated vesicles fused with the membrane in a depolarization- and calcium-dependent manner, and these labeled vesicles can fuse multiple times. Finally, targeted elimination of VGAT in horizontal cells resulted in a loss of tonic, autaptic GABA currents, and of inhibitory feedback modulation of the cone photoreceptor Cai, consistent with the elimination of GABA release from horizontal cell endings. These results in mammalian retina identify the central role of vesicular release of GABA from horizontal cells in the feedback inhibition of photoreceptors.
Collapse
Affiliation(s)
- Arlene A. Hirano
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Helen E. Vuong
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Helen L. Kornmann
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Cataldo Schietroma
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Salvatore L. Stella
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Steven Barnes
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Doheny Eye Institute, University of California, Los Angeles, Los Angeles, CA, United States
| | - Nicholas C. Brecha
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Stein Eye Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
21
|
Grünert U, Martin PR. Cell types and cell circuits in human and non-human primate retina. Prog Retin Eye Res 2020; 78:100844. [PMID: 32032773 DOI: 10.1016/j.preteyeres.2020.100844] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/28/2020] [Accepted: 01/31/2020] [Indexed: 12/12/2022]
Abstract
This review summarizes our current knowledge of primate including human retina focusing on bipolar, amacrine and ganglion cells and their connectivity. We have two main motivations in writing. Firstly, recent progress in non-invasive imaging methods to study retinal diseases mean that better understanding of the primate retina is becoming an important goal both for basic and for clinical sciences. Secondly, genetically modified mice are increasingly used as animal models for human retinal diseases. Thus, it is important to understand to which extent the retinas of primates and rodents are comparable. We first compare cell populations in primate and rodent retinas, with emphasis on how the fovea (despite its small size) dominates the neural landscape of primate retina. We next summarise what is known, and what is not known, about the postreceptoral neurone populations in primate retina. The inventories of bipolar and ganglion cells in primates are now nearing completion, comprising ~12 types of bipolar cell and at least 17 types of ganglion cell. Primate ganglion cells show clear differences in dendritic field size across the retina, and their morphology differs clearly from that of mouse retinal ganglion cells. Compared to bipolar and ganglion cells, amacrine cells show even higher morphological diversity: they could comprise over 40 types. Many amacrine types appear conserved between primates and mice, but functions of only a few types are understood in any primate or non-primate retina. Amacrine cells appear as the final frontier for retinal research in monkeys and mice alike.
Collapse
Affiliation(s)
- Ulrike Grünert
- The University of Sydney, Save Sight Institute, Faculty of Medicine and Health, Sydney, NSW, 2000, Australia; Australian Research Council Centre of Excellence for Integrative Brain Function, Sydney Node, The University of Sydney, Sydney, NSW, 2000, Australia.
| | - Paul R Martin
- The University of Sydney, Save Sight Institute, Faculty of Medicine and Health, Sydney, NSW, 2000, Australia; Australian Research Council Centre of Excellence for Integrative Brain Function, Sydney Node, The University of Sydney, Sydney, NSW, 2000, Australia
| |
Collapse
|
22
|
Caravaggio F, Worhunsky P, Graff-Guerrero A, Matuskey D. Further in vivo characterization of [ 11 C]-(+)-PHNO uptake into a retina-like region of interest in humans. Synapse 2019; 74:e22135. [PMID: 31553807 DOI: 10.1002/syn.22135] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 09/20/2019] [Indexed: 11/12/2022]
Abstract
The neurotransmitter dopamine is present in the retina and is involved in several modulatory functions. Unlike in rodents, dopamine D3 receptors are expressed in the retina of humans. Recently, uptake of the D3 receptor-preferring radiotracer [11 C]-(+)-PHNO has been observed in a retina-like region of interest (ROI) in humans. Here, we attempted to quantify [11 C]-(+)-PHNO uptake into this ROI using an independent sample, employing an extended scan acquisition time (120 min) and arterial kinetic modeling. Data from 14 healthy controls were analyzed (Mean Age: 38.41 ± 9.55, 3 female), 8 of which provided arterial line input function data (Mean Age: 41.07 ± 7.82, 3 female). Using Ichise's multilinear analysis (MA1) method, it was possible to quantify the volume of distribution (VT ) of [11 C]-(+)-PHNO in this retina-like region (Mean VT = 13.56 ± 3.52; Mean χ2 = 2.08 ± 2.20). Notably, the shape of the time activity curve resembled closely that of the globus pallidus. Moreover, the VT values in the retina correlated well with binding potential (BPND ) values calculated using the simplified reference tissue model (Mean BPND = 2.11 ± .94; Mean χ2 = 5.76 ± 2.56), employing the cerebellum as the reference region (r = .76, r2 = .58). In summary, we provide evidence that the in vivo uptake of [11 C]-(+)-PHNO into a retina-like ROI in humans can be quantified using both arterial blood sampling (VT ) and simplified reference tissue methods (BPND ).
Collapse
Affiliation(s)
- Fernando Caravaggio
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Patrick Worhunsky
- Department of Psychiatry, Yale University, New Haven, Connecticut, USA
| | - Ariel Graff-Guerrero
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - David Matuskey
- Department of Psychiatry, Yale University, New Haven, Connecticut, USA.,PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut, USA.,Department of Neurology, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
23
|
Noguez P, Rubí JM, De-Miguel FF. Thermodynamic Efficiency of Somatic Exocytosis of Serotonin. Front Physiol 2019; 10:473. [PMID: 31214038 PMCID: PMC6554442 DOI: 10.3389/fphys.2019.00473] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 04/04/2019] [Indexed: 01/09/2023] Open
Abstract
Through somatic exocytosis neurons liberate immense amounts of transmitter molecules that modulate the functioning of the nervous system. A stream of action potentials triggers an ATP-dependent transport of transmitter-containing vesicles to the plasma membrane, that ends with a large-scale exocytosis. It is commonly assumed that biological processes use metabolic energy with a high thermodynamic efficiency, meaning that most energy generates work with minor dissipation. However, the intricate ultrastructure underlying the pathway for the vesicle flow necessary for somatic exocytosis challenges this possibility. To study this problem here we first applied thermodynamic theory to quantify the efficiency of somatic exocytosis of the vital transmitter serotonin. Then we correlated the efficiency to the ultrastructure of the transport pathway of the vesicles. Exocytosis was evoked in cultured Retzius neurons of the leech by trains of 10 impulses delivered at 20 Hz. The kinetics of exocytosis was quantified from the gradual fluorescence increase of FM1-43 dye as it became incorporated into vesicles that underwent their exo-endocytosis cycle. By fitting a model of the vesicle transport carried by motor forces to the kinetics of exocytosis, we calculated the thermodynamic efficiency of the ATP expenses per vesicle, as the power of the transport divided by total energy ideally produced by the hydrolysis of ATP during the process. The efficiency was remarkably low (0.1-6.4%) and the values formed a W-shape distribution with the transport distances of the vesicles. Electron micrographs and fluorescent staining of the actin cortex indicated that the slopes of the W chart could be explained by the interaction of vesicles with the actin cortex and the calcium-releasing endoplasmic reticulum. We showed that the application of thermodynamic theory permitted to predict aspects of the intracellular structure. Our results suggest that the distribution of subcellular structures that are essential for somatic exocytosis abates the thermodynamic efficiency of the transport by hampering vesicle mobilization. It is remarkable that the modulation of the nervous system occurs at the expenses of an efficient use of metabolic energy.
Collapse
Affiliation(s)
- Paula Noguez
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - J Miguel Rubí
- Facultat de Física, Universitat de Barcelona, Barcelona, Spain
| | - Francisco F De-Miguel
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
24
|
Molecular and Cellular Mechanisms Underlying Somatostatin-Based Signaling in Two Model Neural Networks, the Retina and the Hippocampus. Int J Mol Sci 2019; 20:ijms20102506. [PMID: 31117258 PMCID: PMC6566141 DOI: 10.3390/ijms20102506] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/16/2019] [Accepted: 05/20/2019] [Indexed: 02/06/2023] Open
Abstract
Neural inhibition plays a key role in determining the specific computational tasks of different brain circuitries. This functional "braking" activity is provided by inhibitory interneurons that use different neurochemicals for signaling. One of these substances, somatostatin, is found in several neural networks, raising questions about the significance of its widespread occurrence and usage. Here, we address this issue by analyzing the somatostatinergic system in two regions of the central nervous system: the retina and the hippocampus. By comparing the available information on these structures, we identify common motifs in the action of somatostatin that may explain its involvement in such diverse circuitries. The emerging concept is that somatostatin-based signaling, through conserved molecular and cellular mechanisms, allows neural networks to operate correctly.
Collapse
|
25
|
Rod Photoreceptor Activation Alone Defines the Release of Dopamine in the Retina. Curr Biol 2019; 29:763-774.e5. [PMID: 30799247 DOI: 10.1016/j.cub.2019.01.042] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/27/2018] [Accepted: 01/15/2019] [Indexed: 02/07/2023]
Abstract
Retinal dopamine is released by a specialized subset of amacrine cells in response to light and has a potent influence on how the retina responds to, and encodes, visual information. Here, we address the critical question of which retinal photoreceptor is responsible for coordinating the release of this neuromodulator. Although all three photoreceptor classes-rods, cones, and melanopsin-containing retinal ganglion cells (mRGCs)-have been shown to provide electrophysiological inputs to dopaminergic amacrine cells (DACs), we show here that the release of dopamine is defined only by rod photoreceptors. Remarkably, this rod signal coordinates both a suppressive signal at low intensities and drives dopamine release at very bright light intensities. These data further reveal that dopamine release does not necessarily correlate with electrophysiological activity of DACs and add to a growing body of evidence that rods define aspects of retinal function at very bright light levels.
Collapse
|
26
|
Abstract
The brain hosts a vast and diverse repertoire of neuropeptides, a class of signalling molecules often described as neurotransmitters. Here I argue that this description entails a catalogue of misperceptions, misperceptions that feed into a narrative in which information processing in the brain can be understood only through mapping neuronal connectivity and by studying the transmission of electrically conducted signals through chemical synapses. I argue that neuropeptide signalling in the brain involves primarily autocrine, paracrine and neurohormonal mechanisms that do not depend on synaptic connectivity and that it is not solely dependent on electrical activity but on mechanisms analogous to secretion from classical endocrine cells. As in classical endocrine systems, to understand the role of neuropeptides in the brain, we must understand not only how their release is regulated, but also how their synthesis is regulated and how the sensitivity of their targets is regulated. We must also understand the full diversity of effects of neuropeptides on those targets, including their effects on gene expression.
Collapse
Affiliation(s)
- Gareth Leng
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Correspondence should be addressed to G Leng:
| |
Collapse
|
27
|
Sankaran M, Keeley PW, He L, Iuvone PM, Reese BE. Dopaminergic amacrine cell number, plexus density, and dopamine content in the mouse retina: Strain differences and effects of Bax gene disruption. Exp Eye Res 2018; 177:208-212. [PMID: 30240584 DOI: 10.1016/j.exer.2018.09.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 08/07/2018] [Accepted: 09/17/2018] [Indexed: 01/01/2023]
Abstract
Many types of retinal neuron modulate the distribution of their processes to ensure a uniform coverage of the retinal surface. Dendritic field area, for instance, is inversely related to the variation in cellular density for many cell types, observed either across retinal eccentricity or between different strains of mice that differ in cell number. Dopaminergic amacrine (DA) cells, by contrast, have dendritic arbors that bear no spatial relationship to the presence of their immediate homotypic neighbors, yet it remains to be determined whether their coverage upon the retina, as a population, is conserved across variation in their total number. The present study assessed the overall density of the dopaminergic plexus in the inner plexiform layer in the presence of large variation in the total number of DA cells, as well as their retinal dopamine content, to determine whether either of these features is conserved. We first compared these traits between two strains of mice (C57BL/6J and A/J) that exhibit a two-fold difference in DA cell number. We subsequently examined these same traits in littermate mice for which the pro-apoptotic Bax gene was either intact or knocked out, yielding a five-fold difference in DA cell number. In both comparisons, we found greater plexus density and DA content in the strain or condition with the greater number of DA cells. The population of DA cells, therefore, does not appear to self-regulate its process coverage to achieve a constant density as the DA mosaic is established during development, nor its functional dopamine content in maturity.
Collapse
Affiliation(s)
- Mathangi Sankaran
- Neuroscience Research Institute, University of California, Santa Barbara, CA, 93106, USA; Department of Psychological & Brain Sciences, University of California, Santa Barbara, CA, 93106, USA
| | - Patrick W Keeley
- Department of Psychological & Brain Sciences, University of California, Santa Barbara, CA, 93106, USA
| | - Li He
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - P Michael Iuvone
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, GA, 30322, USA; Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Benjamin E Reese
- Neuroscience Research Institute, University of California, Santa Barbara, CA, 93106, USA; Department of Psychological & Brain Sciences, University of California, Santa Barbara, CA, 93106, USA.
| |
Collapse
|
28
|
Caravaggio F, Scifo E, Sibille EL, Hernandez-Da Mota SE, Gerretsen P, Remington G, Graff-Guerrero A. Expression of dopamine D2 and D3 receptors in the human retina revealed by positron emission tomography and targeted mass spectrometry. Exp Eye Res 2018; 175:32-41. [PMID: 29883636 DOI: 10.1016/j.exer.2018.06.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/24/2018] [Accepted: 06/04/2018] [Indexed: 11/26/2022]
Abstract
Dopamine D2 receptors (D2R) are expressed in the human retina and play an important role in the modulation of neural responses to light-adaptation. However, it is unknown whether dopamine D3 receptors (D3R) are expressed in the human retina. Using positron emission tomography (PET), we have observed significant uptake of the D3R-preferring agonist radiotracer [11C]-(+)-PHNO into the retina of humans in vivo. This led us to examine whether [11C]-(+)-PHNO binding in the retina was quantifiable using reference tissue methods and if D3R are expressed in human post-mortem retinal tissue. [11C]-(+)-PHNO data from 49 healthy controls (mean age: 39.96 ± 14.36; 16 female) and 12 antipsychotic-naïve patients with schizophrenia (mean age: 25.75 ± 6.25; 4 female) were analyzed. We observed no differences in [11C]-(+)-PHNO binding in the retina between first-episode, drug-naïve patients with schizophrenia and healthy controls. Post-mortem retinal tissues from four healthy persons (mean age: 59.75 ± 9.11; 2 female) and four patients with schizophrenia (mean age: 54 ± 17.11; 2 female) were analyzed using a targeted mass spectrometry technique: parallel reaction monitoring (PRM) analysis. Using targeted mass spectrometry, we confirmed that D3R are expressed in human retinal tissue ex vivo. Notably, there was far greater expression of D2R relative to D3R in the healthy human retina (∼12:1). Moreover, PRM analysis revealed reduced D2R, but not D3R, expression in the retinas of non-first episode patients with schizophrenia compared to healthy controls. We confirm that D3R are expressed in the human retina. Future studies are needed to determine what proportion of the [11C]-(+)-PHNO signal in the human retina in vivo is due to binding to D3R versus D2R. Knowledge that both D2R and D3R are expressed in the human retina, and potentially quantifiable in vivo using [11C]-(+)-PHNO, poses new research avenues for better understanding the role of retinal dopamine in human vision. This work may have important implications for elucidating pathophysiological and antipsychotic induced visual deficits in schizophrenia.
Collapse
Affiliation(s)
- Fernando Caravaggio
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, M5T 1R8, Canada; Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario, M5T 1R8, Canada.
| | - Enzo Scifo
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, M5T 1R8, Canada; Molecular and Cellular Cognition Lab, German Center for Neurodegenerative Diseases(DZNE), Bonn, Germany
| | - Etienne L Sibille
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, M5T 1R8, Canada; Department of Pharmacology and Toxicology, University of Toronto, 250 College Street, Toronto, Ontario, M5T 1R8, Canada
| | | | - Philip Gerretsen
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, M5T 1R8, Canada; Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario, M5T 1R8, Canada
| | - Gary Remington
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, M5T 1R8, Canada; Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario, M5T 1R8, Canada
| | - Ariel Graff-Guerrero
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, M5T 1R8, Canada; Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario, M5T 1R8, Canada
| |
Collapse
|
29
|
Vaasjo LO, Quintana AM, Habib MR, Mendez de Jesus PA, Croll RP, Miller MW. GABA-like immunoreactivity in Biomphalaria: Colocalization with tyrosine hydroxylase-like immunoreactivity in the feeding motor systems of panpulmonate snails. J Comp Neurol 2018; 526:1790-1805. [PMID: 29633264 DOI: 10.1002/cne.24448] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 03/24/2018] [Accepted: 03/26/2018] [Indexed: 12/24/2022]
Abstract
The simpler nervous systems of certain invertebrates provide opportunities to examine colocalized classical neurotransmitters in the context of identified neurons and well defined neural circuits. This study examined the distribution of γ-aminobutyric acid-like immunoreactivity (GABAli) in the nervous system of the panpulmonates Biomphalaria glabrata and Biomphalaria alexandrina, major intermediate hosts for intestinal schistosomiasis. GABAli neurons were localized in the cerebral, pedal, and buccal ganglia of each species. With the exception of a projection to the base of the tentacle, GABAli fibers were confined to the CNS. As GABAli was previously reported to be colocalized with markers for dopamine (DA) in five neurons in the feeding network of the euopisthobranch gastropod Aplysia californica (Díaz-Ríos, Oyola, & Miller, 2002), double-labeling protocols were used to compare the distribution of GABAli with tyrosine hydroxylase immunoreactivity (THli). As in Aplysia, GABAli-THli colocalization was limited to five neurons, all of which were located in the buccal ganglion. Five GABAli-THli cells were also observed in the buccal ganglia of two other intensively studied panpulmonate species, Lymnaea stagnalis and Helisoma trivolvis. These findings indicate that colocalization of the classical neurotransmitters GABA and DA in feeding central pattern generator (CPG) interneurons preceded the divergence of euopisthobranch and panpulmonate taxa. These observations also support the hypothesis that heterogastropod feeding CPG networks exhibit a common universal design.
Collapse
Affiliation(s)
- Lee O Vaasjo
- Institute of Neurobiology and Department of Anatomy & Neurobiology, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
| | - Alexandra M Quintana
- Institute of Neurobiology and Department of Anatomy & Neurobiology, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
| | - Mohamed R Habib
- Medical Malacology Laboratory, Theodor Bilharz Research Institute, Giza, Egypt
| | - Paola A Mendez de Jesus
- Institute of Neurobiology and Department of Anatomy & Neurobiology, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
| | - Roger P Croll
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Mark W Miller
- Institute of Neurobiology and Department of Anatomy & Neurobiology, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
| |
Collapse
|
30
|
Chakraborty R, Ostrin LA, Nickla DL, Iuvone PM, Pardue MT, Stone RA. Circadian rhythms, refractive development, and myopia. Ophthalmic Physiol Opt 2018; 38:217-245. [PMID: 29691928 PMCID: PMC6038122 DOI: 10.1111/opo.12453] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/11/2018] [Indexed: 12/14/2022]
Abstract
PURPOSE Despite extensive research, mechanisms regulating postnatal eye growth and those responsible for ametropias are poorly understood. With the marked recent increases in myopia prevalence, robust and biologically-based clinical therapies to normalize refractive development in childhood are needed. Here, we review classic and contemporary literature about how circadian biology might provide clues to develop a framework to improve the understanding of myopia etiology, and possibly lead to rational approaches to ameliorate refractive errors developing in children. RECENT FINDINGS Increasing evidence implicates diurnal and circadian rhythms in eye growth and refractive error development. In both humans and animals, ocular length and other anatomical and physiological features of the eye undergo diurnal oscillations. Systemically, such rhythms are primarily generated by the 'master clock' in the surpachiasmatic nucleus, which receives input from the intrinsically photosensitive retinal ganglion cells (ipRGCs) through the activation of the photopigment melanopsin. The retina also has an endogenous circadian clock. In laboratory animals developing experimental myopia, oscillations of ocular parameters are perturbed. Retinal signaling is now believed to influence refractive development; dopamine, an important neurotransmitter found in the retina, not only entrains intrinsic retinal rhythms to the light:dark cycle, but it also modulates refractive development. Circadian clocks comprise a transcription/translation feedback control mechanism utilizing so-called clock genes that have now been associated with experimental ametropias. Contemporary clinical research is also reviving ideas first proposed in the nineteenth century that light exposures might impact refraction in children. As a result, properties of ambient lighting are being investigated in refractive development. In other areas of medical science, circadian dysregulation is now thought to impact many non-ocular disorders, likely because the patterns of modern artificial lighting exert adverse physiological effects on circadian pacemakers. How, or if, such modern light exposures and circadian dysregulation contribute to refractive development is not known. SUMMARY The premise of this review is that circadian biology could be a productive area worthy of increased investigation, which might lead to the improved understanding of refractive development and improved therapeutic interventions.
Collapse
Affiliation(s)
- Ranjay Chakraborty
- College of Nursing and Health Sciences, Flinders University, Adelaide, Australia
| | | | | | | | - Machelle T. Pardue
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Decatur
| | - Richard A. Stone
- University of Pennsylvania School of Medicine, Philadelphia, USA
| |
Collapse
|
31
|
Park SJH, Pottackal J, Ke JB, Jun NY, Rahmani P, Kim IJ, Singer JH, Demb JB. Convergence and Divergence of CRH Amacrine Cells in Mouse Retinal Circuitry. J Neurosci 2018; 38:3753-3766. [PMID: 29572434 PMCID: PMC5895998 DOI: 10.1523/jneurosci.2518-17.2018] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 03/01/2018] [Accepted: 03/07/2018] [Indexed: 11/21/2022] Open
Abstract
Inhibitory interneurons sculpt the outputs of excitatory circuits to expand the dynamic range of information processing. In mammalian retina, >30 types of amacrine cells provide lateral inhibition to vertical, excitatory bipolar cell circuits, but functional roles for only a few amacrine cells are well established. Here, we elucidate the function of corticotropin-releasing hormone (CRH)-expressing amacrine cells labeled in Cre-transgenic mice of either sex. CRH cells costratify with the ON alpha ganglion cell, a neuron highly sensitive to positive contrast. Electrophysiological and optogenetic analyses demonstrate that two CRH types (CRH-1 and CRH-3) make GABAergic synapses with ON alpha cells. CRH-1 cells signal via graded membrane potential changes, whereas CRH-3 cells fire action potentials. Both types show sustained ON-type responses to positive contrast over a range of stimulus conditions. Optogenetic control of transmission at CRH-1 synapses demonstrates that these synapses are tuned to low temporal frequencies, maintaining GABA release during fast hyperpolarizations during brief periods of negative contrast. CRH amacrine cell output is suppressed by prolonged negative contrast, when ON alpha ganglion cells continue to receive inhibitory input from converging OFF-pathway amacrine cells; the converging ON- and OFF-pathway inhibition balances tonic excitatory drive to ON alpha cells. Previously, it was demonstrated that CRH-1 cells inhibit firing by suppressed-by-contrast (SbC) ganglion cells during positive contrast. Therefore, divergent outputs of CRH-1 cells inhibit two ganglion cell types with opposite responses to positive contrast. The opposing responses of ON alpha and SbC ganglion cells are explained by differing excitation/inhibition balance in the two circuits.SIGNIFICANCE STATEMENT A goal of neuroscience research is to explain the function of neural circuits at the level of specific cell types. Here, we studied the function of specific types of inhibitory interneurons, corticotropin-releasing hormone (CRH) amacrine cells, in the mouse retina. Genetic tools were used to identify and manipulate CRH cells, which make GABAergic synapses with a well studied ganglion cell type, the ON alpha cell. CRH cells converge with other types of amacrine cells to tonically inhibit ON alpha cells and balance their high level of excitation. CRH cells diverge to different types of ganglion cell, the unique properties of which depend on their balance of excitation and inhibition.
Collapse
Affiliation(s)
| | | | - Jiang-Bin Ke
- Department of Biology, University of Maryland, College Park, Maryland 20742
| | | | | | - In-Jung Kim
- Department of Ophthalmology and Visual Science
- Interdepartmental Neuroscience Program
- Department of Neuroscience
| | - Joshua H Singer
- Department of Biology, University of Maryland, College Park, Maryland 20742
| | - Jonathan B Demb
- Department of Ophthalmology and Visual Science,
- Interdepartmental Neuroscience Program
- Department of Cellular and Molecular Physiology, Yale University, New Haven, Connecticut 06511, and
| |
Collapse
|
32
|
Guidolin D, Marcoli M, Maura G, Agnati LF. New dimensions of connectomics and network plasticity in the central nervous system. Rev Neurosci 2018; 28:113-132. [PMID: 28030363 DOI: 10.1515/revneuro-2016-0051] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 09/20/2016] [Indexed: 12/24/2022]
Abstract
Cellular network architecture plays a crucial role as the structural substrate for the brain functions. Therefore, it represents the main rationale for the emerging field of connectomics, defined as the comprehensive study of all aspects of central nervous system connectivity. Accordingly, in the present paper the main emphasis will be on the communication processes in the brain, namely wiring transmission (WT), i.e. the mapping of the communication channels made by cell components such as axons and synapses, and volume transmission (VT), i.e. the chemical signal diffusion along the interstitial brain fluid pathways. Considering both processes can further expand the connectomics concept, since both WT-connectomics and VT-connectomics contribute to the structure of the brain connectome. A consensus exists that such a structure follows a hierarchical or nested architecture, and macro-, meso- and microscales have been defined. In this respect, however, several lines of evidence indicate that a nanoscale (nano-connectomics) should also be considered to capture direct protein-protein allosteric interactions such as those occurring, for example, in receptor-receptor interactions at the plasma membrane level. In addition, emerging evidence points to novel mechanisms likely playing a significant role in the modulation of intercellular connectivity, increasing the plasticity of the system and adding complexity to its structure. In particular, the roamer type of VT (i.e. the intercellular transfer of RNA, proteins and receptors by extracellular vesicles) will be discussed since it allowed us to introduce a new concept of 'transient changes of cell phenotype', that is the transient acquisition of new signal release capabilities and/or new recognition/decoding apparatuses.
Collapse
|
33
|
Kubrusly RC, Günter A, Sampaio L, Martins RS, Schitine CS, Trindade P, Fernandes A, Borelli-Torres R, Miya-Coreixas VS, Rego Costa AC, Freitas HR, Gardino PF, de Mello FG, Calaza KC, Reis RA. Neuro-glial cannabinoid receptors modulate signaling in the embryonic avian retina. Neurochem Int 2018; 112:27-37. [DOI: 10.1016/j.neuint.2017.10.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 10/24/2017] [Accepted: 10/30/2017] [Indexed: 10/18/2022]
|
34
|
Dai H, Jackson CR, Davis GL, Blakely RD, McMahon DG. Is dopamine transporter-mediated dopaminergic signaling in the retina a noninvasive biomarker for attention-deficit/ hyperactivity disorder? A study in a novel dopamine transporter variant Val559 transgenic mouse model. J Neurodev Disord 2017; 9:38. [PMID: 29281965 PMCID: PMC5745861 DOI: 10.1186/s11689-017-9215-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 09/14/2017] [Indexed: 12/14/2022] Open
Abstract
Background Dopamine (DA) is a critical neuromodulator in the retina. Disruption of retinal DA synthesis and signaling significantly attenuates light-adapted, electroretinogram (ERG) responses, as well as contrast sensitivity and acuity. As these measures can be detected noninvasively, they may provide opportunities to detect disease processes linked to perturbed DA signaling. Recently, we identified a rare, functional DA transporter (DAT, SLC6A3) coding substitution, Ala559Val, in subjects with attention-deficit/hyperactivity disorder (ADHD), demonstrating that DAT Val559 imparts anomalous DA efflux (ADE) with attendant physiological, pharmacological, and behavioral phenotypes. To understand the broader impact of ADE on ADHD, noninvasive measures sensitive to DAT reversal are needed. Methods Here, we explored this question through ERG-based analysis of retinal light responses, as well as HPLC measurements of retinal DA in DAT Val559 mice. Results Male mice homozygous (HOM) for the DAT Val559 variant demonstrated increased, light-adapted ERG b-wave amplitudes compared to wild type (WT) and heterozygous (HET) mice, whereas dark-adapted responses were indistinguishable across genotypes. The elevated amplitude of the photopic light responses in HOM mice could be mimicked in WT mice by applying D1 and D4 DA receptor agonists and suppressed in HOM mice by introducing D4 antagonist, supporting elevated retinal DA signaling arising from ADE. Following the challenge with amphetamine, WT exhibited an increase in light-adapted response amplitudes, while HOM did not. Total retinal DA content was similar across genotypes. Interestingly, female DAT Val559 HOM animals revealed no significant difference in photopic ERG responses when compared with WT and HET littermates. Conclusions These data reveal that noninvasive, in vivo evaluation of retinal responses to light can reveal physiological signatures of ADE, suggesting a possible approach to the segregation of neurobehavioral disorders based on the DAT-dependent control of DA signaling.
Collapse
Affiliation(s)
- Heng Dai
- Department of Biological Sciences, Vanderbilt University, Box 35-1634 Station B, Nashville, TN, 37235-1634, USA
| | - Chad R Jackson
- Department of Biological Sciences, Vanderbilt University, Box 35-1634 Station B, Nashville, TN, 37235-1634, USA.,Present address: Defense Threat Reduction Agency, 8211 Terminal Road, Lorton, VA, 22079, USA
| | - Gwynne L Davis
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Randy D Blakely
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Douglas G McMahon
- Department of Biological Sciences, Vanderbilt University, Box 35-1634 Station B, Nashville, TN, 37235-1634, USA.
| |
Collapse
|
35
|
Liu LL, Spix NJ, Zhang DQ. NMDA Receptors Contribute to Retrograde Synaptic Transmission from Ganglion Cell Photoreceptors to Dopaminergic Amacrine Cells. Front Cell Neurosci 2017; 11:279. [PMID: 28959188 PMCID: PMC5603656 DOI: 10.3389/fncel.2017.00279] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 08/29/2017] [Indexed: 11/13/2022] Open
Abstract
Recently, a line of evidence has demonstrated that the vertebrate retina possesses a novel retrograde signaling pathway. In this pathway, phototransduction is initiated by the photopigment melanopsin, which is expressed in a small population of retinal ganglion cells. These ganglion cell photoreceptors then signal to dopaminergic amacrine cells (DACs) through glutamatergic synapses, influencing visual light adaptation. We have previously demonstrated that in Mg2+-containing solution, α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate (AMPA) receptors mediate this glutamatergic transmission. Here, we demonstrate that removing extracellular Mg2+ enhances melanopsin-based DAC light responses at membrane potentials more negative than −40 mV. Melanopsin-based responses in Mg2+-free solution were profoundly suppressed by the selective N-methyl-D-aspartate (NMDA) receptor antagonist D-AP5. In addition, application of NMDA to the retina produced excitatory inward currents in DACs. These data strongly suggest that DACs express functional NMDA receptors. We further found that in the presence of Mg2+, D-AP5 reduced the peak amplitude of melanopsin-based DAC responses by ~70% when the cells were held at their resting membrane potential (−50 mV), indicating that NMDA receptors are likely to contribute to retrograde signal transmission to DACs under physiological conditions. Moreover, our data show that melanopsin-based NMDA-receptor-mediated responses in DACs are suppressed by antagonists specific to either the NR2A or NR2B receptor subtype. Immunohistochemical results show that NR2A and NR2B subunits are expressed on DAC somata and processes. These results suggest that DACs express functional NMDA receptors containing both NR2A and NR2B subunits. Collectively, our data reveal that, along with AMPA receptors, NR2A- and NR2B-containing NMDA receptors mediate retrograde signal transmission from ganglion cell photoreceptors to DACs.
Collapse
Affiliation(s)
- Lei-Lei Liu
- Eye Research Institute, Oakland UniversityRochester, MI, United States
| | - Nathan J Spix
- Eye Research Institute, Oakland UniversityRochester, MI, United States
| | - Dao-Qi Zhang
- Eye Research Institute, Oakland UniversityRochester, MI, United States
| |
Collapse
|
36
|
Goodings L, He J, Wood AJ, Harris WA, Currie PD, Jusuf PR. In vivo expression of Nurr1/Nr4a2a in developing retinal amacrine subtypes in zebrafish Tg(nr4a2a:eGFP) transgenics. J Comp Neurol 2017; 525:1962-1979. [PMID: 28177524 DOI: 10.1002/cne.24185] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 01/29/2017] [Accepted: 01/30/2017] [Indexed: 12/12/2022]
Abstract
The Nuclear receptor subfamily 4 group A member 2 (Nr4a2) is crucial for the formation or maintenance of dopaminergic neurons in the central nervous system including the retina, where dopaminergic amacrine cells contribute to visual function. Little is known about which cells express Nr4a2 at which developmental stage. Furthermore, whether Nr4a2 functions in combination with other genes is poorly understood. Thus, we generated a novel transgenic to visualize Nr4a2 expression in vivo during zebrafish retinogenesis. A 4.1 kb fragment of the nr4a2a promoter was used to drive green fluorescent protein expression in this Tg(nr4a2a:eGFP) line. In situ hybridization showed that transgene expression follows endogenous RNA expression at a cellular level. Temporal expression and lineages were quantified using in vivo time-lapse imaging in embryos. Nr4a2 expressing retinal subtypes were characterized immunohistochemically. Nr4a2a:eGFP labeled multiple neuron subtypes including 24.5% of all amacrine interneurons. Nr4a2a:eGFP labels all tyrosine hydroxylase labeled dopaminergic amacrine cells, and other nondopaminergic GABAergic amacrine populations. Nr4a2a:eGFP is confined to a specific progenitor lineage identified by sequential expression of the bhlh transcription factor Atonal7 (Atoh7) and Pancreas transcription factor 1a (Ptf1a), and labels postmitotic postmigratory amacrine cells. Thus, developmental Nr4a2a expression indicates a role during late differentiation of specific amacrine interneurons. Tg(nr4a2a:eGFP) is an early marker of distinct neurons including dopaminergic amacrine cells. It can be utilized to assess consequences of gene manipulations and understand whether Nr4a2 only carries out its role in the presence of specific coexpressed genes. This will allow Nr4a2 use to be refined for regenerative approaches.
Collapse
Affiliation(s)
- Liana Goodings
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Jie He
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Alasdair J Wood
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - William A Harris
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Peter D Currie
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Patricia R Jusuf
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,School of Biosciences, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
37
|
High-resolution imaging of cellular dopamine efflux using a fluorescent nanosensor array. Proc Natl Acad Sci U S A 2017; 114:1789-1794. [PMID: 28179565 DOI: 10.1073/pnas.1613541114] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Intercellular communication via chemical signaling proceeds with both spatial and temporal components, but analytical tools, such as microfabricated electrodes, have been limited to just a few probes per cell. In this work, we use a nonphotobleaching fluorescent nanosensor array based on single-walled carbon nanotubes (SWCNTs) rendered selective to dopamine to study its release from PC12 neuroprogenitor cells at a resolution exceeding 20,000 sensors per cell. This allows the spatial and temporal dynamics of dopamine release, following K+ stimulation, to be measured at exceedingly high resolution. We observe localized, unlabeled release sites of dopamine spanning 100 ms to seconds that correlate with protrusions but not predominately the positive curvature associated with the tips of cellular protrusions as intuitively expected. The results illustrate how directionality of chemical signaling is shaped by membrane morphology, and highlight the advantages of nanosensor arrays that can provide high spatial and temporal resolution of chemical signaling.
Collapse
|
38
|
Rodríguez-Sosa L, Calderón-Rosete G, Ortega-Cambranis A, De-Miguel FF. Octopamine cyclic release and its modulation of visual sensitivity in crayfish. Comp Biochem Physiol A Mol Integr Physiol 2016; 203:83-90. [PMID: 27593450 DOI: 10.1016/j.cbpa.2016.08.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 08/30/2016] [Accepted: 08/30/2016] [Indexed: 11/27/2022]
Abstract
The biogenic amine octopamine (OA) modulates invertebrate behavior by changing neuronal responses from sensory inputs to motor outputs. However, the OA modulation of visual sensitivity and its possible coupling to diurnal cycles remains unexplored. Here we studied the diurnal variations in the OA levels in the hemolymph of the crayfish Procambarus clarkii, its release from the structures in the eyestalk and its modulation of the retinal light sensitivity. The hemolymph concentration of OA and its amino acid precursor tyrosine was measured by high-resolution liquid chromatography; OA varied along the 24-hcycle. The peak value appeared about 2h before the light offset which preceded the peak locomotor activity. OA was found in every structure of the eyestalk but displayed higher levels in the retina-lamina ganglionaris. Moreover, OA was released from isolated eyestalks at a rate of 92nmol/eyestalk/min and a calcium-dependent release was evoked by incubation in a high potassium solution. OA injected into dark-adapted crayfish or applied to the isolated retina at concentrations of 1, 10 and 100μM produced a proportionally increasing reduction in the amplitude of the photoreceptor light responses. These OA concentrations did not affect the position of the visual accessory pigments. Our results suggest that OA release in the crayfish eyestalk is coupled to the 24-hcycle to regulate the diurnal reduction of the photoreceptor sensitivity and to favor the expression of exploratory locomotion during the dark phase of the circadian cycle.
Collapse
Affiliation(s)
- Leonardo Rodríguez-Sosa
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad Universitaria, C.P. 04510 Ciudad de México, México.
| | - Gabina Calderón-Rosete
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad Universitaria, C.P. 04510 Ciudad de México, México
| | - Aída Ortega-Cambranis
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, C.P. 72570 Puebla, Pue., México
| | - Francisco F De-Miguel
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, Ciudad Universitaria, C.P. 04510 Ciudad de México, México
| |
Collapse
|
39
|
Caravaggio F, Kegeles LS, Wilson AA, Remington G, Borlido C, Mamo DC, Graff-Guerrero A. Estimating the effect of endogenous dopamine on baseline [(11) C]-(+)-PHNO binding in the human brain. Synapse 2016; 70:453-60. [PMID: 27341789 DOI: 10.1002/syn.21920] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 06/03/2016] [Accepted: 06/20/2016] [Indexed: 11/10/2022]
Abstract
Endogenous dopamine (DA) levels at dopamine D2/3 receptors (D2/3 R) have been quantified in the living human brain using the agonist radiotracer [(11) C]-(+)-PHNO. As an agonist radiotracer, [(11) C]-(+)-PHNO is more sensitive to endogenous DA levels than antagonist radiotracers. We sought to determine the proportion of the variance in baseline [(11) C]-(+)-PHNO binding to D2/3 Rs which can be accounted for by variation in endogenous DA levels. This was done by computing the Pearson's coefficient for the correlation between baseline binding potential (BPND ) and the change in BPND after acute DA depletion, using previously published data. All correlations were inverse, and the proportion of the variance in baseline [(11) C]-(+)-PHNO BPND that can be accounted for by variation in endogenous DA levels across the striatal subregions ranged from 42-59%. These results indicate that lower baseline values of [(11) C]-(+)-PHNO BPND reflect greater stimulation by endogenous DA. To further validate this interpretation, we sought to examine whether these data could be used to estimate the dissociation constant (Kd) of DA at D2/3 R. In line with previous in vitro work, we estimated the in vivo Kd of DA to be around 20 nM. In summary, the agonist radiotracer [(11) C]-(+)-PHNO can detect the impact of endogenous DA levels at D2/3 R in the living human brain from a single baseline scan, and may be more sensitive to this impact than other commonly employed radiotracers.
Collapse
Affiliation(s)
- Fernando Caravaggio
- Centre for Addiction and Mental Health, Research Imaging Centre, Toronto, Ontario, M5T 1R8, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Lawrence S Kegeles
- Department of Psychiatry and Radiology, Columbia University College of Physicians and Surgeons, New York State Psychiatric Institute, New York
| | - Alan A Wilson
- Centre for Addiction and Mental Health, Research Imaging Centre, Toronto, Ontario, M5T 1R8, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, M5S 1A8, Canada.,Department of Psychiatry, University of Toronto, Ontario, M5T 1R8, Canada
| | - Gary Remington
- Centre for Addiction and Mental Health, Research Imaging Centre, Toronto, Ontario, M5T 1R8, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, M5S 1A8, Canada.,Department of Psychiatry, University of Toronto, Ontario, M5T 1R8, Canada
| | - Carol Borlido
- Centre for Addiction and Mental Health, Research Imaging Centre, Toronto, Ontario, M5T 1R8, Canada
| | - David C Mamo
- Department of Psychiatry, Faculties of Medicine and Health Science, University of Malta, Msida, Malta
| | - Ariel Graff-Guerrero
- Centre for Addiction and Mental Health, Research Imaging Centre, Toronto, Ontario, M5T 1R8, Canada. .,Institute of Medical Science, University of Toronto, Toronto, Ontario, M5S 1A8, Canada. .,Department of Psychiatry, University of Toronto, Ontario, M5T 1R8, Canada.
| |
Collapse
|
40
|
De-Miguel FF, Nicholls JG. Release of chemical transmitters from cell bodies and dendrites of nerve cells. Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2014.0181. [PMID: 26009760 DOI: 10.1098/rstb.2014.0181] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Papers in this issue concern extrasynaptic transmission, namely release of signalling molecules by exocytosis or diffusion from neuronal cell bodies, dendrites, axons and glia. Problems discussed concern the molecules, their secretion and importance for normal function and disease. Molecules secreted extrasynaptically include transmitters, peptides, hormones and nitric oxide. For extrasynaptic secretion, trains of action potentials are required, and the time course of release is slower than at synapses. Questions arise concerning the mechanism of extrasynaptic secretion: how does it differ from the release observed at synaptic terminals and gland cells? What kinds of vesicles take part? Is release accomplished through calcium entry, SNAP and SNARE proteins? A clear difference is in the role of molecules released synaptically and extrasynaptically. After extrasynaptic release, molecules reach distant as well as nearby cells, and thereby produce long-lasting changes over large volumes of brain. Such changes can affect circuits for motor performance and mood states. An example with clinical relevance is dyskinesia of patients treated with l-DOPA for Parkinson's disease. Extrasynaptically released transmitters also evoke responses in glial cells, which in turn release molecules that cause local vasodilatation and enhanced circulation in regions of the brain that are active.
Collapse
Affiliation(s)
- Francisco F De-Miguel
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, Distrito Federal, Mexico
| | - John G Nicholls
- Scuola Internazionale Superiore di Studi Avanzati, SISSA, Trieste, Italy
| |
Collapse
|
41
|
Euler T, Schubert T. Multiple Independent Oscillatory Networks in the Degenerating Retina. Front Cell Neurosci 2015; 9:444. [PMID: 26617491 PMCID: PMC4637421 DOI: 10.3389/fncel.2015.00444] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 10/26/2015] [Indexed: 01/09/2023] Open
Abstract
During neuronal degenerative diseases, microcircuits undergo severe structural alterations, leading to remodeling of synaptic connectivity. This can be particularly well observed in the retina, where photoreceptor degeneration triggers rewiring of connections in the retina’s first synaptic layer (e.g., Strettoi et al., 2003; Haq et al., 2014), while the synaptic organization of inner retinal circuits appears to be little affected (O’Brien et al., 2014; Figures 1A,B). Remodeling of (outer) retinal circuits and diminishing light-driven activity due to the loss of functional photoreceptors lead to spontaneous activity that can be observed at different retinal levels (Figure 1C), including the retinal ganglion cells, which display rhythmic spiking activity in the degenerative retina (Margolis et al., 2008; Stasheff, 2008; Menzler and Zeck, 2011; Stasheff et al., 2011). Two networks have been suggested to drive the oscillatory activity in the degenerating retina: a network of remnant cone photoreceptors, rod bipolar cells (RBCs) and horizontal cells in the outer retina (Haq et al., 2014), and the AII amacrine cell-cone bipolar cell network in the inner retina (Borowska et al., 2011). Notably, spontaneous rhythmic activity in the inner retinal network can be triggered in the absence of synaptic remodeling in the outer retina, for example, in the healthy retina after photo-bleaching (Menzler et al., 2014). In addition, the two networks show remarkable differences in their dominant oscillation frequency range as well as in the types and numbers of involved cells (Menzler and Zeck, 2011; Haq et al., 2014). Taken together this suggests that the two networks are self-sustained and can be active independently from each other. However, it is not known if and how they modulate each other. In this mini review, we will discuss: (i) commonalities and differences between these two oscillatory networks as well as possible interaction pathways; (ii) how multiple self-sustained networks may hamper visual restoration strategies employing, for example, microelectronic implants, optogenetics or stem cells, and briefly; and (iii) how the finding of diverse (independent) networks in the degenerative retina may relate to other parts of the neurodegenerative central nervous system.
Collapse
Affiliation(s)
- Thomas Euler
- Werner Reichardt Centre for Integrative Neuroscience (CIN)/Institute for Ophathalmic Research, University of Tübingen Tübingen, Germany ; Bernstein Centre for Computational Neuroscience, University of Tübingen Tübingen, Germany
| | - Timm Schubert
- Werner Reichardt Centre for Integrative Neuroscience (CIN)/Institute for Ophathalmic Research, University of Tübingen Tübingen, Germany
| |
Collapse
|