151
|
Shao YQ, Fan L, Wu WY, Zhu YJ, Xu HT. A developmental switch between electrical and neuropeptide communication in the ventromedial hypothalamus. Curr Biol 2022; 32:3137-3145.e3. [PMID: 35659861 DOI: 10.1016/j.cub.2022.05.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 04/15/2022] [Accepted: 05/11/2022] [Indexed: 12/29/2022]
Abstract
Dissecting neural connectivity patterns within local brain regions is an essential step to understanding the function of the brain.1 Neural microcircuits in brain regions, such as the neocortex and the hippocampus, have been extensively studied.2 By contrast, the microcircuit in the hypothalamus remains largely uncharacterized. The hypothalamus is crucial for animals' survival and reproduction.3 Knowledge of how different hypothalamic nuclei coordinate with each other and outside brain regions for hypothalamus-related functions has been significantly advanced.4-9 Although there are limited studies on the neural microcircuit in the lateral hypothalamus (LHA)10,11 and the suprachiasmatic nucleus (SCN),12,13 the patterns of neural microcircuits in most of the given hypothalamic nuclei remain largely unknown. This study applied combinatory approaches to address the local neural circuit pattern in the ventromedial hypothalamus (VMH) and other hypothalamic nuclei. We discovered a unique neural circuit design in the VMH. Neurons in the VMH were electrically coupled at the early postnatal stage like ones in the neocortex.14 However, unlike neocortical neurons,14,15 they developed very few chemical synapses after the disappearance of electrical synapses. Instead, VMH neurons communicated with neuropeptides. The similar scarceness of synaptic connectivity found in other hypothalamic nuclei further indicated that the lack of synaptic connections is a unique feature for local neural circuits in most adult hypothalamic nuclei. Thus, our findings provide a solid synaptic basis at the cellular level to understand hypothalamic functions better.
Collapse
Affiliation(s)
- Yin-Qi Shao
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liu Fan
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wen-Yan Wu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yi-Jun Zhu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hua-Tai Xu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai 201210, China.
| |
Collapse
|
152
|
Guidolin D, Tortorella C, Marcoli M, Maura G, Agnati LF. Intercellular Communication in the Central Nervous System as Deduced by Chemical Neuroanatomy and Quantitative Analysis of Images: Impact on Neuropharmacology. Int J Mol Sci 2022; 23:5805. [PMID: 35628615 PMCID: PMC9145073 DOI: 10.3390/ijms23105805] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 01/25/2023] Open
Abstract
In the last decades, new evidence on brain structure and function has been acquired by morphological investigations based on synergic interactions between biochemical anatomy approaches, new techniques in microscopy and brain imaging, and quantitative analysis of the obtained images. This effort produced an expanded view on brain architecture, illustrating the central nervous system as a huge network of cells and regions in which intercellular communication processes, involving not only neurons but also other cell populations, virtually determine all aspects of the integrative function performed by the system. The main features of these processes are described. They include the two basic modes of intercellular communication identified (i.e., wiring and volume transmission) and mechanisms modulating the intercellular signaling, such as cotransmission and allosteric receptor-receptor interactions. These features may also open new possibilities for the development of novel pharmacological approaches to address central nervous system diseases. This aspect, with a potential major impact on molecular medicine, will be also briefly discussed.
Collapse
Affiliation(s)
- Diego Guidolin
- Department of Neuroscience, Section of Anatomy, University of Padova, 35121 Padova, Italy;
| | - Cinzia Tortorella
- Department of Neuroscience, Section of Anatomy, University of Padova, 35121 Padova, Italy;
| | - Manuela Marcoli
- Department of Pharmacy, Center of Excellence for Biomedical Research, University of Genova, 16126 Genova, Italy; (M.M.); (G.M.)
| | - Guido Maura
- Department of Pharmacy, Center of Excellence for Biomedical Research, University of Genova, 16126 Genova, Italy; (M.M.); (G.M.)
| | - Luigi F. Agnati
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy;
| |
Collapse
|
153
|
Zup SL, Park JH, Dominguez JM. Editorial: Intersection of Hormones and Neuropeptides in the Brain. Front Behav Neurosci 2022; 16:886591. [PMID: 35530729 PMCID: PMC9074826 DOI: 10.3389/fnbeh.2022.886591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 03/28/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Susan L Zup
- Developmental and Brain Sciences Program (DBS), Department of Psychology, University of Massachusetts Boston, Boston, MA, United States
| | - Jin Ho Park
- Developmental and Brain Sciences Program (DBS), Department of Psychology, University of Massachusetts Boston, Boston, MA, United States
| | - Juan M Dominguez
- Departments of Psychology, The University of Texas at Austin, Austin, TX, United States.,Department of Pharmacology & Toxicology, The University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
154
|
Wang M, Duong TL, Rea BJ, Waite JS, Huebner MW, Flinn HC, Russo AF, Sowers LP. CGRP Administration Into the Cerebellum Evokes Light Aversion, Tactile Hypersensitivity, and Nociceptive Squint in Mice. FRONTIERS IN PAIN RESEARCH 2022; 3:861598. [PMID: 35547239 PMCID: PMC9082264 DOI: 10.3389/fpain.2022.861598] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/17/2022] [Indexed: 12/16/2022] Open
Abstract
The neuropeptide calcitonin gene-related peptide (CGRP) is a major player in migraine pathophysiology. Previous preclinical studies demonstrated that intracerebroventricular administration of CGRP caused migraine-like behaviors in mice, but the sites of action in the brain remain unidentified. The cerebellum has the most CGRP binding sites in the central nervous system and is increasingly recognized as both a sensory and motor integration center. The objective of this study was to test whether the cerebellum, particularly the medial cerebellar nuclei (MN), might be a site of CGRP action. In this study, CGRP was directly injected into the right MN of C57BL/6J mice via a cannula. A battery of tests was done to assess preclinical behaviors that are surrogates of migraine-like symptoms. CGRP caused light aversion measured as decreased time in the light zone even with dim light. The mice also spent more time resting in the dark zone, but not the light, along with decreased rearing and transitions between zones. These behaviors were similar for both sexes. Moreover, significant responses to CGRP were seen in the open field assay, von Frey test, and automated squint assay, indicating anxiety, tactile hypersensitivity, and spontaneous pain, respectively. Interestingly, CGRP injection caused significant anxiety and spontaneous pain responses only in female mice, and a more robust tactile hypersensitivity in female mice. No detectable effect of CGRP on gait was observed in either sex. These results suggest that CGRP injection in the MN causes light aversion accompanied by increased anxiety, tactile hypersensitivity, and spontaneous pain. A caveat is that we cannot exclude contributions from other cerebellar regions in addition to the MN due to diffusion of the injected peptide. These results reveal the cerebellum as a new site of CGRP actions that may contribute to migraine-like hypersensitivity.
Collapse
Affiliation(s)
- Mengya Wang
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, United States
| | - Thomas L. Duong
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, United States
| | - Brandon J. Rea
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, United States
- Center for the Prevention and Treatment of Visual Loss, Veterans Administration Health Center, Iowa City, IA, United States
| | - Jayme S. Waite
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, United States
| | - Michael W. Huebner
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, United States
| | - Harold C. Flinn
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, United States
| | - Andrew F. Russo
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, United States
- Center for the Prevention and Treatment of Visual Loss, Veterans Administration Health Center, Iowa City, IA, United States
- Department of Neurology, University of Iowa, Iowa City, IA, United States
| | - Levi P. Sowers
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, United States
- Center for the Prevention and Treatment of Visual Loss, Veterans Administration Health Center, Iowa City, IA, United States
- *Correspondence: Levi P. Sowers
| |
Collapse
|
155
|
Urrutia-Piñones J, Morales-Moraga C, Sanguinetti-González N, Escobar AP, Chiu CQ. Long-Range GABAergic Projections of Cortical Origin in Brain Function. Front Syst Neurosci 2022; 16:841869. [PMID: 35392440 PMCID: PMC8981584 DOI: 10.3389/fnsys.2022.841869] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/10/2022] [Indexed: 12/12/2022] Open
Abstract
The study of long-range GABAergic projections has traditionally been focused on those with subcortical origin. In the last few years, cortical GABAergic neurons have been shown to not only mediate local inhibition, but also extend long-range axons to remote cortical and subcortical areas. In this review, we delineate the different types of long-range GABAergic neurons (LRGNs) that have been reported to arise from the hippocampus and neocortex, paying attention to the anatomical and functional circuits they form to understand their role in behavior. Although cortical LRGNs are similar to their interneuron and subcortical counterparts, they comprise distinct populations that show specific patterns of cortico-cortical and cortico-fugal connectivity. Functionally, cortical LRGNs likely induce timed disinhibition in target regions to synchronize network activity. Thus, LRGNs are emerging as a new element of cortical output, acting in concert with long-range excitatory projections to shape brain function in health and disease.
Collapse
Affiliation(s)
- Jocelyn Urrutia-Piñones
- Ph.D. Program in Neuroscience, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Facultad de Ciencias, Instituto de Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Camila Morales-Moraga
- Facultad de Ciencias, Instituto de Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Nicole Sanguinetti-González
- Ph.D. Program in Neuroscience, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Facultad de Ciencias, Instituto de Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Angelica P. Escobar
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Centro de Neurobiología y Fisiopatología Integrativa, Universidad de Valparaíso, Valparaíso, Chile
| | - Chiayu Q. Chiu
- Facultad de Ciencias, Instituto de Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
156
|
CRISPR/Cas9-Based Mutagenesis of Histone H3.1 in Spinal Dynorphinergic Neurons Attenuates Thermal Sensitivity in Mice. Int J Mol Sci 2022; 23:ijms23063178. [PMID: 35328599 PMCID: PMC8955318 DOI: 10.3390/ijms23063178] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 02/01/2023] Open
Abstract
Burn injury is a trauma resulting in tissue degradation and severe pain, which is processed first by neuronal circuits in the spinal dorsal horn. We have recently shown that in mice, excitatory dynorphinergic (Pdyn) neurons play a pivotal role in the response to burn-injury-associated tissue damage via histone H3.1 phosphorylation-dependent signaling. As Pdyn neurons were mostly associated with mechanical allodynia, their involvement in thermonociception had to be further elucidated. Using a custom-made AAV9_mutH3.1 virus combined with the CRISPR/cas9 system, here we provide evidence that blocking histone H3.1 phosphorylation at position serine 10 (S10) in spinal Pdyn neurons significantly increases the thermal nociceptive threshold in mice. In contrast, neither mechanosensation nor acute chemonociception was affected by the transgenic manipulation of histone H3.1. These results suggest that blocking rapid epigenetic tagging of S10H3 in spinal Pdyn neurons alters acute thermosensation and thus explains the involvement of Pdyn cells in the immediate response to burn-injury-associated tissue damage.
Collapse
|
157
|
Wu G, Heck I, Zhang N, Phaup G, Zhang X, Wu Y, Stalla DE, Weng Z, Sun H, Li H, Zhang Z, Ding S, Li DP, Zhang Y. Wireless, battery-free push-pull microsystem for membrane-free neurochemical sampling in freely moving animals. SCIENCE ADVANCES 2022; 8:eabn2277. [PMID: 35196090 PMCID: PMC8865804 DOI: 10.1126/sciadv.abn2277] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 12/29/2021] [Indexed: 06/12/2023]
Abstract
Extensive studies in both animals and humans have demonstrated that high molecular weight neurochemicals, such as neuropeptides and other polypeptide neurochemicals, play critical roles in various neurological disorders. Despite many attempts, existing methods are constrained by detecting neuropeptide release in small animal models during behavior tasks, which leaves the molecular mechanisms underlying many neurological and psychological disorders unresolved. Here, we report a wireless, programmable push-pull microsystem for membrane-free neurochemical sampling with cellular spatial resolution in freely moving animals. In vitro studies demonstrate the sampling of various neurochemicals with high recovery (>80%). Open-field tests reveal that the device implantation does not affect the natural behavior of mice. The probe successfully captures the pharmacologically evoked release of neuropeptide Y in freely moving mice. This wireless push-pull microsystem creates opportunities for neuroscientists to understand where, when, and how the release of neuropeptides modulates diverse behavioral outputs of the brain.
Collapse
Affiliation(s)
- Guangfu Wu
- Department of Biomedical Engineering and the Institute of Materials Science, University of Connecticut, Storrs, CT 06269, USA
| | - Ian Heck
- Department of Biomedical, Biological, and Chemical Engineering, University of Missouri, Columbia, MO 65211, USA
| | - Nannan Zhang
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA
| | - Glenn Phaup
- Center for Precision Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Xincheng Zhang
- Department of Biomedical Engineering and the Institute of Materials Science, University of Connecticut, Storrs, CT 06269, USA
| | - Yixin Wu
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL 60208, USA
| | - David E. Stalla
- Electron Microscopy Core, University of Missouri, Columbia, MO 65211, USA
| | - Zhengyan Weng
- Department of Biomedical Engineering and the Institute of Materials Science, University of Connecticut, Storrs, CT 06269, USA
| | - He Sun
- Department of Biomedical Engineering and the Institute of Materials Science, University of Connecticut, Storrs, CT 06269, USA
| | - Huijie Li
- Department of Biomedical Engineering and the Institute of Materials Science, University of Connecticut, Storrs, CT 06269, USA
| | - Zhe Zhang
- Department of Biomedical, Biological, and Chemical Engineering, University of Missouri, Columbia, MO 65211, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA
| | - Shinghua Ding
- Department of Biomedical, Biological, and Chemical Engineering, University of Missouri, Columbia, MO 65211, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA
| | - De-Pei Li
- Center for Precision Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Yi Zhang
- Department of Biomedical Engineering and the Institute of Materials Science, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
158
|
Huang M, Xu L, Liu J, Huang P, Tan Y, Chen S. Cell–Cell Communication Alterations via Intercellular Signaling Pathways in Substantia Nigra of Parkinson’s Disease. Front Aging Neurosci 2022; 14:828457. [PMID: 35283752 PMCID: PMC8914319 DOI: 10.3389/fnagi.2022.828457] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative movement disorder characterized with dopaminergic neuron (DaN) loss within the substantia nigra (SN). Despite bulk studies focusing on intracellular mechanisms of PD inside DaNs, few studies have explored the pathogeneses outside DaNs, or between DaNs and other cells. Here, we set out to probe the implication of intercellular communication involving DaNs in the pathogeneses of PD at a systemic level with bioinformatics methods. We harvested three online published single-cell/single-nucleus transcriptomic sequencing (sc/snRNA-seq) datasets of human SN (GSE126838, GSE140231, and GSE157783) from the Gene Expression Omnibus (GEO) database, and integrated them with one of the latest integration algorithms called Harmony. We then applied CellChat, the latest cell–cell communication analytic algorithm, to our integrated dataset. We first found that the overall communication quantity was decreased while the overall communication strength was enhanced in PD sample compared with control sample. We then focused on the intercellular communication where DaNs are involved, and found that the communications between DaNs and other cell types via certain signaling pathways were selectively altered in PD, including some growth factors, neurotrophic factors, chemokines, etc. pathways. Our bioinformatics analysis showed that the alteration in intercellular communications involving DaNs might be a previously underestimated aspect of PD pathogeneses with novel translational potential.
Collapse
Affiliation(s)
- Maoxin Huang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liang Xu
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jin Liu
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pei Huang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuyan Tan
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yuyan Tan,
| | - Shengdi Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Lab for Translational Research of Neurodegenerative Diseases, Shanghai Institute for Advanced Immunochemical Studies, Shanghai Tech University, Shanghai, China
- Shengdi Chen,
| |
Collapse
|
159
|
Somatostatin and Somatostatin-Containing Interneurons—From Plasticity to Pathology. Biomolecules 2022; 12:biom12020312. [PMID: 35204812 PMCID: PMC8869243 DOI: 10.3390/biom12020312] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/27/2022] [Accepted: 02/11/2022] [Indexed: 01/27/2023] Open
Abstract
Despite the obvious differences in the pathophysiology of distinct neuropsychiatric diseases or neurodegenerative disorders, some of them share some general but pivotal mechanisms, one of which is the disruption of excitation/inhibition balance. Such an imbalance can be generated by changes in the inhibitory system, very often mediated by somatostatin-containing interneurons (SOM-INs). In physiology, this group of inhibitory interneurons, as well as somatostatin itself, profoundly shapes the brain activity, thus influencing the behavior and plasticity; however, the changes in the number, density and activity of SOM-INs or levels of somatostatin are found throughout many neuropsychiatric and neurological conditions, both in patients and animal models. Here, we (1) briefly describe the brain somatostatinergic system, characterizing the neuropeptide somatostatin itself, its receptors and functions, as well the physiology and circuitry of SOM-INs; and (2) summarize the effects of the activity of somatostatin and SOM-INs in both physiological brain processes and pathological brain conditions, focusing primarily on learning-induced plasticity and encompassing selected neuropsychological and neurodegenerative disorders, respectively. The presented data indicate the somatostatinergic-system-mediated inhibition as a substantial factor in the mechanisms of neuroplasticity, often disrupted in a plethora of brain pathologies.
Collapse
|
160
|
Tear film and ocular surface neuropeptides: Characteristics, synthesis, signaling and implications for ocular surface and systemic diseases. Exp Eye Res 2022; 218:108973. [PMID: 35149082 DOI: 10.1016/j.exer.2022.108973] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 12/14/2021] [Accepted: 02/01/2022] [Indexed: 01/13/2023]
Abstract
Ocular surface neuropeptides are vital molecules primarily involved in maintaining ocular surface integrity and homeostasis. They also serve as communication channels between the nervous system and the immune system, maintaining the homeostasis of the ocular surface. Tear film and ocular surface neuropeptides have a role in disease often due to abnormalities in their synthesis (either high or low production), signaling through defective receptors, or both. This creates imbalances in otherwise normal physiological processes. They have been observed to be altered in many ocular surface and systemic diseases including dry eye disease, ocular allergy, keratoconus, LASIK-induced dry eye, pterygium, neurotrophic keratitis, corneal graft rejection, microbial keratitis, headaches and diabetes. This review examines the characteristics of neuropeptides, their synthesis and their signaling through G-protein coupled receptors. The review also explores the types of neuropeptides within the tears and ocular surface, and how they change in ocular and systemic diseases.
Collapse
|
161
|
Xia X, Wang Y, Qin Y, Zhao S, Zheng JC. Exosome: A novel neurotransmission modulator or non-canonical neurotransmitter? Ageing Res Rev 2022; 74:101558. [PMID: 34990846 DOI: 10.1016/j.arr.2021.101558] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 08/13/2021] [Accepted: 12/30/2021] [Indexed: 02/08/2023]
Abstract
Neurotransmission is the electrical impulse-triggered propagation of signals between neurons or between neurons and other cell types such as skeletal muscle cells. Recent studies point out the involvement of exosomes, a type of small bilipid layer-enclosed extracellular vesicles, in regulating neurotransmission. Through horizontally transferring proteins, lipids, and nucleic acids, exosomes can modulate synaptic activities rapidly by controlling neurotransmitter release or progressively by regulating neural plasticity including synapse formation, neurite growth & removal, and axon guidance & elongation. In this review, we summarize the similarities and differences between exosomes and synaptic vesicles in their biogenesis, contents, and release. We also highlight the recent progress made in demonstrating the biological roles of exosome in regulating neurotransmission, and propose a modified model of neurotransmission, in which exosomes act as novel neurotransmitters. Lastly, we provide a comprehensive discussion of the enlightenment of the current knowledge on neurotransmission to the future directions of exosome research.
Collapse
|
162
|
Ferdos S, Brockhaus J, Missler M, Rohlmann A. Deletion of β-Neurexins in Mice Alters the Distribution of Dense-Core Vesicles in Presynapses of Hippocampal and Cerebellar Neurons. Front Neuroanat 2022; 15:757017. [PMID: 35173587 PMCID: PMC8841415 DOI: 10.3389/fnana.2021.757017] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 12/16/2021] [Indexed: 11/17/2022] Open
Abstract
Communication between neurons through synapses includes the release of neurotransmitter-containing synaptic vesicles (SVs) and of neuromodulator-containing dense-core vesicles (DCVs). Neurexins (Nrxns), a polymorphic family of cell surface molecules encoded by three genes in vertebrates (Nrxn1–3), have been proposed as essential presynaptic organizers and as candidates for cell type-specific or even synapse-specific regulation of synaptic vesicle exocytosis. However, it remains unknown whether Nrxns also regulate DCVs. Here, we report that at least β-neurexins (β-Nrxns), an extracellularly smaller Nrxn variant, are involved in the distribution of presynaptic DCVs. We found that conditional deletion of all three β-Nrxn isoforms in mice by lentivirus-mediated Cre recombinase expression in primary hippocampal neurons reduces the number of ultrastructurally identified DCVs in presynaptic boutons. Consistently, colabeling against marker proteins revealed a diminished population of chromogranin A- (ChrgA-) positive DCVs in synapses and axons of β-Nrxn-deficient neurons. Moreover, we validated the impaired DCV distribution in cerebellar brain tissue from constitutive β-Nrxn knockout (β-TKO) mice, where DCVs are normally abundant and β-Nrxn isoforms are prominently expressed. Finally, we observed that the ultrastructure and marker proteins of the Golgi apparatus, responsible for packaging neuropeptides into DCVs, seem unchanged. In conclusion, based on the validation from the two deletion strategies in conditional and constitutive KO mice, two neuronal populations from the hippocampus and cerebellum, and two experimental protocols in cultured neurons and in the brain tissue, this study presented morphological evidence that the number of DCVs at synapses is altered in the absence of β-Nrxns. Our results therefore point to an unexpected contribution of β-Nrxns to the organization of neuropeptide and neuromodulator function, in addition to their more established role in synaptic vesicle release.
Collapse
|
163
|
The Role of Neuropeptide-Stimulated cAMP-EPACs Signalling in Cancer Cells. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27010311. [PMID: 35011543 PMCID: PMC8746471 DOI: 10.3390/molecules27010311] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 02/06/2023]
Abstract
Neuropeptides are autocrine and paracrine signalling factors and mainly bind to G protein-coupled receptors (GPCRs) to trigger intracellular secondary messenger release including adenosine 3′, 5′-cyclic monophosphate (cAMP), thus modulating cancer progress in different kind of tumours. As one of the downstream effectors of cAMP, exchange proteins directly activated by cAMP (EPACs) play dual roles in cancer proliferation and metastasis. More evidence about the relationship between neuropeptides and EPAC pathways have been proposed for their potential role in cancer development; hence, this review focuses on the role of neuropeptide/GPCR system modulation of cAMP/EPACs pathways in cancers. The correlated downstream pathways between neuropeptides and EPACs in cancer cell proliferation, migration, and metastasis is discussed to glimmer the direction of future research.
Collapse
|
164
|
Hypothalamic melanin-concentrating hormone regulates hippocampus-dorsolateral septum activity. Nat Neurosci 2022; 25:61-71. [PMID: 34980924 PMCID: PMC8741735 DOI: 10.1038/s41593-021-00984-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 11/10/2021] [Indexed: 12/15/2022]
Abstract
Hypothalamic melanin-concentrating hormone (MCH) polypeptide contributes to regulating energy homeostasis, sleep, and memory, though the mechanistic bases of its effects are unknown. Here, in mice, we uncover the physiological mechanism underlying the functional role of MCH signaling in projections to the dorsolateral septum (dLS), a region involved in routing hippocampal firing rhythms and encoding spatial memory based on such rhythms. Firing activity within the dLS in response to dorsal CA3 (dCA3) excitation is limited by strong feed-forward inhibition (FFI). We find that MCH synchronizes dLS neuronal firing with its dCA3 inputs by enhancing GABA release, which subsequently reduces the FFI and augments dCA3 excitatory input strength, both via presynaptic mechanisms. At the functional level, our data reveal a role for MCH signaling in the dLS in facilitating spatial memory. These findings support a model in which peptidergic signaling within the dLS modulates dorsal hippocampal output and supports memory encoding.
Collapse
|
165
|
Han Y, Ding K. Imaging Neuropeptide Release at Drosophila Neuromuscular Junction with a Genetically Engineered Neuropeptide Release Reporter. Methods Mol Biol 2022; 2417:193-203. [PMID: 35099801 DOI: 10.1007/978-1-0716-1916-2_15] [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: 06/14/2023]
Abstract
Despite the important roles of neuropeptides in a variety of physiological processes, there still lacks a method to probe neuropeptide release events in vivo with satisfying temporal and spatial resolution. Neuropeptide Release Reporter (NPRR) was recently introduced as a novel genetically encoded indicator of neuropeptide release with a high temporal resolution and peptide specificity based on GCaMP molecule. Here we describe a method for using NPRR to image selective neuropeptide release at Drosophila neuromuscular junction in semi-dissected larvae. This method provides a quantitative analysis of activity-dependent neuropeptide release as real-time changes in fluorescence intensity of GCaMP reporter with sub-second temporal resolution and single bouton specificity.
Collapse
Affiliation(s)
- Yifu Han
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA.
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA.
| | - Keke Ding
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| |
Collapse
|
166
|
Cell-type-specific neuromodulation guides synaptic credit assignment in a spiking neural network. Proc Natl Acad Sci U S A 2021; 118:2111821118. [PMID: 34916291 PMCID: PMC8713766 DOI: 10.1073/pnas.2111821118] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2021] [Indexed: 12/27/2022] Open
Abstract
Synaptic connectivity provides the foundation for our present understanding of neuronal network function, but static connectivity cannot explain learning and memory. We propose a computational role for the diversity of cortical neuronal types and their associated cell-type–specific neuromodulators in improving the efficiency of synaptic weight adjustments for task learning in neuronal networks. Brains learn tasks via experience-driven differential adjustment of their myriad individual synaptic connections, but the mechanisms that target appropriate adjustment to particular connections remain deeply enigmatic. While Hebbian synaptic plasticity, synaptic eligibility traces, and top-down feedback signals surely contribute to solving this synaptic credit-assignment problem, alone, they appear to be insufficient. Inspired by new genetic perspectives on neuronal signaling architectures, here, we present a normative theory for synaptic learning, where we predict that neurons communicate their contribution to the learning outcome to nearby neurons via cell-type–specific local neuromodulation. Computational tests suggest that neuron-type diversity and neuron-type–specific local neuromodulation may be critical pieces of the biological credit-assignment puzzle. They also suggest algorithms for improved artificial neural network learning efficiency.
Collapse
|
167
|
Feng KL, Weng JY, Chen CC, Abubaker MB, Lin HW, Charng CC, Lo CC, de Belle JS, Tully T, Lien CC, Chiang AS. Neuropeptide F inhibits dopamine neuron interference of long-term memory consolidation in Drosophila. iScience 2021; 24:103506. [PMID: 34934925 DOI: 10.1016/j.isci.2021.103506] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/13/2021] [Accepted: 11/22/2021] [Indexed: 11/28/2022] Open
Abstract
Long-term memory (LTM) formation requires consolidation processes to overcome interfering signals that erode memory formation. Olfactory memory in Drosophila involves convergent projection neuron (PN; odor) and dopaminergic neuron (DAN; reinforcement) input to the mushroom body (MB). How post-training DAN activity in the posterior lateral protocerebrum (PPL1) continues to regulate memory consolidation remains unknown. Here we address this question using targeted transgenes in behavior and electrophysiology experiments to show that (1) persistent post-training activity of PPL1-α2α'2 and PPL1-α3 DANs interferes with aversive LTM formation; (2) neuropeptide F (NPF) signaling blocks this interference in PPL1-α2α'2 and PPL1-α3 DANs after spaced training to enable LTM formation; and (3) training-induced NPF release and neurotransmission from two upstream dorsal-anterior-lateral (DAL2) neurons are required to form LTM. Thus, NPF signals from DAL2 neurons to specific PPL1 DANs disinhibit the memory circuit, ensuring that periodic events are remembered as consolidated LTM.
Collapse
Affiliation(s)
- Kuan-Lin Feng
- Institute of Biotechnology, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Ju-Yun Weng
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Chun-Chao Chen
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan
| | | | - Hsuan-Wen Lin
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Ching-Che Charng
- Institute of Systems Neuroscience and Department of Life Science, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Chung-Chuan Lo
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan.,Institute of Systems Neuroscience and Department of Life Science, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - J Steven de Belle
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan.,Department of Psychological Sciences, University of San Diego, San Diego, CA 92110, USA.,School of Life Sciences, University of Nevada, Las Vegas, NV 89154, USA.,MnemOdyssey LLC, Escondido, CA 92027, USA
| | - Tim Tully
- Institute of Biotechnology, National Tsing Hua University, Hsinchu 30013, Taiwan.,Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Cheng-Chang Lien
- Institute of Neuroscience and Brain Research Center, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Ann-Shyn Chiang
- Institute of Biotechnology, National Tsing Hua University, Hsinchu 30013, Taiwan.,Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan.,Institute of Systems Neuroscience and Department of Life Science, National Tsing Hua University, Hsinchu 30013, Taiwan.,Kaohsiung Medical University, Kaohsiung 80708, Taiwan.,National Health Research Institutes, Zhunan 35053, Taiwan.,China Medical University, Taichung 40402, Taiwan.,Kavli Institute for Brain and Mind, University of California at San Diego, La Jolla, CA 92093-0526, USA
| |
Collapse
|
168
|
Pagella S, Deussing JM, Kopp-Scheinpflug C. Expression Patterns of the Neuropeptide Urocortin 3 and Its Receptor CRFR2 in the Mouse Central Auditory System. Front Neural Circuits 2021; 15:747472. [PMID: 34867212 PMCID: PMC8633543 DOI: 10.3389/fncir.2021.747472] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/18/2021] [Indexed: 11/16/2022] Open
Abstract
Sensory systems have to be malleable to context-dependent modulations occurring over different time scales, in order to serve their evolutionary function of informing about the external world while also eliciting survival-promoting behaviors. Stress is a major context-dependent signal that can have fast and delayed effects on sensory systems, especially on the auditory system. Urocortin 3 (UCN3) is a member of the corticotropin-releasing factor family. As a neuropeptide, UCN3 regulates synaptic activity much faster than the classic steroid hormones of the hypothalamic-pituitary-adrenal axis. Moreover, due to the lack of synaptic re-uptake mechanisms, UCN3 can have more long-lasting and far-reaching effects. To date, a modest number of studies have reported the presence of UCN3 or its receptor CRFR2 in the auditory system, particularly in the cochlea and the superior olivary complex, and have highlighted the importance of this stress neuropeptide for protecting auditory function. However, a comprehensive map of all neurons synthesizing UCN3 or CRFR2 within the auditory pathway is lacking. Here, we utilize two reporter mouse lines to elucidate the expression patterns of UCN3 and CRFR2 in the auditory system. Additional immunolabelling enables further characterization of the neurons that synthesize UCN3 or CRFR2. Surprisingly, our results indicate that within the auditory system, UCN3 is expressed predominantly in principal cells, whereas CRFR2 expression is strongest in non-principal, presumably multisensory, cell types. Based on the presence or absence of overlap between UCN3 and CRFR2 labeling, our data suggest unusual modes of neuromodulation by UCN3, involving volume transmission and autocrine signaling.
Collapse
Affiliation(s)
- Sara Pagella
- Division of Neurobiology, Faculty of Biology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Jan M Deussing
- Research Group Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Conny Kopp-Scheinpflug
- Division of Neurobiology, Faculty of Biology, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
169
|
Bhat US, Shahi N, Surendran S, Babu K. Neuropeptides and Behaviors: How Small Peptides Regulate Nervous System Function and Behavioral Outputs. Front Mol Neurosci 2021; 14:786471. [PMID: 34924955 PMCID: PMC8674661 DOI: 10.3389/fnmol.2021.786471] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/11/2021] [Indexed: 11/13/2022] Open
Abstract
One of the reasons that most multicellular animals survive and thrive is because of the adaptable and plastic nature of their nervous systems. For an organism to survive, it is essential for the animal to respond and adapt to environmental changes. This is achieved by sensing external cues and translating them into behaviors through changes in synaptic activity. The nervous system plays a crucial role in constantly evaluating environmental cues and allowing for behavioral plasticity in the organism. Multiple neurotransmitters and neuropeptides have been implicated as key players for integrating sensory information to produce the desired output. Because of its simple nervous system and well-established neuronal connectome, C. elegans acts as an excellent model to understand the mechanisms underlying behavioral plasticity. Here, we critically review how neuropeptides modulate a wide range of behaviors by allowing for changes in neuronal and synaptic signaling. This review will have a specific focus on feeding, mating, sleep, addiction, learning and locomotory behaviors in C. elegans. With a view to understand evolutionary relationships, we explore the functions and associated pathophysiology of C. elegans neuropeptides that are conserved across different phyla. Further, we discuss the mechanisms of neuropeptidergic signaling and how these signals are regulated in different behaviors. Finally, we attempt to provide insight into developing potential therapeutics for neuropeptide-related disorders.
Collapse
Affiliation(s)
- Umer Saleem Bhat
- Centre for Neuroscience, Indian Institute of Science, Bengaluru, India
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, India
| | - Navneet Shahi
- Centre for Neuroscience, Indian Institute of Science, Bengaluru, India
| | - Siju Surendran
- Centre for Neuroscience, Indian Institute of Science, Bengaluru, India
| | - Kavita Babu
- Centre for Neuroscience, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
170
|
Blumenfeld A, Durham PL, Feoktistov A, Hay DL, Russo AF, Turner I. Hypervigilance, Allostatic Load, and Migraine Prevention: Antibodies to CGRP or Receptor. Neurol Ther 2021; 10:469-497. [PMID: 34076848 PMCID: PMC8571459 DOI: 10.1007/s40120-021-00250-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 04/09/2021] [Indexed: 01/03/2023] Open
Abstract
Migraine involves brain hypersensitivity with episodic dysfunction triggered by behavioral or physiological stressors. During an acute migraine attack the trigeminal nerve is activated (peripheral sensitization). This leads to central sensitization with activation of the central pathways including the trigeminal nucleus caudalis, the trigemino-thalamic tract, and the thalamus. In episodic migraine the sensitization process ends with the individual act, but with chronic migraine central sensitization may continue interictally. Increased allostatic load, the consequence of chronic, repeated exposure to stressors, leads to central sensitization, lowering the threshold for future neuronal activation (hypervigilance). Ostensibly innocuous stressors are then sufficient to trigger an attack. Medications that reduce sensitization may help patients who are hypervigilant and help to balance allostatic load. Acute treatments and drugs for migraine prevention have traditionally been used to reduce attack duration and frequency. However, since many patients do not fully respond, an unmet treatment need remains. Calcitonin gene-related peptide (CGRP) is a vasoactive neuropeptide involved in nociception and in the sensitization of peripheral and central neurons of the trigeminovascular system, which is implicated in migraine pathophysiology. Elevated CGRP levels are associated with dysregulated signaling in the trigeminovascular system, leading to maladaptive responses to behavioral or physiological stressors. CGRP may, therefore, play a key role in the underlying pathophysiology of migraine. Increased understanding of the role of CGRP in migraine led to the development of small-molecule antagonists (gepants) and monoclonal antibodies (mAbs) that target either CGRP or the receptor (CGRP-R) to restore homeostasis, reducing the frequency, duration, and severity of attacks. In clinical trials, US Food and Drug Administration-approved anti-CGRP-R/CGRP mAbs were well tolerated and effective as preventive migraine treatments. Here, we explore the role of CGRP in migraine pathophysiology and the use of gepants or mAbs to suppress CGRP-R signaling via inhibition of the CGRP ligand or receptor.
Collapse
Affiliation(s)
- Andrew Blumenfeld
- The Headache Center of Southern California, The Neurology Center, Carlsbad, CA, USA.
| | - Paul L Durham
- Department of Biology, Center for Biomedical and Life Sciences, Missouri State University, Springfield, MO, USA
| | | | - Debbie L Hay
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand
| | - Andrew F Russo
- Departments of Molecular Physiology and Biophysics, Neurology, University of Iowa, Iowa City, IA, USA
- Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, Iowa City, IA, USA
| | - Ira Turner
- Island Neurological Associates, Plainview, NY, USA
| |
Collapse
|
171
|
Kuznetsov IA, Kuznetsov AV. Simulation of a sudden drop-off in distal dense core vesicle concentration in Drosophila type II motoneuron terminals. INTERNATIONAL JOURNAL FOR NUMERICAL METHODS IN BIOMEDICAL ENGINEERING 2021; 37:e3523. [PMID: 34418891 DOI: 10.1002/cnm.3523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 08/14/2021] [Indexed: 06/13/2023]
Abstract
Recent experimental observations have shown evidence of an unexpected sudden drop-off in the dense core vesicles (DCVs) content at the ends of certain types of axon endings. This article seeks to determine whether these observations may be explained without modifying the parameters characterizing the ability of distal en passant boutons to capture and accumulate DCVs. We developed a mathematical model that is based on the conservation of captured and transiting DCVs in boutons. The model consists of 77 ordinary differential equations and is solved using a standard Matlab solver. We hypothesize that the drop in DCV content in distal boutons is due to an insufficient supply of anterogradely moving DCVs coming from the soma. As anterogradely moving DCVs are captured (and eventually destroyed) in more proximal boutons on their way to the end of the terminal, the fluxes of anterogradely moving DCVs between the boutons become increasingly smaller, and the most distal boutons are left without DCVs. We tested this hypothesis by modifying the flux of DCVs entering the terminal and found that the number of most distal boutons left unfilled increases if the DCV flux entering the terminal is decreased. The number of anterogradely moving DCVs in the axon can be increased either by the release of a portion of captured DCVs into the anterograde component or by an increase of the anterograde DCV flux into the terminal. This increase could lead to having enough anterogradely moving DCVs such that they could reach the most distal bouton and then turn around by changing molecular motors that propel them. The model suggests that this could result in an increased concentration of resident DCVs in distal boutons beginning with bouton 2 (the most distal is bouton 1). This is because in distal boutons, DCVs have a larger chance to be captured from the transiting state as they pass the boutons moving anterogradely and then again as they pass the same boutons moving retrogradely.
Collapse
Affiliation(s)
- Ivan A Kuznetsov
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Andrey V Kuznetsov
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
172
|
Data-driven analysis of kappa opioid receptor binding in major depressive disorder measured by positron emission tomography. Transl Psychiatry 2021; 11:602. [PMID: 34839360 PMCID: PMC8627509 DOI: 10.1038/s41398-021-01729-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/26/2021] [Accepted: 11/02/2021] [Indexed: 11/08/2022] Open
Abstract
Preclinical studies have implicated kappa opioid receptors (KORs) in stress responses and depression-related behaviors, but evidence from human studies is limited. Here we present results of a secondary analysis of data acquired using positron emission tomography (PET) with the KOR radiotracer [11C]GR103545 in 10 unmedicated, currently depressed individuals with major depressive disorder (MDD; 32.6 ± 6.5 years, 5 women) and 13 healthy volunteers (34.8 ± 10 years, 6 women). Independent component analysis was performed to identify spatial patterns of coherent variance in KOR binding (tracer volume of distribution, VT) across all subjects. Expression of each component was compared between groups and relationships to symptoms were explored using the 17-item Hamilton Depression Rating Scale (HDRS). Three components of variation in KOR availability across ROIs were identified, spatially characterized by [11C]GR103545 VT in (1) bilateral frontal lobe; (2) occipital and parietal cortices, right hippocampus, and putamen; and (3) right anterior cingulate, right superior frontal gyrus and insula, coupled to negative loading in left middle cingulate. In MDD patients, component 3 was negatively associated with symptom severity on the HDRS (r = -0.85, p = 0.0021). There were no group-wise differences in expression of any component between patients and controls. These preliminary findings suggest that KOR signaling in cortical regions relevant to depression, particularly right anterior cingulate, could reflect MDD pathophysiology.
Collapse
|
173
|
Imambocus BN, Zhou F, Formozov A, Wittich A, Tenedini FM, Hu C, Sauter K, Macarenhas Varela E, Herédia F, Casimiro AP, Macedo A, Schlegel P, Yang CH, Miguel-Aliaga I, Wiegert JS, Pankratz MJ, Gontijo AM, Cardona A, Soba P. A neuropeptidergic circuit gates selective escape behavior of Drosophila larvae. Curr Biol 2021; 32:149-163.e8. [PMID: 34798050 DOI: 10.1016/j.cub.2021.10.069] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 10/05/2021] [Accepted: 10/29/2021] [Indexed: 12/26/2022]
Abstract
Animals display selective escape behaviors when faced with environmental threats. Selection of the appropriate response by the underlying neuronal network is key to maximizing chances of survival, yet the underlying network mechanisms are so far not fully understood. Using synapse-level reconstruction of the Drosophila larval network paired with physiological and behavioral readouts, we uncovered a circuit that gates selective escape behavior for noxious light through acute and input-specific neuropeptide action. Sensory neurons required for avoidance of noxious light and escape in response to harsh touch, each converge on discrete domains of neuromodulatory hub neurons. We show that acute release of hub neuron-derived insulin-like peptide 7 (Ilp7) and cognate relaxin family receptor (Lgr4) signaling in downstream neurons are required for noxious light avoidance, but not harsh touch responses. Our work highlights a role for compartmentalized circuit organization and neuropeptide release from regulatory hubs, acting as central circuit elements gating escape responses.
Collapse
Affiliation(s)
- Bibi Nusreen Imambocus
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany; Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Fangmin Zhou
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany; Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Andrey Formozov
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Annika Wittich
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Federico M Tenedini
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Chun Hu
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Kathrin Sauter
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Ednilson Macarenhas Varela
- Integrative Biomedicine Laboratory, CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Rua do Instituto Bacteriológico 5, 1150-082 Lisbon, Portugal
| | - Fabiana Herédia
- Integrative Biomedicine Laboratory, CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Rua do Instituto Bacteriológico 5, 1150-082 Lisbon, Portugal
| | - Andreia P Casimiro
- Integrative Biomedicine Laboratory, CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Rua do Instituto Bacteriológico 5, 1150-082 Lisbon, Portugal
| | - André Macedo
- Integrative Biomedicine Laboratory, CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Rua do Instituto Bacteriológico 5, 1150-082 Lisbon, Portugal
| | - Philipp Schlegel
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Chung-Hui Yang
- Department of Neurobiology, Duke University Medical School, 427E Bryan Research, Durham, NC 27710, USA
| | - Irene Miguel-Aliaga
- MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - J Simon Wiegert
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Michael J Pankratz
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Alisson M Gontijo
- Integrative Biomedicine Laboratory, CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Rua do Instituto Bacteriológico 5, 1150-082 Lisbon, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Albert Cardona
- HHMI Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA; MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK; Department of Physiology, Development, and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
| | - Peter Soba
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany; Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany.
| |
Collapse
|
174
|
Marques F, Falquet L, Vandewyer E, Beets I, Glauser DA. Signaling via the FLP-14/FRPR-19 neuropeptide pathway sustains nociceptive response to repeated noxious stimuli in C. elegans. PLoS Genet 2021; 17:e1009880. [PMID: 34748554 PMCID: PMC8601619 DOI: 10.1371/journal.pgen.1009880] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 11/18/2021] [Accepted: 10/15/2021] [Indexed: 12/13/2022] Open
Abstract
In order to thrive in constantly changing environments, animals must adaptively respond to threatening events. Noxious stimuli are not only processed according to their absolute intensity, but also to their context. Adaptation processes can cause animals to habituate at different rates and degrees in response to permanent or repeated stimuli. Here, we used a forward genetic approach in Caenorhabditis elegans to identify a neuropeptidergic pathway, essential to prevent fast habituation and maintain robust withdrawal responses to repeated noxious stimuli. This pathway involves the FRPR-19A and FRPR-19B G-protein coupled receptor isoforms produced from the frpr-19 gene by alternative splicing. Loss or overexpression of each or both isoforms can impair withdrawal responses caused by the optogenetic activation of the polymodal FLP nociceptor neuron. Furthermore, we identified FLP-8 and FLP-14 as FRPR-19 ligands in vitro. flp-14, but not flp-8, was essential to promote withdrawal response and is part of the same genetic pathway as frpr-19 in vivo. Expression and cell-specific rescue analyses suggest that FRPR-19 acts both in the FLP nociceptive neurons and downstream interneurons, whereas FLP-14 acts from interneurons. Importantly, genetic impairment of the FLP-14/FRPR-19 pathway accelerated the habituation to repeated FLP-specific optogenetic activation, as well as to repeated noxious heat and harsh touch stimuli. Collectively, our data suggest that well-adjusted neuromodulation via the FLP-14/FRPR-19 pathway contributes to promote nociceptive signals in C. elegans and counteracts habituation processes that otherwise tend to rapidly reduce aversive responses to repeated noxious stimuli.
Collapse
Affiliation(s)
- Filipe Marques
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Laurent Falquet
- Department of Biology, University of Fribourg, Fribourg, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Elke Vandewyer
- Neural Signaling and Circuit Plasticity Group, Department of Biology, KU Leuven, Leuven, Belgium
| | - Isabel Beets
- Neural Signaling and Circuit Plasticity Group, Department of Biology, KU Leuven, Leuven, Belgium
| | | |
Collapse
|
175
|
Yang T, Yuan Z, Liu C, Liu T, Zhang W. A neural circuit integrates pharyngeal sensation to control feeding. Cell Rep 2021; 37:109983. [PMID: 34758309 DOI: 10.1016/j.celrep.2021.109983] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 08/20/2021] [Accepted: 10/20/2021] [Indexed: 11/18/2022] Open
Abstract
Swallowing is an essential step of eating and drinking. However, how the quality of a food bolus is sensed by pharyngeal neurons is largely unknown. Here we find that mechanical receptors along the Drosophila pharynx are required for control of meal size, especially for food of high viscosity. The mechanical force exerted by the bolus passing across the pharynx is detected by neurons expressing the mechanotransduction channel NOMPC (no mechanoreceptor potential C) and is relayed, together with gustatory information, to IN1 neurons in the subesophageal zone (SEZ) of the brain. IN1 (ingestion neurons) neurons act directly upstream of a group of peptidergic neurons that encode satiety. Prolonged activation of IN1 neurons suppresses feeding. IN1 neurons receive inhibition from DSOG1 (descending subesophageal neurons) neurons, a group of GABAergic neurons that non-selectively suppress feeding. Our results reveal the function of pharyngeal mechanoreceptors and their downstream neural circuits in the control of food ingestion.
Collapse
Affiliation(s)
- Tingting Yang
- School of Life Sciences, Tsinghua-Peking Joint Center for Life Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
| | - Zixuan Yuan
- School of Life Sciences, Tsinghua-Peking Joint Center for Life Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
| | - Chenxi Liu
- School of Life Sciences, Tsinghua-Peking Joint Center for Life Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
| | - Ting Liu
- School of Life Sciences, Tsinghua-Peking Joint Center for Life Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
| | - Wei Zhang
- School of Life Sciences, Tsinghua-Peking Joint Center for Life Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China.
| |
Collapse
|
176
|
Quinn JP, Kandigian SE, Trombetta BA, Arnold SE, Carlyle BC. VGF as a biomarker and therapeutic target in neurodegenerative and psychiatric diseases. Brain Commun 2021; 3:fcab261. [PMID: 34778762 PMCID: PMC8578498 DOI: 10.1093/braincomms/fcab261] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 09/01/2021] [Accepted: 09/13/2021] [Indexed: 12/20/2022] Open
Abstract
Neurosecretory protein VGF (non-acronymic) belongs to the granin family of neuropeptides. VGF and VGF-derived peptides have been repeatedly identified in well-powered and well-designed multi-omic studies as dysregulated in neurodegenerative and psychiatric diseases. New therapeutics is urgently needed for these devastating and costly diseases, as are new biomarkers to improve disease diagnosis and mechanistic understanding. From a list of 537 genes involved in Alzheimer's disease pathogenesis, VGF was highlighted by the Accelerating Medicines Partnership in Alzheimer's disease as the potential therapeutic target of greatest interest. VGF levels are consistently decreased in brain tissue and CSF samples from patients with Alzheimer's disease compared to controls, and its levels correlate with disease severity and Alzheimer's disease pathology. In the brain, VGF exists as multiple functional VGF-derived peptides. Full-length human VGF1-615 undergoes proteolytic processing by prohormone convertases and other proteases in the regulated secretory pathway to produce at least 12 active VGF-derived peptides. In cell and animal models, these VGF-derived peptides have been linked to energy balance regulation, neurogenesis, synaptogenesis, learning and memory, and depression-related behaviours throughout development and adulthood. The C-terminal VGF-derived peptides, TLQP-62 (VGF554-615) and TLQP-21 (VGF554-574) have differential effects on Alzheimer's disease pathogenesis, neuronal and microglial activity, and learning and memory. TLQP-62 activates neuronal cell-surface receptors and regulates long-term hippocampal memory formation. TLQP-62 also prevents immune-mediated memory impairment, depression-like and anxiety-like behaviours in mice. TLQP-21 binds to microglial cell-surface receptors, triggering microglial chemotaxis and phagocytosis. These actions were reported to reduce amyloid-β plaques and decrease neuritic dystrophy in a transgenic mouse model of familial Alzheimer's disease. Expression differences of VGF-derived peptides have also been associated with frontotemporal lobar dementias, amyotrophic lateral sclerosis, Lewy body diseases, Huntington's disease, pain, schizophrenia, bipolar disorder, depression and antidepressant response. This review summarizes current knowledge and highlights questions for future investigation regarding the roles of VGF and its dysregulation in neurodegenerative and psychiatric disease. Finally, the potential of VGF and VGF-derived peptides as biomarkers and novel therapeutic targets for neurodegenerative and psychiatric diseases is highlighted.
Collapse
Affiliation(s)
- James P Quinn
- Department of Neurology, Alzheimer's Clinical & Translational Research Unit, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Savannah E Kandigian
- Department of Neurology, Alzheimer's Clinical & Translational Research Unit, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Bianca A Trombetta
- Department of Neurology, Alzheimer's Clinical & Translational Research Unit, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Steven E Arnold
- Department of Neurology, Alzheimer's Clinical & Translational Research Unit, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Becky C Carlyle
- Department of Neurology, Alzheimer's Clinical & Translational Research Unit, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
177
|
NPF activates a specific NPF receptor and regulates food intake in Pacific abalone Haliotis discus hannai. Sci Rep 2021; 11:20912. [PMID: 34686694 PMCID: PMC8536682 DOI: 10.1038/s41598-021-00238-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 10/07/2021] [Indexed: 01/13/2023] Open
Abstract
Neuropeptides function through G protein-coupled receptors (GPCRs) with high specificity, implying a significant degree of neuropeptide-GPCR coevolution. However, potential neuropeptide signaling systems in non-chordates are relatively elusive. We determined the specificity of the neuropeptide F (Hdh-NPF) signaling system with a cognate receptor (Hdh-NPFR) in the Pacific abalone, Haliotis discus hannai. Phylogenetic and exon–intron arrangement analyses of bilaterian NPF and the chordate ortholog NPY with their receptor sequences revealed a likely common ancestor, and Hdh-NPFR was similar to the NPYR2 subtype among the NPYR1, NPYR2, and NPYR5 subtypes. Among four Hdh-NPFR-related receptors, Hdh-NPFR specifically responded to Hdh-NPF peptide, supported by the dose–response luciferase reporter curve, intracellular Ca2+ mobilization, and phosphorylation of ERK1/2 and its inhibition with a protein kinase C inhibitor. Peptide fragmentations and shuffling of Hdh-NPF with human NPY could not activate the cellular response of Hdh-NPFR. Three-dimensional in silico modeling suggested that interaction of Hdh-NPF C-terminal amino acids with the extracellular loops of Hdh-NPFR is critical for Hdh-NPFR activation. In vivo injection of Hdh-NPF peptide increased food consumption, and knockdown of Hdh-NPF expression decreased food consumption in Pacific abalone. These findings provide evidence for co-evolution of the NPF/Y ligand-receptor system, enabling further research on mollusk orexigenic neuropeptides.
Collapse
|
178
|
Abstract
Itch is one of the most primal sensations, being both ubiquitous and important for the well-being of animals. For more than a century, a desire to understand how itch is encoded by the nervous system has prompted the advancement of many theories. Within the past 15 years, our understanding of the molecular and neural mechanisms of itch has undergone a major transformation, and this remarkable progress continues today without any sign of abating. Here I describe accumulating evidence that indicates that itch is distinguished from pain through the actions of itch-specific neuropeptides that relay itch information to the spinal cord. According to this model, classical neurotransmitters transmit, inhibit and modulate itch information in a context-, space- and time-dependent manner but do not encode itch specificity. Gastrin-releasing peptide (GRP) is proposed to be a key itch-specific neuropeptide, with spinal neurons expressing GRP receptor (GRPR) functioning as a key part of a convergent circuit for the conveyance of peripheral itch information to the brain.
Collapse
|
179
|
Pomrenze MB, Walker LC, Giardino WJ. Gray areas: Neuropeptide circuits linking the Edinger-Westphal and Dorsal Raphe nuclei in addiction. Neuropharmacology 2021; 198:108769. [PMID: 34481834 PMCID: PMC8484048 DOI: 10.1016/j.neuropharm.2021.108769] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/20/2021] [Accepted: 08/25/2021] [Indexed: 01/16/2023]
Abstract
The circuitry of addiction comprises several neural networks including the midbrain - an expansive region critically involved in the control of motivated behaviors. Midbrain nuclei like the Edinger-Westphal (EW) and dorsal raphe (DR) contain unique populations of neurons that synthesize many understudied neuroactive molecules and are encircled by the periaqueductal gray (PAG). Despite the proximity of these special neuron classes to the ventral midbrain complex and surrounding PAG, functions of the EW and DR remain substantially underinvestigated by comparison. Spanning approximately -3.0 to -5.2 mm posterior from bregma in the mouse, these various cell groups form a continuum of neurons that we refer to collectively as the subaqueductal paramedian zone. Defining how these pathways modulate affective behavioral states presents a difficult, yet conquerable challenge for today's technological advances in neuroscience. In this review, we cover the known contributions of different neuronal subtypes of the subaqueductal paramedian zone. We catalogue these cell types based on their spatial, molecular, connectivity, and functional properties and integrate this information with the existing data on the EW and DR in addiction. We next discuss evidence that links the EW and DR anatomically and functionally, highlighting the potential contributions of an EW-DR circuit to addiction-related behaviors. Overall, we aim to derive an integrated framework that emphasizes the contributions of EW and DR nuclei to addictive states and describes how these cell groups function in individuals suffering from substance use disorders. This article is part of the special Issue on 'Neurocircuitry Modulating Drug and Alcohol Abuse'.
Collapse
Affiliation(s)
- Matthew B Pomrenze
- Dept. of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, 94305-5453, USA
| | - Leigh C Walker
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, 3052, Australia
| | - William J Giardino
- Dept. of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, 94305-5453, USA; Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, 94305-5453, USA.
| |
Collapse
|
180
|
Levite M, Safadi R, Milgrom Y, Massarwa M, Galun E. Neurotransmitters and Neuropeptides decrease PD-1 in T cells of healthy subjects and patients with hepatocellular carcinoma (HCC), and increase their proliferation and eradication of HCC cells. Neuropeptides 2021; 89:102159. [PMID: 34293596 DOI: 10.1016/j.npep.2021.102159] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/04/2021] [Accepted: 05/09/2021] [Indexed: 01/29/2023]
Abstract
T cells of aged people, and of patients with either cancer or severe infections (including COVID-19), are often exhausted, senescent and dysfunctional, leading to increased susceptibilities, complications and mortality. Neurotransmitters and Neuropeptides bind their receptors in T cells, and induce multiple beneficial T cell functions. Yet, T cells of different people vary in the expression levels of Neurotransmitter and Neuropeptide receptors, and in the magnitude of the corresponding effects. Therefore, we performed an individual-based study on T cells of 3 healthy subjects, and 3 Hepatocellular Carcinoma (HCC) patients. HCC usually develops due to chronic inflammation. The inflamed liver induces reduction and inhibition of CD4+ T cells and Natural Killer (NK) cells. Immune-based therapies for HCC are urgently needed. We tested if selected Neurotransmitters and Neuropeptides decrease the key checkpoint protein PD-1 in human T cells, and increase proliferation and killing of HCC cells. First, we confirmed human T cells express all dopamine receptors (DRs), and glutamate receptors (GluRs): AMPA-GluR3, NMDA-R and mGluR. Second, we discovered that either Dopamine, Glutamate, GnRH-II, Neuropeptide Y and/or CGRP (10nM), as well as DR and GluR agonists, induced the following effects: 1. Decreased significantly both %PD-1+ T cells and PD-1 expression level per cell (up to 60% decrease, within 1 h only); 2. Increased significantly the number of T cells that proliferated in the presence of HCC cells (up to 7 fold increase), 3. Increased significantly T cell killing of HCC cells (up to 2 fold increase). 4. Few non-conventional combinations of Neurotransmitters and Neuropeptides had surprising synergistic beneficial effects. We conclude that Dopamine, Glutamate, GnRH-II, Neuropeptide Y and CGRP, alone or in combinations, can decrease % PD-1+ T cells and PD-1 expression per cell, in T cells of both healthy subjects and HCC patients, and increase their proliferation in response to HCC cells and killing of HCC cells. Yet, testing T cells of many more cancer patients is absolutely needed. Based on these findings and previous ones, we designed a novel "Personalized Adoptive Neuro-Immunotherapy", calling for validation of safety and efficacy in clinical trials.
Collapse
Affiliation(s)
- Mia Levite
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel; Institute of Gene Therapy, Hadassah Hebrew University Hospital, Ein Karem, Jerusalem 91120, Israel.
| | - Rifaat Safadi
- The Liver Unit, Hadassah Hebrew University Hospital, Ein Karem, Jerusalem 91120, Israel
| | - Yael Milgrom
- The Liver Unit, Hadassah Hebrew University Hospital, Ein Karem, Jerusalem 91120, Israel
| | - Muhammad Massarwa
- The Liver Unit, Hadassah Hebrew University Hospital, Ein Karem, Jerusalem 91120, Israel
| | - Eithan Galun
- Institute of Gene Therapy, Hadassah Hebrew University Hospital, Ein Karem, Jerusalem 91120, Israel
| |
Collapse
|
181
|
Barranco N, Plá V, Alcolea D, Sánchez-Domínguez I, Fischer-Colbrie R, Ferrer I, Lleó A, Aguado F. Dense core vesicle markers in CSF and cortical tissues of patients with Alzheimer's disease. Transl Neurodegener 2021; 10:37. [PMID: 34565482 PMCID: PMC8466657 DOI: 10.1186/s40035-021-00263-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 09/14/2021] [Indexed: 12/19/2022] Open
Abstract
Background New fluid biomarkers for Alzheimer's disease (AD) that reveal synaptic and neural network dysfunctions are needed for clinical practice and therapeutic trial design. Dense core vesicle (DCV) cargos are promising cerebrospinal fluid (CSF) indicators of synaptic failure in AD patients. However, their value as biomarkers has not yet been determined. Methods Immunoassays were performed to analyze the secretory proteins prohormone convertases PC1/3 and PC2, carboxypeptidase E (CPE), secretogranins SgIII and SgII, and Cystatin C in the cerebral cortex (n = 45, provided by Bellvitge University Hospital) and CSF samples (n = 66, provided by The Sant Pau Initiative on Neurodegeneration cohort) from AD patients (n = 56) and age-matched controls (n = 55).
Results In AD tissues, most DCV proteins were aberrantly accumulated in dystrophic neurites and activated astrocytes, whereas PC1/3, PC2 and CPE were also specifically accumulated in hippocampal granulovacuolar degeneration bodies. AD individuals displayed an overall decline of secretory proteins in the CSF. Interestingly, in AD patients, the CSF levels of prohormone convertases strongly correlated inversely with those of neurodegeneration markers and directly with cognitive impairment status. Conclusions These results demonstrate marked alterations of neuronal-specific prohormone convertases in CSF and cortical tissues of AD patients. The neuronal DCV cargos are biomarker candidates for synaptic dysfunction and neurodegeneration in AD. Supplementary Information The online version contains supplementary material available at 10.1186/s40035-021-00263-0.
Collapse
Affiliation(s)
- Neus Barranco
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, 08028, Barcelona, Spain.,Institute of Neurosciences, University of Barcelona, 08028, Barcelona, Spain
| | - Virginia Plá
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, 08028, Barcelona, Spain.,Institute of Neurosciences, University of Barcelona, 08028, Barcelona, Spain.,Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Daniel Alcolea
- Memory Unit, Department of Neurology, Sant Pau Biomedical Research Institute. Sant Pau Hospital, Autonomous University of Barcelona, 08041, Barcelona, Spain.,Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), 28031, Madrid, Spain
| | - Irene Sánchez-Domínguez
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, 08028, Barcelona, Spain.,Institute of Neurosciences, University of Barcelona, 08028, Barcelona, Spain
| | | | - Isidro Ferrer
- Institute of Neurosciences, University of Barcelona, 08028, Barcelona, Spain.,Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), 28031, Madrid, Spain.,Department of Pathology and Experimental Therapeutics, University of Barcelona, and Bellvitge University Hospital, Bellvitge Biomedical Research Institute, Hospitalet de Llobregat, Spain
| | - Alberto Lleó
- Memory Unit, Department of Neurology, Sant Pau Biomedical Research Institute. Sant Pau Hospital, Autonomous University of Barcelona, 08041, Barcelona, Spain.,Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), 28031, Madrid, Spain
| | - Fernando Aguado
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, 08028, Barcelona, Spain. .,Institute of Neurosciences, University of Barcelona, 08028, Barcelona, Spain.
| |
Collapse
|
182
|
Chrobok L, Jeczmien-Lazur JS, Bubka M, Pradel K, Klekocinska A, Klich JD, Ridla Rahim A, Myung J, Kepczynski M, Lewandowski MH. Daily coordination of orexinergic gating in the rat superior colliculus-Implications for intrinsic clock activities in the visual system. FASEB J 2021; 35:e21930. [PMID: 34533886 DOI: 10.1096/fj.202100779rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/24/2021] [Accepted: 08/31/2021] [Indexed: 01/07/2023]
Abstract
The orexinergic system delivers excitation for multiple brain centers to facilitate behavioral arousal, with its malfunction resulting in narcolepsy, somnolence, and notably, visual hallucinations. Since the circadian clock underlies the daily arousal, a timed coordination is expected between the orexin system and its target subcortical visual system, including the superior colliculus (SC). Here, we use a combination of electrophysiological, immunohistochemical, and molecular approaches across 24 h, together with the neuronal tract-tracing methods to investigate the daily coordination between the orexin system and the rodent SC. Higher orexinergic input was found to occur nocturnally in the superficial layers of the SC, in time for nocturnal silencing of spontaneous firing in this visual brain area. We identify autonomous daily and circadian expression of clock genes in the SC, which may underlie these day-night changes. Additionally, we establish the lateral hypothalamic origin of the orexin innervation to the SC and that the SC neurons robustly respond to orexin A via OX2 receptor in both excitatory and GABAA receptor-dependent inhibitory manners. Together, our evidence elucidates the combination of intrinsic and extrinsic clock mechanisms that shape the daily function of the visual layers of the SC.
Collapse
Affiliation(s)
- Lukasz Chrobok
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Jagoda Stanislawa Jeczmien-Lazur
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Monika Bubka
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Kamil Pradel
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Aleksandra Klekocinska
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Jasmin Daniela Klich
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Amalia Ridla Rahim
- Graduate Institute of Mind, Brain, and Consciousness, Taipei Medical University, Taipei, Taiwan
| | - Jihwan Myung
- Graduate Institute of Mind, Brain, and Consciousness, Taipei Medical University, Taipei, Taiwan.,Brain and Consciousness Research Centre, Taipei Medical University-Shuang Ho Hospital, Ministry of Health and Welfare, New Taipei City, Taiwan
| | - Mariusz Kepczynski
- Department of Physical Chemistry and Electrochemistry, Faculty of Chemistry, Jagiellonian University in Krakow, Krakow, Poland
| | - Marian Henryk Lewandowski
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| |
Collapse
|
183
|
Wong WLE, Dawe GS, Young AH. The putative role of the relaxin-3/RXFP3 system in clinical depression and anxiety: A systematic literature review. Neurosci Biobehav Rev 2021; 131:429-450. [PMID: 34537263 DOI: 10.1016/j.neubiorev.2021.09.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 09/13/2021] [Accepted: 09/13/2021] [Indexed: 12/14/2022]
Abstract
The relaxin-3/RXFP3 system is one of several neuropeptidergic systems putatively implicated in regulating the behavioural alterations that characterise clinical depression and anxiety, making it a potential target for clinical translation. Accordingly, this systematic review identified published reports on the role of relaxin-3/RXFP3 signalling in these neuropsychiatric disorders and their behavioural endophenotypes, evaluating evidence from animal and human studies to ascertain any relationship. We searched PubMed, EMBASE, PsycINFO and Google Scholar databases up to February 2021, finding 609 relevant records. After stringent screening, 51 of these studies were included in the final synthesis. There was considerable heterogeneity in study designs and some inconsistency across study outcomes. However, experimental evidence is consistent with an ability of relaxin-3/RXFP3 signalling to promote arousal and suppress depressive- and anxiety-like behaviour. Moreover, meta-analyses of six to eight articles investigating food intake revealed that acute RXFP3 activation had strong orexigenic effects in rats. This appraisal also identified the lack of high-quality clinical studies pertinent to the relaxin-3/RXFP3 system, a gap that future research should attempt to bridge.
Collapse
Affiliation(s)
- Win Lee Edwin Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom.
| | - Gavin Stewart Dawe
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Neurobiology and Ageing Programme, Life Sciences Institute, National University of Singapore, Singapore
| | - Allan H Young
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom; South London & Maudsley NHS Foundation Trust, Bethlem Royal Hospital, Monks Orchard Road, London, United Kingdom
| |
Collapse
|
184
|
Wen G, Pang H, Wu X, Jiang E, Zhang X, Zhan X. Proteomic characterization of secretory granules in dopaminergic neurons indicates chromogranin/secretogranin-mediated protein processing impairment in Parkinson's disease. Aging (Albany NY) 2021; 13:20335-20358. [PMID: 34420933 PMCID: PMC8436928 DOI: 10.18632/aging.203415] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 08/03/2021] [Indexed: 11/25/2022]
Abstract
Parkinson’s disease (PD) is an aging disorder related to vesicle transport dysfunctions and neurotransmitter secretion. Secretory granules (SGs) are large dense-core vesicles for the biosynthesis of neuropeptides and hormones. At present, the involvement of SGs impairment in PD remains unclear. In the current study, we found that the number of SGs in tyrosine hydroxylase-positive neurons and the marker proteins secretogranin III (Scg3) significantly decreased in the substantia nigra and striatum regions of 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) exposed mice. Proteomic study of SGs purified from the dopaminergic SH-sy5Y cells under 1-methyl-4-phenylpyridinium (MPP+) treatments (ProteomeXchange PXD023937) identified 536 significantly differentially expressed proteins. The result indicated that disabled lysosome and peroxisome, lipid and energy metabolism disorders are three characteristic features. Protein-protein interaction analysis of 56 secretory proteins and 140 secreted proteins suggested that the peptide processing mediated by chromogranin/secretogranin in SGs was remarkably compromised, accompanied by decreased candidate proteins and peptides neurosecretory protein (VGF), neuropeptide Y, apolipoprotein E, and an increased level of proenkephalin. The current study provided an extensive proteinogram of SGs in PD. It is helpful to understand the molecular mechanisms in the disease.
Collapse
Affiliation(s)
- Gehua Wen
- School of Forensic Medicine, China Medical University, Shenyang, PR China
| | - Hao Pang
- School of Forensic Medicine, China Medical University, Shenyang, PR China
| | - Xu Wu
- School of Forensic Medicine, China Medical University, Shenyang, PR China
| | - Enzhu Jiang
- School of Forensic Medicine, China Medical University, Shenyang, PR China
| | - Xique Zhang
- Department of Geriatrics, The First Affiliated Hospital of China Medical University, Shenyang, PR China
| | - Xiaoni Zhan
- School of Forensic Medicine, China Medical University, Shenyang, PR China
| |
Collapse
|
185
|
Selective Expression of a SNARE-Cleaving Protease in Peripheral Sensory Neurons Attenuates Pain-Related Gene Transcription and Neuropeptide Release. Int J Mol Sci 2021; 22:ijms22168826. [PMID: 34445536 PMCID: PMC8396265 DOI: 10.3390/ijms22168826] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/08/2021] [Accepted: 08/12/2021] [Indexed: 12/31/2022] Open
Abstract
Chronic pain is a leading health and socioeconomic problem and an unmet need exists for long-lasting analgesics. SNAREs (soluble N-ethylmaleimide-sensitive factor attachment protein receptors) are required for neuropeptide release and noxious signal transducer surface trafficking, thus, selective expression of the SNARE-cleaving light-chain protease of botulinum neurotoxin A (LCA) in peripheral sensory neurons could alleviate chronic pain. However, a safety concern to this approach is the lack of a sensory neuronal promoter to prevent the expression of LCA in the central nervous system. Towards this, we exploit the unique characteristics of Pirt (phosphoinositide-interacting regulator of TRP), which is expressed in peripheral nociceptive neurons. For the first time, we identified a Pirt promoter element and cloned it into a lentiviral vector driving transgene expression selectively in peripheral sensory neurons. Pirt promoter driven-LCA expression yielded rapid and concentration-dependent cleavage of SNAP-25 in cultured sensory neurons. Moreover, the transcripts of pain-related genes (TAC1, tachykinin precursor 1; CALCB, calcitonin gene-related peptide 2; HTR3A, 5-hydroxytryptamine receptor 3A; NPY2R, neuropeptide Y receptor Y2; GPR52, G protein-coupled receptor 52; SCN9A, sodium voltage-gated channel alpha subunit 9; TRPV1 and TRPA1, transient receptor potential cation channel subfamily V member 1 and subfamily A member 1) in pro-inflammatory cytokines stimulated sensory neurons were downregulated by viral mediated expression of LCA. Furthermore, viral expression of LCA yielded long-lasting inhibition of pain mediator release. Thus, we show that the engineered Pirt-LCA virus may provide a novel means for long lasting pain relief.
Collapse
|
186
|
Cellular context shapes cyclic nucleotide signaling in neurons through multiple levels of integration. J Neurosci Methods 2021; 362:109305. [PMID: 34343574 DOI: 10.1016/j.jneumeth.2021.109305] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/22/2021] [Accepted: 07/29/2021] [Indexed: 02/06/2023]
Abstract
Intracellular signaling with cyclic nucleotides are ubiquitous signaling pathways, yet the dynamics of these signals profoundly differ in different cell types. Biosensor imaging experiments, by providing direct measurements in intact cellular environment, reveal which receptors are activated by neuromodulators and how the coincidence of different neuromodulators is integrated at various levels in the signaling cascade. Phosphodiesterases appear as one important determinant of cross-talk between different signaling pathways. Finally, analysis of signal dynamics reveal that striatal medium-sized spiny neuron obey a different logic than other brain regions such as cortex, probably in relation with the function of this brain region which efficiently detects transient dopamine.
Collapse
|
187
|
Kalkan ÖF, Şahin Z, Öztürk H, Keser H, Aydın-Abidin S, Abidin İ. Phoenixin-14 reduces the frequency of interictal-like events in mice brain slices. Exp Brain Res 2021; 239:2841-2849. [PMID: 34283252 DOI: 10.1007/s00221-021-06179-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/13/2021] [Indexed: 01/17/2023]
Abstract
Phoenixin-14 (PNX-14) has a wide bioactivity in the central nervous system. Its role in the hypothalamus has been investigated, and it has been reported that it is involved in the regulation of excitability in hypothalamic neurons. However, its role in the regulation of excitability in entorhinal cortex and the hippocampus is unknown. In this study, we investigated whether i. PNX-14 induces any synchronous discharges or epileptiform activity and ii. PNX-14 has any effect on already initiated epileptiform discharges. We used 350 µm thick acute horizontal hippocampal-entorhinal cortex slices obtained from 30- to 35-day-old mice. Extracellular field potential recordings were evaluated in the entorhinal cortex and hippocampus CA1 region. Bath application of PNX-14 did not initiate any epileptiform activity or abnormal discharges. 4-Aminopyridine was applied to induce epileptiform activity in the slices. We found that 200 nM PNX-14 reduced the frequency of interictal-like events in both the entorhinal cortex and hippocampus CA1 region which was induced by 4-aminopyridine. Furthermore, PNX-14 led to a similar suppression in the total power of local field potentials of 1-120 Hz. The frequency or the duration of the ictal events was not affected. These results exhibited for the first time that PNX-14 has a modulatory effect on synchronized neuronal discharges which should be considered in future therapeutic approaches.
Collapse
Affiliation(s)
- Ömer Faruk Kalkan
- Department of Physiology, Faculty of Medicine, Karadeniz Technical University, 61080, Trabzon, Turkey.
| | - Zafer Şahin
- Department of Physiology, Faculty of Medicine, Karadeniz Technical University, 61080, Trabzon, Turkey
| | - Hilal Öztürk
- Department of Physiology, Faculty of Medicine, Karadeniz Technical University, 61080, Trabzon, Turkey
| | - Hatice Keser
- Department of Physiology, Faculty of Medicine, Karadeniz Technical University, 61080, Trabzon, Turkey
| | - Selcen Aydın-Abidin
- Department of Physiology, Faculty of Medicine, Karadeniz Technical University, 61080, Trabzon, Turkey
| | - İsmail Abidin
- Department of Physiology, Faculty of Medicine, Karadeniz Technical University, 61080, Trabzon, Turkey
| |
Collapse
|
188
|
Intranasal Administration for Pain: Oxytocin and Other Polypeptides. Pharmaceutics 2021; 13:pharmaceutics13071088. [PMID: 34371778 PMCID: PMC8309171 DOI: 10.3390/pharmaceutics13071088] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/09/2021] [Accepted: 07/13/2021] [Indexed: 11/16/2022] Open
Abstract
Pain, particularly chronic pain, remains one of the most debilitating and difficult-to-treat conditions in medicine. Chronic pain is difficult to treat, in part because it is associated with plastic changes in the peripheral and central nervous systems. Polypeptides are linear organic polymers that are highly selective molecules for neurotransmitter and other nervous system receptors sites, including those associated with pain and analgesia, and so have tremendous potential in pain therapeutics. However, delivery of polypeptides to the nervous system is largely limited due to rapid degradation within the peripheral circulation as well as the blood–brain barrier. One strategy that has been shown to be successful in nervous system deposition of polypeptides is intranasal (IN) delivery. In this narrative review, we discuss the delivery of polypeptides to the peripheral and central nervous systems following IN administration. We briefly discuss the mechanism of delivery via the nasal–cerebral pathway. We review recent studies that demonstrate that polypeptides such as oxytocin, delivered IN, not only reach key pain-modulating regions in the nervous system but, in doing so, evoke significant analgesic effects. IN administration of polypeptides has tremendous potential to provide a non-invasive, rapid and effective method of delivery to the nervous system for chronic pain treatment and management.
Collapse
|
189
|
Corradi L, Filosa A. Neuromodulation and Behavioral Flexibility in Larval Zebrafish: From Neurotransmitters to Circuits. Front Mol Neurosci 2021; 14:718951. [PMID: 34335183 PMCID: PMC8319623 DOI: 10.3389/fnmol.2021.718951] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 06/25/2021] [Indexed: 11/13/2022] Open
Abstract
Animals adapt their behaviors to their ever-changing needs. Internal states, such as hunger, fear, stress, and arousal are important behavioral modulators controlling the way an organism perceives sensory stimuli and reacts to them. The translucent zebrafish larva is an ideal model organism for studying neuronal circuits regulating brain states, owning to the possibility of easy imaging and manipulating activity of genetically identified neurons while the animal performs stereotyped and well-characterized behaviors. The main neuromodulatory circuits present in mammals can also be found in the larval zebrafish brain, with the advantage that they contain small numbers of neurons. Importantly, imaging and behavioral techniques can be combined with methods for generating targeted genetic modifications to reveal the molecular underpinnings mediating the functions of such circuits. In this review we discuss how studying the larval zebrafish brain has contributed to advance our understanding of circuits and molecular mechanisms regulating neuromodulation and behavioral flexibility.
Collapse
Affiliation(s)
- Laura Corradi
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Alessandro Filosa
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| |
Collapse
|
190
|
Ouyang Q, Hu S, Li L, Ran M, Zhu J, Zhao Y, Hu B, Hu J, He H, Li L, Wang J. Integrated mRNA and miRNA transcriptome analysis provides novel insights into the molecular mechanisms underlying goose pituitary development during the embryo-to-hatchling transition. Poult Sci 2021; 100:101380. [PMID: 34358958 PMCID: PMC8350522 DOI: 10.1016/j.psj.2021.101380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 07/01/2021] [Accepted: 07/04/2021] [Indexed: 11/25/2022] Open
Abstract
It is well established that the endocrine system plays a pivotal role in preparing the avian embryos for the abrupt switch from chorioallantoic to pulmonary respiration during the critical embryo-to-hatchling transition. However, as the master gland of the endocrine system, there has been little research focusing on the molecular mechanisms controlling the development and function of the pituitary gland during the peri-hatch period in birds. In the present study, we aimed to determine the genome-wide mRNA and miRNA transcriptome profiles of the pituitary during the embryo-to-hatchling transition period from embryonic day 22 (E22) to post-hatching day 6 (P6) in the goose (Anser cygnoides). Of note, expression of Anser_cygnoides_newGene_32456 and LOC106031011 were significantly different among these 4 stages (i.e., E22, E26, P2, and P6). Meanwhile, the neuroactive ligand-receptor interaction pathway was significantly enriched by the DEGs commonly identified among three pairwise comparisons. At the miRNA transcriptome level, there were not commonly identified DE miRNAs among these 4 stages, while the 418 of their predicted target genes were mutually shared. Both the target genes of DE miRNAs in each comparison and these 418 shared target genes were significantly enriched in the ECM-receptor interaction and focal adhesion pathways. In the predicted miRNA-mRNA interaction networks of these 2 pathways, novel_miRNA_467, novel_miRNA_154, and novel_miRNA_340 were the hub miRNAs. In addition, multiple DE miRNAs also showed predicted target relationships with the DEGs associated with extracellular matrix (ECM) components. Among them, expression of novel_miR_120, tgu-miR-92-3p, and novel_miR_398 was significantly negatively correlated with that of LAMC3 (laminin subunit gamma3), suggesting that these miRNAs may regulate pituitary tissue remodeling and functional changes through targeting LAMC3 during development. These identified DE mRNAs and miRNAs as well as their predicted interaction networks involved in regulation of tissue remodeling and cellular functions were most likely to play critical roles in facilitating the embryo-to-hatchling transition. These results provide novel insights into the early developmental process of avian pituitary gland and will help better understand the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Qingyuan Ouyang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Shenqiang Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Li Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Mingxia Ran
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Jiaran Zhu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Yiting Zhao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Bo Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Jiwei Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Hua He
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Liang Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Jiwen Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| |
Collapse
|
191
|
Jiang Z, Lietz CB, Podvin S, Yoon MC, Toneff T, Hook V, O’Donoghue AJ. Differential Neuropeptidomes of Dense Core Secretory Vesicles (DCSV) Produced at Intravesicular and Extracellular pH Conditions by Proteolytic Processing. ACS Chem Neurosci 2021; 12:2385-2398. [PMID: 34153188 PMCID: PMC8267839 DOI: 10.1021/acschemneuro.1c00133] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
![]()
Neuropeptides mediate
cell–cell signaling in the nervous
and endocrine systems. The neuropeptidome is the spectrum of peptides
generated from precursors by proteolysis within dense core secretory
vesicles (DCSV). DCSV neuropeptides and contents are released to the
extracellular environment where further processing for neuropeptide
formation may occur. To assess the DCSV proteolytic capacity for production
of neuropeptidomes at intravesicular pH 5.5 and extracellular pH 7.2,
neuropeptidomics, proteomics, and protease assays were conducted using
chromaffin granules (CG) purified from adrenal medulla. CG are an
established model of DCSV. The CG neuropeptidome consisted of 1239
unique peptides derived from 15 proneuropeptides that were colocalized
with 64 proteases. Distinct CG neuropeptidomes were generated at the
internal DCSV pH of 5.5 compared to the extracellular pH of 7.2. Class-specific
protease inhibitors differentially regulated neuropeptidome production
involving aspartic, cysteine, serine, and metallo proteases. The substrate
cleavage properties of CG proteases were assessed by multiplex substrate
profiling by mass spectrometry (MSP-MS) that uses a synthetic peptide
library containing diverse cleavage sites for endopeptidases and exopeptidases.
Parallel inhibitor-sensitive cleavages for neuropeptidome production
and peptide library proteolysis led to elucidation of six CG proteases
involved in neuropeptidome production, represented by cathepsins A,
B, C, D, and L and carboxypeptidase E (CPE). The MSP-MS profiles of
these six enzymes represented the majority of CG proteolytic cleavages
utilized for neuropeptidome production. These findings provide new
insight into the DCSV proteolytic system for production of distinct
neuropeptidomes at the internal CG pH of 5.5 and at the extracellular
pH of 7.2.
Collapse
Affiliation(s)
- Zhenze Jiang
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093, United States
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California 92093, United States
| | - Christopher B. Lietz
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093, United States
| | - Sonia Podvin
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093, United States
| | - Michael C. Yoon
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093, United States
| | - Thomas Toneff
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093, United States
| | - Vivian Hook
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093, United States
- Department of Neuroscience and Department of Pharmacology, University of California, San Diego, La Jolla, California 92093, United States
| | - Anthony J. O’Donoghue
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093, United States
| |
Collapse
|
192
|
Cocaine-induced neural adaptations in the lateral hypothalamic melanin-concentrating hormone neurons and the role in regulating rapid eye movement sleep after withdrawal. Mol Psychiatry 2021; 26:3152-3168. [PMID: 33093653 PMCID: PMC8060355 DOI: 10.1038/s41380-020-00921-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 09/30/2020] [Accepted: 10/07/2020] [Indexed: 12/21/2022]
Abstract
Sleep abnormalities are often a prominent contributor to withdrawal symptoms following chronic drug use. Notably, rapid eye movement (REM) sleep regulates emotional memory, and persistent REM sleep impairment after cocaine withdrawal negatively impacts relapse-like behaviors in rats. However, it is not understood how cocaine experience may alter REM sleep regulatory machinery, and what may serve to improve REM sleep after withdrawal. Here, we focus on the melanin-concentrating hormone (MCH) neurons in the lateral hypothalamus (LH), which regulate REM sleep initiation and maintenance. Using adult male Sprague-Dawley rats trained to self-administer intravenous cocaine, we did transcriptome profiling of LH MCH neurons after long-term withdrawal using RNA-sequencing, and performed functional assessment using slice electrophysiology. We found that 3 weeks after withdrawal from cocaine, LH MCH neurons exhibit a wide range of gene expression changes tapping into cell membrane signaling, intracellular signaling, and transcriptional regulations. Functionally, they show reduced membrane excitability and decreased glutamatergic receptor activity, consistent with increased expression of voltage-gated potassium channel gene Kcna1 and decreased expression of metabotropic glutamate receptor gene Grm5. Finally, chemogenetic or optogenetic stimulations of LH MCH neural activity increase REM sleep after long-term withdrawal with important differences. Whereas chemogenetic stimulation promotes both wakefulness and REM sleep, optogenetic stimulation of these neurons in sleep selectively promotes REM sleep. In summary, cocaine exposure persistently alters gene expression profiles and electrophysiological properties of LH MCH neurons. Counteracting cocaine-induced hypoactivity of these neurons selectively in sleep enhances REM sleep quality and quantity after long-term withdrawal.
Collapse
|
193
|
Analgesic effect of central relaxin receptor activation on persistent inflammatory pain in mice: behavioral and neurochemical data. Pain Rep 2021; 6:e937. [PMID: 34159282 PMCID: PMC8213244 DOI: 10.1097/pr9.0000000000000937] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/26/2021] [Accepted: 04/23/2021] [Indexed: 01/02/2023] Open
Abstract
Supplemental Digital Content is Available in the Text. Relaxin peptide analogues produce strong but transient analgesia in inflammatory pain in mouse. Relaxin and its RXFP1 receptor represent a new peptidergic system that modulates pain processing in the forebrain areas. Introduction: The relaxin peptide signaling system is involved in diverse physiological processes, but its possible roles in the brain, including nociception, are largely unexplored. Objective: In light of abundant expression of relaxin receptor (RXFP1) mRNA/protein in brain regions involved in pain processing, we investigated the effects of central RXFP1 activation on nociceptive behavior in a mouse model of inflammatory pain and examined the neurochemical phenotype and connectivity of relaxin and RXFP1 mRNA-positive neurons. Methods: Mice were injected with Complete Freund Adjuvant (CFA) into a hind paw. After 4 days, the RXFP1 agonist peptides, H2-relaxin or B7-33, ± the RXFP1 antagonist, B-R13/17K-H2, were injected into the lateral cerebral ventricle, and mechanical and thermal sensitivity were assessed at 30 to 120 minutes. Relaxin and RXFP1 mRNA in excitatory and inhibitory neurons were examined using multiplex, fluorescent in situ hybridization. Relaxin-containing neurons were detected using immunohistochemistry and their projections assessed using fluorogold retrograde tract-tracing. Results: Both H2-relaxin and B7-33 produced a strong, but transient, reduction in mechanical and thermal sensitivity of the CFA-injected hind paw alone, at 30 minutes postinjection. Notably, coinjection of B-R13/17K-H2 blocked mechanical, but not thermal, analgesia. In the claustrum, cingulate cortex, and subiculum, RXFP1 mRNA was expressed in excitatory neurons. Relaxin immunoreactivity was detected in neurons in forebrain and midbrain areas involved in pain processing and sending projections to the RXFP1-rich, claustrum and cingulate cortex. No changes were detected in CFA mice. Conclusion: Our study identified a previously unexplored peptidergic system that can control pain processing in the brain and produce analgesia.
Collapse
|
194
|
Jamieson BB, Bouwer GT, Campbell RE, Piet R. Estrous Cycle Plasticity in the Central Clock Output to Kisspeptin Neurons: Implications for the Preovulatory Surge. Endocrinology 2021; 162:6213415. [PMID: 33824970 DOI: 10.1210/endocr/bqab071] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Indexed: 11/19/2022]
Abstract
Coordination of ovulation and behavior is critical to reproductive success in many species. During the female estrous cycle, the preovulatory gonadotropin surge occurs when ovarian follicles reach maturity and, in rodents, it begins just before the daily onset of activity, ensuring that ovulation coincides with sex behavior. Timing of the surge relies on projections from the suprachiasmatic nucleus (SCN), the locus of the central circadian clock, to hypothalamic circuits that regulate gonadotropin secretion. The cellular mechanisms through which the SCN controls these circuits and gates the preovulatory surge to the appropriate estrous cycle stage, however, are poorly understood. We investigated in mice the functional impact of SCN arginine-vasopressin (AVP) neuron projections to kisspeptin (Kiss1) neurons in the rostral periventricular area of the third ventricle (RP3VKiss1), responsible for generating the preovulatory surge. Conditional anterograde tracing revealed that SCNAVP neurons innervate approximately half of the RP3VKiss1 neurons. Optogenetic activation of SCNAVP projections in brain slices caused an AVP-mediated stimulation of RP3VKiss1 action potential firing in proestrus, the cycle stage when the surge is generated. This effect was less prominent in diestrus, the preceding cycle stage, and absent in estrus, following ovulation. Remarkably, in estrus, activation of SCNAVP projections resulted in GABA-mediated inhibition of RP3VKiss1 neuron firing, an effect rarely encountered in other cycle stages. Together, these data reveal functional plasticity in SCNAVP neuron output that drives opposing effects on RP3VKiss1 neuron activity across the ovulatory cycle. This might contribute to gating activation of the preovulatory surge to the appropriate estrous cycle stage.
Collapse
Affiliation(s)
- Bradley B Jamieson
- Centre for Neuroendocrinology & Department of Physiology, University of Otago, Dunedin 9054, New Zealand
| | - Gregory T Bouwer
- Centre for Neuroendocrinology & Department of Physiology, University of Otago, Dunedin 9054, New Zealand
| | - Rebecca E Campbell
- Centre for Neuroendocrinology & Department of Physiology, University of Otago, Dunedin 9054, New Zealand
| | - Richard Piet
- Centre for Neuroendocrinology & Department of Physiology, University of Otago, Dunedin 9054, New Zealand
- Brain Health Research Institute & Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
| |
Collapse
|
195
|
Holt MK, Rinaman L. The role of nucleus of the solitary tract glucagon-like peptide-1 and prolactin-releasing peptide neurons in stress: anatomy, physiology and cellular interactions. Br J Pharmacol 2021; 179:642-658. [PMID: 34050926 PMCID: PMC8820208 DOI: 10.1111/bph.15576] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 05/04/2021] [Accepted: 05/17/2021] [Indexed: 02/06/2023] Open
Abstract
Neuroendocrine, behavioural and autonomic responses to stressful stimuli are orchestrated by complex neural circuits. The caudal nucleus of the solitary tract (cNTS) in the dorsomedial hindbrain is uniquely positioned to integrate signals of both interoceptive and psychogenic stress. Within the cNTS, glucagon‐like peptide‐1 (GLP‐1) and prolactin‐releasing peptide (PrRP) neurons play crucial roles in organising neural responses to a broad range of stressors. In this review we discuss the anatomical and functional overlap between PrRP and GLP‐1 neurons. We outline their co‐activation in response to stressful stimuli and their importance as mediators of behavioural and physiological stress responses. Finally, we review evidence that PrRP neurons are downstream of GLP‐1 neurons and outline unexplored areas of the research field. Based on the current state‐of‐knowledge, PrRP and GLP‐1 neurons may be compelling targets in the treatment of stress‐related disorders.
Collapse
Affiliation(s)
- Marie K Holt
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Linda Rinaman
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida, USA
| |
Collapse
|
196
|
Hidalgo S, Campusano JM, Hodge JJL. Assessing olfactory, memory, social and circadian phenotypes associated with schizophrenia in a genetic model based on Rim. Transl Psychiatry 2021; 11:292. [PMID: 34001859 PMCID: PMC8128896 DOI: 10.1038/s41398-021-01418-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 04/22/2021] [Accepted: 04/30/2021] [Indexed: 02/04/2023] Open
Abstract
Schizophrenia shows high heritability and several of the genes associated with this disorder are involved in calcium (Ca2+) signalling and synaptic function. One of these is the Rab-3 interacting molecule-1 (RIM1), which has recently been associated with schizophrenia by Genome Wide Association Studies (GWAS). However, its contribution to the pathophysiology of this disorder remains unexplored. In this work, we use Drosophila mutants of the orthologue of RIM1, Rim, to model some aspects of the classical and non-classical symptoms of schizophrenia. Rim mutants showed several behavioural features relevant to schizophrenia including social distancing and altered olfactory processing. These defects were accompanied by reduced evoked Ca2+ influx and structural changes in the presynaptic terminals sent by the primary olfactory neurons to higher processing centres. In contrast, expression of Rim-RNAi in the mushroom bodies (MBs), the main memory centre in flies, spared learning and memory suggesting a differential role of Rim in different synapses. Circadian deficits have been reported in schizophrenia. We observed circadian locomotor activity deficits in Rim mutants, revealing a role of Rim in the pacemaker ventral lateral clock neurons (LNvs). These changes were accompanied by impaired day/night remodelling of dorsal terminal synapses from a subpopulation of LNvs and impaired day/night release of the circadian neuropeptide pigment dispersing factor (PDF) from these terminals. Lastly, treatment with the commonly used antipsychotic haloperidol rescued Rim locomotor deficits to wildtype. This work characterises the role of Rim in synaptic functions underlying behaviours disrupted in schizophrenia.
Collapse
Affiliation(s)
- Sergio Hidalgo
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- School of Physiology, Pharmacology and Neuroscience, Faculty of Life Science, University of Bristol, Bristol, UK
| | - Jorge M Campusano
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - James J L Hodge
- School of Physiology, Pharmacology and Neuroscience, Faculty of Life Science, University of Bristol, Bristol, UK.
| |
Collapse
|
197
|
Zhang Y, Zhang H, Jiang B, Tong X, Yan S, Lu J. Current views on neuropeptides in atopic dermatitis. Exp Dermatol 2021; 30:1588-1597. [PMID: 33963624 DOI: 10.1111/exd.14382] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 12/19/2022]
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease involving skin barrier dysfunction and immune imbalance. However, the mechanism of AD is not clear completely and may be related to heredity and environment. Neuropeptides are a class of peptides secreted by nerve endings, they may play roles in promoting vasodilation, plasma extravasation, chemotaxis of inflammatory cells and mediating pruritus. Since itching and immune cell infiltration are the main manifestations of atopic dermatitis, to further investigate the impact of neuropeptides on AD, our review summarized the mechanisms of several common neuropeptides in AD and hypothesized that neuropeptides may be the novel potential targets in AD treatment.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China.,Xiangya School of Medicine, Central South University, Changsha, China
| | - Hanyi Zhang
- Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China.,Xiangya School of Medicine, Central South University, Changsha, China
| | - Boyue Jiang
- Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China.,Xiangya School of Medicine, Central South University, Changsha, China
| | - Xiaoliang Tong
- Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Siyu Yan
- Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jianyun Lu
- Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
198
|
Key B, Zalucki O, Brown DJ. Neural Design Principles for Subjective Experience: Implications for Insects. Front Behav Neurosci 2021; 15:658037. [PMID: 34025371 PMCID: PMC8131515 DOI: 10.3389/fnbeh.2021.658037] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/07/2021] [Indexed: 02/04/2023] Open
Abstract
How subjective experience is realized in nervous systems remains one of the great challenges in the natural sciences. An answer to this question should resolve debate about which animals are capable of subjective experience. We contend that subjective experience of sensory stimuli is dependent on the brain's awareness of its internal neural processing of these stimuli. This premise is supported by empirical evidence demonstrating that disruption to either processing streams or awareness states perturb subjective experience. Given that the brain must predict the nature of sensory stimuli, we reason that conscious awareness is itself dependent on predictions generated by hierarchically organized forward models of the organism's internal sensory processing. The operation of these forward models requires a specialized neural architecture and hence any nervous system lacking this architecture is unable to subjectively experience sensory stimuli. This approach removes difficulties associated with extrapolations from behavioral and brain homologies typically employed in addressing whether an animal can feel. Using nociception as a model sensation, we show here that the Drosophila brain lacks the required internal neural connectivity to implement the computations required of hierarchical forward models. Consequently, we conclude that Drosophila, and those insects with similar neuroanatomy, do not subjectively experience noxious stimuli and therefore cannot feel pain.
Collapse
Affiliation(s)
- Brian Key
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Oressia Zalucki
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Deborah J. Brown
- School of Historical and Philosophical Inquiry, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
199
|
van Westen R, Poppinga J, Díez Arazola R, Toonen RF, Verhage M. Neuromodulator release in neurons requires two functionally redundant calcium sensors. Proc Natl Acad Sci U S A 2021; 118:e2012137118. [PMID: 33903230 PMCID: PMC8106342 DOI: 10.1073/pnas.2012137118] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Neuropeptides and neurotrophic factors secreted from dense core vesicles (DCVs) control many brain functions, but the calcium sensors that trigger their secretion remain unknown. Here, we show that in mouse hippocampal neurons, DCV fusion is strongly and equally reduced in synaptotagmin-1 (Syt1)- or Syt7-deficient neurons, but combined Syt1/Syt7 deficiency did not reduce fusion further. Cross-rescue, expression of Syt1 in Syt7-deficient neurons, or vice versa, completely restored fusion. Hence, both sensors are rate limiting, operating in a single pathway. Overexpression of either sensor in wild-type neurons confirmed this and increased fusion. Syt1 traveled with DCVs and was present on fusing DCVs, but Syt7 supported fusion largely from other locations. Finally, the duration of single DCV fusion events was reduced in Syt1-deficient but not Syt7-deficient neurons. In conclusion, two functionally redundant calcium sensors drive neuromodulator secretion in an expression-dependent manner. In addition, Syt1 has a unique role in regulating fusion pore duration.
Collapse
Affiliation(s)
- Rhodé van Westen
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Department of Clinical Genetics, Amsterdam University Medical Centers, 1081 HV Amsterdam, The Netherlands
| | - Josse Poppinga
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Rocío Díez Arazola
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Ruud F Toonen
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands;
| | - Matthijs Verhage
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands;
- Department of Clinical Genetics, Amsterdam University Medical Centers, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
200
|
Arginine Vasopressin-Containing Neurons of the Suprachiasmatic Nucleus Project to CSF. eNeuro 2021; 8:ENEURO.0363-20.2021. [PMID: 33472866 PMCID: PMC8174031 DOI: 10.1523/eneuro.0363-20.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 12/30/2020] [Accepted: 01/04/2021] [Indexed: 02/01/2023] Open
Abstract
While it is well established that there are robust circadian rhythms of arginine vasopressin (AVP) in the cerebrospinal fluid (CSF), the route whereby the peptide reaches the CSF is not clear. A, AVP neurons constitute the largest fraction of the SCN neuronal population. Here, we show that processes of AVP-expressing SCN neurons cross the epithelium of the 3rd ventricular wall to reach the CSF (black arrows). Additionally, we report rostro-caudal differences in AVP neuron size and demonstrate that the localization of cells expressing the clock protein PER2 extend beyond the AVP population, thereby indicating that the size of this nucleus is somewhat larger than previously understood. B, Following lateral ventricle (LV) injection of cholera toxin β subunit (CTβ ; magenta) the retrograde tracer is seen in AVP neurons of the SCN, supporting the anatomical evidence that AVP neuronal processes directly contact the CSF. Arginine vasopressin (AVP) expressing neurons form the major population in the brain’s circadian clock located in the hypothalamic suprachiasmatic nucleus (SCN). They participate in inter-neuronal coupling and provide an output signal for synchronizing daily rhythms. AVP is present at high concentrations in the cerebrospinal fluid (CSF) and fluctuates on a circadian timescale. While it is assumed that rhythms in CSF AVP are of SCN origin, a route of communication between these compartments has not been delineated. Using immunochemistry (ICC) and cell filling techniques, we determine the morphology and location of AVP neurons in mouse and delineate their axonal and dendritic processes. Cholera toxin β subunit (CTβ) tracer injected into the lateral ventricle tests whether AVP neurons communicate with CSF. Most importantly, the results indicate that AVP neurons lie in close proximity to the third ventricle, and their processes cross the ventricular wall into the CSF. We also report that contrary to widely held assumptions, AVP neurons do not fully delineate the SCN borders as PER2 expression extends beyond the AVP region. Also, AVP neurons form a rostral prong originating in the SCN medial-most and ventral-most aspect. AVP is lacking in the mid-dorsal shell but does occur at the base of the SCN just above the optic tract. Finally, neurons of the rostral SCN are smaller than those lying caudally. These findings extend our understanding of AVP signaling potential, demonstrate the heterogeneity of AVP neurons, and highlight limits in using this peptide to delineate the mouse SCN.
Collapse
|