51
|
Barranca VJ, Bhuiyan A, Sundgren M, Xing F. Functional Implications of Dale's Law in Balanced Neuronal Network Dynamics and Decision Making. Front Neurosci 2022; 16:801847. [PMID: 35295091 PMCID: PMC8919085 DOI: 10.3389/fnins.2022.801847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 02/02/2022] [Indexed: 11/28/2022] Open
Abstract
The notion that a neuron transmits the same set of neurotransmitters at all of its post-synaptic connections, typically known as Dale's law, is well supported throughout the majority of the brain and is assumed in almost all theoretical studies investigating the mechanisms for computation in neuronal networks. Dale's law has numerous functional implications in fundamental sensory processing and decision-making tasks, and it plays a key role in the current understanding of the structure-function relationship in the brain. However, since exceptions to Dale's law have been discovered for certain neurons and because other biological systems with complex network structure incorporate individual units that send both positive and negative feedback signals, we investigate the functional implications of network model dynamics that violate Dale's law by allowing each neuron to send out both excitatory and inhibitory signals to its neighbors. We show how balanced network dynamics, in which large excitatory and inhibitory inputs are dynamically adjusted such that input fluctuations produce irregular firing events, are theoretically preserved for a single population of neurons violating Dale's law. We further leverage this single-population network model in the context of two competing pools of neurons to demonstrate that effective decision-making dynamics are also produced, agreeing with experimental observations from honeybee dynamics in selecting a food source and artificial neural networks trained in optimal selection. Through direct comparison with the classical two-population balanced neuronal network, we argue that the one-population network demonstrates more robust balanced activity for systems with less computational units, such as honeybee colonies, whereas the two-population network exhibits a more rapid response to temporal variations in network inputs, as required by the brain. We expect this study will shed light on the role of neurons violating Dale's law found in experiment as well as shared design principles across biological systems that perform complex computations.
Collapse
|
52
|
Chakrabarty B, Aitchison K, White P, McCarthy CJ, Kanai AJ, Fry CH. Frequency-dependent characteristics of nerve-mediated ATP and acetylcholine release from detrusor smooth muscle. Exp Physiol 2022; 107:350-358. [PMID: 35165960 PMCID: PMC9360561 DOI: 10.1113/ep090238] [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: 11/25/2021] [Accepted: 01/28/2022] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS The frequency-dependencies of acetylcholine (ACh) and ATP co-transmitter release are different. ACh release can be modelled to a one-compartment process, whereas ATP release requires a two-compartment model. Nerve-mediated release of ACh and ATP can be independently regulated, for example by the phosphodiesterase-type 5 inhibitor, sildenafil. What is the central question of this study? Is the frequency-dependency of co-transmitter release from postganglionic nerve fibres different for each transmitter? What is the main finding and its importance? Release of co-transmitters from the parasympathetic supply to detrusor smooth muscle can be independently regulated. This offers a targeted drug model to reduce selectively the release of transmitter associated with human pathologies (ATP) and may also be applicable to other smooth muscle-based disorders of visceral tissues. ABSTRACT Nerve-mediated contractions of detrusor smooth muscle are mediated by acetylcholine (ACh) and ATP release in most animals. However, with the normal human bladder only ACh is a functional transmitter but in benign pathologies such as overactive bladder (OAB), ATP re-emerges a secondary transmitter. The selective regulation of ATP release offers a therapeutic approach to manage OAB, in contrast to current primary strategies that target ACh actions. However, the release characteristics of nerve-mediated ACh and ATP are poorly defined and this study aimed to measure the frequency-dependence of ACh and ATP release and determine if selective regulation of ATP or ACh was possible. Experiments were carried out in vitro with mouse detrusor with nerve-mediated ATP and ACh release measured, simultaneously with tension recording. ATP was released in two frequency-dependent components, both at lower frequencies (mid-range 0.4 and 5.5 Hz stimulation) compared to a single compartment release of ACh at 14 Hz. Intervention with the phosphodiesterase type-5 inhibitor, sildenafil, attenuated ATP release, equally from both components, but had no effect on ACh release. These data demonstrate that nerve-mediated ACh and ATP release characteristics are distinct and may be separately manipulated. This offers a potential targeted drug model to manage benign lower urinary tract conditions such as OAB. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Basu Chakrabarty
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK
| | - Katie Aitchison
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK
| | - Paul White
- Faculty of Engineering, Design & Mathematics, University of the West of England, Bristol, UK
| | - Carly J McCarthy
- Instituto de Investigaciones en Medicina Traslacional (IIMT), Facultad de Ciencias Biomédicas, Austral University, Buenos Aires, Argentina
| | - Anthony J Kanai
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Christopher H Fry
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK
| |
Collapse
|
53
|
Dvořáček J, Bednářová A, Krishnan N, Kodrík D. Dopaminergic muhsroom body neurons in Drosophila: flexibility of neuron identity in a model organism? Neurosci Biobehav Rev 2022; 135:104570. [DOI: 10.1016/j.neubiorev.2022.104570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/03/2022] [Accepted: 02/03/2022] [Indexed: 11/28/2022]
|
54
|
Lee CA, Romanova EV, Southey BR, Gillette R, Sweedler JV. Comparative Analysis of Neuropeptides in Homologous Interneurons and Prohormone Annotation in Nudipleuran Sea Slugs. Front Physiol 2022; 12:809529. [PMID: 35002782 PMCID: PMC8735849 DOI: 10.3389/fphys.2021.809529] [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: 11/05/2021] [Accepted: 12/02/2021] [Indexed: 02/06/2023] Open
Abstract
Despite substantial research on neuronal circuits in nudipleuran gastropods, few peptides have been implicated in nudipleuran behavior. In this study, we expanded the understanding of peptides in this clade, using three species with well-studied nervous systems, Hermissenda crassicornis, Melibe leonina, and Pleurobranchaea californica. For each species, we performed sequence homology analysis of de novo transcriptome predictions to identify homologs to 34 of 36 prohormones previously characterized in the gastropods Aplysia californica and Lymnaea stagnalis. We then used single-cell mass spectrometry to characterize peptide profiles in homologous feeding interneurons: the multifunctional ventral white cell (VWC) in P. californica and the small cardioactive peptide B large buccal (SLB) cells in H. crassicornis and M. leonina. The neurons produced overlapping, but not identical, peptide profiles. The H. crassicornis SLB cells expressed peptides from homologs to the FMRFamide (FMRFa), small cardioactive peptide (SCP), LFRFamide (LFRFa), and feeding circuit activating peptides prohormones. The M. leonina SLB cells expressed peptides from homologs to the FMRFa, SCP, LFRFa, and MIP-related peptides prohormones. The VWC, previously shown to express peptides from the FMRFa and QNFLa (a homolog of A. californica pedal peptide 4) prohormones, was shown to also contain SCP peptides. Thus, each neuron expressed peptides from the FMRFa and SCP families, the H. crassicornis and M. leonina SLB cells expressed peptides from the LFRFa family, and each neuron contained peptides from a prohormone not found in the others. These data suggest each neuron performs complex co-transmission, which potentially facilitates a multifunctional role in feeding. Additionally, the unique feeding characteristics of each species may relate, in part, to differences in the peptide profiles of these neurons. These data add chemical insight to enhance our understanding of the neuronal basis of behavior in nudipleurans and other gastropods.
Collapse
Affiliation(s)
- Colin A Lee
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, United States
| | - Elena V Romanova
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, United States.,Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, IL, United States
| | - Bruce R Southey
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, United States
| | - Rhanor Gillette
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, United States.,Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL, United States
| | - Jonathan V Sweedler
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, United States.,Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, IL, United States
| |
Collapse
|
55
|
Zhang X, Lian P, Su M, Ji Z, Deng J, Zheng G, Wang W, Ren X, Jiang T, Zhang P, Li H. Single-cell transcriptome analysis identifies a unique tumor cell type producing multiple hormones in ectopic ACTH and CRH secreting pheochromocytoma. eLife 2021; 10:68436. [PMID: 34905486 PMCID: PMC8719890 DOI: 10.7554/elife.68436] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 12/13/2021] [Indexed: 01/04/2023] Open
Abstract
Ectopic Cushing’s syndrome due to ectopic ACTH&CRH-secreting by pheochromocytoma is extremely rare and can be fatal if not properly diagnosed. It remains unclear whether a unique cell type is responsible for multiple hormones secreting. In this work, we performed single-cell RNA sequencing to three different anatomic tumor tissues and one peritumoral tissue based on a rare case with ectopic ACTH&CRH-secreting pheochromocytoma. And in addition to that, three adrenal tumor specimens from common pheochromocytoma and adrenocortical adenomas were also involved in the comparison of tumor cellular heterogeneity. A total of 16 cell types in the tumor microenvironment were identified by unbiased cell clustering of single-cell transcriptomic profiles from all specimens. Notably, we identified a novel multi-functionally chromaffin-like cell type with high expression of both POMC (the precursor of ACTH) and CRH, called ACTH+&CRH + pheochromocyte. We hypothesized that the molecular mechanism of the rare case harbor Cushing’s syndrome is due to the identified novel tumor cell type, that is, the secretion of ACTH had a direct effect on the adrenal gland to produce cortisol, while the secretion of CRH can indirectly stimulate the secretion of ACTH from the anterior pituitary. Besides, a new potential marker (GAL) co-expressed with ACTH and CRH might be involved in the regulation of ACTH secretion. The immunohistochemistry results confirmed its multi-functionally chromaffin-like properties with positive staining for CRH, POMC, ACTH, GAL, TH, and CgA. Our findings also proved to some extent the heterogeneity of endothelial and immune microenvironment in different adrenal tumor subtypes.
Collapse
Affiliation(s)
- Xuebin Zhang
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Penghu Lian
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Mingming Su
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Zhigang Ji
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jianhua Deng
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Guoyang Zheng
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wenda Wang
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xinyu Ren
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Taijiao Jiang
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China.,Suzhou Institute of Systems Medicine, Jiangsu, China
| | - Peng Zhang
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Hanzhong Li
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
56
|
Cifuentes F, Morales MA. Functional Implications of Neurotransmitter Segregation. Front Neural Circuits 2021; 15:738516. [PMID: 34720888 PMCID: PMC8548464 DOI: 10.3389/fncir.2021.738516] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/21/2021] [Indexed: 11/13/2022] Open
Abstract
Here, we present and discuss the characteristics and properties of neurotransmitter segregation, a subtype of neurotransmitter cotransmission. We review early evidence of segregation and discuss its properties, such as plasticity, while placing special emphasis on its probable functional implications, either in the central nervous system (CNS) or the autonomic nervous system. Neurotransmitter segregation is a process by which neurons separately route transmitters to independent and distant or to neighboring neuronal processes; it is a plastic phenomenon that changes according to synaptic transmission requirements and is regulated by target-derived signals. Distant neurotransmitter segregation in the CNS has been shown to be related to an autocrine/paracrine function of some neurotransmitters. In retinal amacrine cells, segregation of acetylcholine (ACh) and GABA, and glycine and glutamate to neighboring terminals has been related to the regulation of the firing rate of direction-selective ganglion cells. In the rat superior cervical ganglion, segregation of ACh and GABA to neighboring varicosities shows a heterogeneous regional distribution, which is correlated to a similar regional distribution in transmission strength. We propose that greater segregation of ACh and GABA produces less GABAergic inhibition, strengthening ganglionic transmission. Segregation of ACh and GABA varies in different physiopathological conditions; specifically, segregation increases in acute sympathetic hyperactivity that occurs in cold stress, does not vary in chronic hyperactivity that occurs in hypertension, and rises in early ages of normotensive and hypertensive rats. Given this, we propose that variations in the extent of transmitter segregation may contribute to the alteration of neural activity that occurs in some physiopathological conditions and with age.
Collapse
Affiliation(s)
- Fredy Cifuentes
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Miguel Angel Morales
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
57
|
Huang H, Possidente DR, Vecsey CG. Optogenetic activation of SIFamide (SIFa) neurons induces a complex sleep-promoting effect in the fruit fly Drosophila melanogaster. Physiol Behav 2021; 239:113507. [PMID: 34175361 DOI: 10.1016/j.physbeh.2021.113507] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 05/20/2021] [Accepted: 06/22/2021] [Indexed: 10/21/2022]
Abstract
Sleep is a universal and extremely complicated function. Sleep is regulated by two systems-sleep homeostasis and circadian rhythms. In a wide range of species, neuropeptides have been found to play a crucial role in the communication and synchronization between different components of both systems. In the fruit fly Drosophila melanogaster, SIFamide (SIFa) is a neuropeptide that has been reported to be expressed in 4 neurons in the pars intercerebralis (PI) area of the brain. Previous work has shown that transgenic ablation of SIFa neurons, mutation of SIFa itself, or knockdown of SIFa receptors reduces sleep, suggesting that SIFa is sleep-promoting. However, those were all constitutive manipulations that could have affected development or resulted in compensation, so the role of SIFa signaling in sleep regulation during adulthood remains unclear. In the current study, we examined the sleep-promoting effect of SIFa through an optogenetic approach, which allowed for neuronal activation with high temporal resolution, while leaving development unaffected. We found that activation of the red-light sensor Chrimson in SIFa neurons promoted sleep in flies in a sexually dimorphic manner, where the magnitude of the sleep effect was greater in females than in males. Because neuropeptidergic neurons often also release other transmitters, we used RNA interference to knock down SIFa while also optogenetically activating SIFa neurons. SIFa knockdown only partially reduced the magnitude of the sleep effect, suggesting that release of other transmitters may contribute to the sleep induction when SIFa neurons are activated. Video-based analysis showed that activation of SIFa neurons for as brief a period as 1 second was able to decrease walking behavior for minutes after the stimulus. Future studies should aim to identify the transmitters that are utilized by SIFa neurons and characterize their upstream activators and downstream targets. It would also be of interest to determine how acute optogenetic activation of SIFa neurons alters other behaviors that have been linked to SIFa, such as mating and feeding.
Collapse
Affiliation(s)
- Haoyang Huang
- Neuroscience Program, Skidmore College, 815 N. Broadway, Saratoga Springs, NY 12866
| | - Debra R Possidente
- Neuroscience Program, Skidmore College, 815 N. Broadway, Saratoga Springs, NY 12866
| | - Christopher G Vecsey
- Neuroscience Program, Skidmore College, 815 N. Broadway, Saratoga Springs, NY 12866.
| |
Collapse
|
58
|
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: 1.3] [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
|
59
|
Cahaya N, Wijaya SP, Anwar K. Incidence of Orthostatic Hypotension in Schizophrenic Patients Using Antipsychotics at Sambang Lihum Mental Health Hospital, South Kalimantan. BORNEO JOURNAL OF PHARMACY 2021. [DOI: 10.33084/bjop.v4i3.1959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Schizophrenia is a psychiatric disorder that requires antipsychotics therapy. Antipsychotics cause many side effects, including orthostatic hypotension. The study aimed to describe the incidence of orthostatic hypotensive side effects experiences by schizophrenia patients at the Sambang Lihum Mental Health Hospital, South Kalimantan. This research was observational description research with data sampling by medical records. This research was conducted to 300 medical records of patients period January-December 2018 which received antipsychotics medication and data analyzed by univariate analysis. The results showed the number of patients who experienced orthostatic hypotension was 98 patients (32.67%) and no experienced were 202 patients (67.33%). Incidence of orthostatic hypotension in haloperidol 54.35% (N=46); trifluoperazine 100% (N=1); clozapine 84.62% (N=13); olanzapine 100% (N=1); haloperidol-chlorpromazine 27.27% (N=11); haloperidol-haloperidol 42.86% (N=7); clozapine-risperidone 16.67% (N=6); haloperidol-clozapine 15.05% (N=93); haloperidol-olanzapine 50% (N=2); haloperidol-risperidone 31.82% (N=22); trifluoperazine-olanzapine 100% (N=1); trifluoperazine-clozapine 22.22% (N=9); trifluoperazine-risperidone 5.56% (N=18); chlorpromazine-haloperidol-haloperidol 33.3% (N=3); chlorpromazine-haloperidol-trifluoperazine 100% (N=3); haloperidol-trifluoperazine-chlorpromazine 100% (N=1); chlorpromazine-haloperidol-clozapine 42.86% (N=7); chlorpromazine-trifluoperazine-clozapine 100% (N=1); chlorpromazine-trifluoperazine-olanzapine 100% (N=1); chlorpromazine-trifluoperazine-risperidone 50% (N=2); trifluoperazine-haloperidol-risperidone 100% (N=4); haloperidol-trifluoperazine-risperidone 100% (N=1); trifluoperazine-haloperidol-clozapine 40% (N=5); haloperidol-haloperidol-clozapine 80% (N=5); clozapine-risperidone-trifluoperazine 100% (N=4); risperidone-clozapine-haloperidol 20% (N=10). The conclusion from this study was the percentage of orthostatic hypotension on schizophrenia patients at the Sambang Lihum Mental Health Hospital was 32.67% (N=98).
Collapse
|
60
|
Moroz LL. Multiple Origins of Neurons From Secretory Cells. Front Cell Dev Biol 2021; 9:669087. [PMID: 34307354 PMCID: PMC8293673 DOI: 10.3389/fcell.2021.669087] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 05/26/2021] [Indexed: 12/12/2022] Open
Affiliation(s)
- Leonid L. Moroz
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
- Whitney Laboratory for Marine Biosciences, University of Florida, St. Augustine, FL, United States
| |
Collapse
|
61
|
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: 12] [Impact Index Per Article: 3.0] [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
|
62
|
Stone TW. Relationships and Interactions between Ionotropic Glutamate Receptors and Nicotinic Receptors in the CNS. Neuroscience 2021; 468:321-365. [PMID: 34111447 DOI: 10.1016/j.neuroscience.2021.06.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 02/07/2023]
Abstract
Although ionotropic glutamate receptors and nicotinic receptors for acetylcholine (ACh) have usually been studied separately, they are often co-localized and functionally inter-dependent. The objective of this review is to survey the evidence for interactions between the two receptor families and the mechanisms underlying them. These include the mutual regulation of subunit expression, which change the NMDA:AMPA response balance, and the existence of multi-functional receptor complexes which make it difficult to distinguish between individual receptor sites, especially in vivo. This is followed by analysis of the functional relationships between the receptors from work on transmitter release, cellular electrophysiology and aspects of behavior where these can contribute to understanding receptor interactions. It is clear that nicotinic receptors (nAChRs) on axonal terminals directly regulate the release of glutamate and other neurotransmitters, α7-nAChRs generally promoting release. Hence, α7-nAChR responses will be prevented not only by a nicotinic antagonist, but also by compounds blocking the indirectly activated glutamate receptors. This accounts for the apparent anticholinergic activity of some glutamate antagonists, including the endogenous antagonist kynurenic acid. The activation of presynaptic nAChRs is by the ambient levels of ACh released from pre-terminal synapses, varicosities and glial cells, acting as a 'volume neurotransmitter' on synaptic and extrasynaptic sites. In addition, ACh and glutamate are released as CNS co-transmitters, including 'cholinergic' synapses onto spinal Renshaw cells. It is concluded that ACh should be viewed primarily as a modulator of glutamatergic neurotransmission by regulating the release of glutamate presynaptically, and the location, subunit composition, subtype balance and sensitivity of glutamate receptors, and not primarily as a classical fast neurotransmitter. These conclusions and caveats should aid clarification of the sites of action of glutamate and nicotinic receptor ligands in the search for new centrally-acting drugs.
Collapse
Affiliation(s)
- Trevor W Stone
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK; Institute of Neuroscience, University of Glasgow, G12 8QQ, UK.
| |
Collapse
|
63
|
Kitcher SR, Pederson AM, Weisz CJC. Diverse identities and sites of action of cochlear neurotransmitters. Hear Res 2021; 419:108278. [PMID: 34108087 DOI: 10.1016/j.heares.2021.108278] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 04/30/2021] [Accepted: 05/18/2021] [Indexed: 11/18/2022]
Abstract
Accurate encoding of acoustic stimuli requires temporally precise responses to sound integrated with cellular mechanisms that encode the complexity of stimuli over varying timescales and orders of magnitude of intensity. Sound in mammals is initially encoded in the cochlea, the peripheral hearing organ, which contains functionally specialized cells (including hair cells, afferent and efferent neurons, and a multitude of supporting cells) to allow faithful acoustic perception. To accomplish the demanding physiological requirements of hearing, the cochlea has developed synaptic arrangements that operate over different timescales, with varied strengths, and with the ability to adjust function in dynamic hearing conditions. Multiple neurotransmitters interact to support the precision and complexity of hearing. Here, we review the location of release, action, and function of neurotransmitters in the mammalian cochlea with an emphasis on recent work describing the complexity of signaling.
Collapse
Affiliation(s)
- Siân R Kitcher
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD 20892, United States
| | - Alia M Pederson
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD 20892, United States
| | - Catherine J C Weisz
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD 20892, United States.
| |
Collapse
|
64
|
Eskenazi D, Malave L, Mingote S, Yetnikoff L, Ztaou S, Velicu V, Rayport S, Chuhma N. Dopamine Neurons That Cotransmit Glutamate, From Synapses to Circuits to Behavior. Front Neural Circuits 2021; 15:665386. [PMID: 34093138 PMCID: PMC8170480 DOI: 10.3389/fncir.2021.665386] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/16/2021] [Indexed: 11/21/2022] Open
Abstract
Discovered just over 20 years ago, dopamine neurons have the ability to cotransmit both dopamine and glutamate. Yet, the functional roles of dopamine neuron glutamate cotransmission and their implications for therapeutic use are just emerging. This review article encompasses the current body of evidence investigating the functions of dopamine neurons of the ventral midbrain that cotransmit glutamate. Since its discovery in dopamine neuron cultures, further work in vivo confirmed dopamine neuron glutamate cotransmission across species. From there, growing interest has led to research related to neural functioning including roles in synaptic signaling, development, and behavior. Functional connectome mapping reveals robust connections in multiple forebrain regions to various cell types, most notably to cholinergic interneurons in both the medial shell of the nucleus accumbens and the lateral dorsal striatum. Glutamate markers in dopamine neurons reach peak levels during embryonic development and increase in response to various toxins, suggesting dopamine neuron glutamate cotransmission may serve neuroprotective roles. Findings from behavioral analyses reveal prominent roles for dopamine neuron glutamate cotransmission in responses to psychostimulants, in positive valence and cognitive systems and for subtle roles in negative valence systems. Insight into dopamine neuron glutamate cotransmission informs the pathophysiology of neuropsychiatric disorders such as addiction, schizophrenia and Parkinson Disease, with therapeutic implications.
Collapse
Affiliation(s)
- Daniel Eskenazi
- Department of Psychiatry, Columbia University, New York, NY, United States
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
| | - Lauren Malave
- Department of Psychiatry, Columbia University, New York, NY, United States
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
| | - Susana Mingote
- Department of Psychiatry, Columbia University, New York, NY, United States
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
- Neuroscience Initiative, Advanced Science Research Center, Graduate Center of The City University of New York, New York, NY, United States
| | - Leora Yetnikoff
- Department of Psychology, College of Staten Island, City University of New York, Staten Island, NY, United States
- CUNY Neuroscience Collaborative, The Graduate Center, City University of New York, New York, NY, United States
| | - Samira Ztaou
- Department of Psychiatry, Columbia University, New York, NY, United States
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
| | - Vlad Velicu
- Department of Psychiatry, Columbia University, New York, NY, United States
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
| | - Stephen Rayport
- Department of Psychiatry, Columbia University, New York, NY, United States
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
| | - Nao Chuhma
- Department of Psychiatry, Columbia University, New York, NY, United States
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
| |
Collapse
|
65
|
The Phosphoprotein Synapsin Ia Regulates the Kinetics of Dense-Core Vesicle Release. J Neurosci 2021; 41:2828-2841. [PMID: 33632727 DOI: 10.1523/jneurosci.2593-19.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 02/02/2021] [Accepted: 02/04/2021] [Indexed: 12/19/2022] Open
Abstract
Common fusion machinery mediates the Ca2+-dependent exocytosis of synaptic vesicles (SVs) and dense-core vesicles (DCVs). Previously, Synapsin Ia (Syn Ia) was found to localize to SVs, essential for mobilizing SVs to the plasma membrane through phosphorylation. However, whether (or how) the phosphoprotein Syn Ia plays a role in regulating DCV exocytosis remains unknown. To answer these questions, we measured the dynamics of DCV exocytosis by using single-vesicle amperometry in PC12 cells (derived from the pheochromocytoma of rats of unknown sex) overexpressing wild-type or phosphodeficient Syn Ia. We found that overexpression of phosphodeficient Syn Ia decreased the DCV secretion rate, specifically via residues previously shown to undergo calmodulin-dependent kinase (CaMK)-mediated phosphorylation (S9, S566, and S603). Moreover, the fusion pore kinetics during DCV exocytosis were found to be differentially regulated by Syn Ia and two phosphodeficient Syn Ia mutants (Syn Ia-S62A and Syn Ia-S9,566,603A). Kinetic analysis suggested that Syn Ia may regulate the closure and dilation of DCV fusion pores via these sites, implying the potential interactions of Syn Ia with certain DCV proteins involved in the regulation of fusion pore dynamics. Furthermore, we predicted the interaction of Syn Ia with several DCV proteins, including Synaptophysin (Syp) and soluble N-ethylmaleimide-sensitive factor attachment receptor (SNARE) proteins. By immunoprecipitation, we found that Syn Ia interacted with Syp via phosphorylation. Moreover, a proximity ligation assay (PLA) confirmed their phosphorylation-dependent, in situ interaction on DCVs. Together, these findings reveal a phosphorylation-mediated regulation of DCV exocytosis by Syn Ia.SIGNIFICANCE STATEMENT Although they exhibit distinct exocytosis dynamics upon stimulation, synaptic vesicles (SVs) and dense-core vesicles (DCVs) may undergo co-release in neurons and neuroendocrine cells through an undefined molecular mechanism. Synapsin Ia (Syn Ia) is known to recruit SVs to the plasma membrane via phosphorylation. Here, we examined whether Syn Ia also affects the dynamics of DCV exocytosis. We showed that Syn Ia regulates the DCV secretion rate and fusion pore kinetics during DCV exocytosis. Moreover, Syn Ia-mediated regulation of DCV exocytosis depends on phosphorylation. We further found that Syn Ia interacts with Synaptophysin (Syp) on DCVs in a phosphorylation-dependent manner. Thus, these results suggest that Syn Ia may regulate the release of DCVs via phosphorylation.
Collapse
|
66
|
Grininger D, Birmingham JT. Dual modulatory effects on feedback from a proprioceptor in the crustacean stomatogastric nervous system. J Neurophysiol 2021; 125:1755-1767. [PMID: 33760675 DOI: 10.1152/jn.00080.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Neuromodulatory actions that change the properties of proprioceptors or the muscle movements to which they respond necessarily affect the feedback provided to the central network. Here we further characterize the responses of the gastropyloric receptor 1 (GPR1) and gastropyloric receptor 2 (GPR2) neurons in the stomatogastric nervous system of the crab Cancer borealis to movements and contractions of muscles, and we report how neuromodulation modifies those responses. We observed that the GPR1 response to contractions of the gastric mill 4 muscle (gm4) was absent, or nearly so, when the neuron was quiescent but robust when it was spontaneously active. We also found that the effects of four neuromodulatory substances (GABA, serotonin, proctolin, and TNRNFLRFamide) on the GPR1 response to muscle stretch were similar to those previously reported for GPR2. Finally, we showed that an excitatory action on gm4 due to proctolin combined with an inhibitory action on GPR2 due to GABA can allow for larger muscle contractions without increased proprioceptive feedback.NEW & NOTEWORTHY We report that the combination of GABA and the peptide proctolin increases contraction of a stomatogastric muscle while decreasing the corresponding response of the proprioceptor that reports on it. These results suggest a general mechanism by which muscle movements can be modified while sensory feedback is conserved, one that may be particularly well suited for providing flexibility to central pattern generator networks.
Collapse
Affiliation(s)
- Davis Grininger
- Department of Physics, Santa Clara University, Santa Clara, California
| | - John T Birmingham
- Department of Physics, Santa Clara University, Santa Clara, California
| |
Collapse
|
67
|
Guillaumin MCC, Burdakov D. Neuropeptides as Primary Mediators of Brain Circuit Connectivity. Front Neurosci 2021; 15:644313. [PMID: 33776641 PMCID: PMC7991401 DOI: 10.3389/fnins.2021.644313] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 02/18/2021] [Indexed: 11/21/2022] Open
Abstract
Across sleep and wakefulness, brain function requires inter-neuronal interactions lasting beyond seconds. Yet, most studies of neural circuit connectivity focus on millisecond-scale interactions mediated by the classic fast transmitters, GABA and glutamate. In contrast, neural circuit roles of the largest transmitter family in the brain–the slow-acting peptide transmitters–remain relatively overlooked, or described as “modulatory.” Neuropeptides may efficiently implement sustained neural circuit connectivity, since they are not rapidly removed from the extracellular space, and their prolonged action does not require continuous presynaptic firing. From this perspective, we review actions of evolutionarily-conserved neuropeptides made by brain-wide-projecting hypothalamic neurons, focusing on lateral hypothalamus (LH) neuropeptides essential for stable consciousness: the orexins/hypocretins. Action potential-dependent orexin release inside and outside the hypothalamus evokes slow postsynaptic excitation. This excitation does not arise from modulation of classic neurotransmission, but involves direct action of orexins on their specific G-protein coupled receptors (GPCRs) coupled to ion channels. While millisecond-scale, GABA/glutamate connectivity within the LH may not be strong, re-assessing LH microcircuits from the peptidergic viewpoint is consistent with slow local microcircuits. The sustained actions of neuropeptides on neuronal membrane potential may enable core brain functions, such as temporal integration and the creation of lasting permissive signals that act as “eligibility traces” for context-dependent information routing and plasticity. The slowness of neuropeptides has unique advantages for efficient neuronal processing and feedback control of consciousness.
Collapse
Affiliation(s)
| | - Denis Burdakov
- Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland
| |
Collapse
|
68
|
Nässel DR. Leucokinin and Associated Neuropeptides Regulate Multiple Aspects of Physiology and Behavior in Drosophila. Int J Mol Sci 2021; 22:1940. [PMID: 33669286 PMCID: PMC7920058 DOI: 10.3390/ijms22041940] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 12/17/2022] Open
Abstract
Leucokinins (LKs) constitute a family of neuropeptides identified in numerous insects and many other invertebrates. LKs act on G-protein-coupled receptors that display only distant relations to other known receptors. In adult Drosophila, 26 neurons/neurosecretory cells of three main types express LK. The four brain interneurons are of two types, and these are implicated in several important functions in the fly's behavior and physiology, including feeding, sleep-metabolism interactions, state-dependent memory formation, as well as modulation of gustatory sensitivity and nociception. The 22 neurosecretory cells (abdominal LK neurons, ABLKs) of the abdominal neuromeres co-express LK and a diuretic hormone (DH44), and together, these regulate water and ion homeostasis and associated stress as well as food intake. In Drosophila larvae, LK neurons modulate locomotion, escape responses and aspects of ecdysis behavior. A set of lateral neurosecretory cells, ALKs (anterior LK neurons), in the brain express LK in larvae, but inconsistently so in adults. These ALKs co-express three other neuropeptides and regulate water and ion homeostasis, feeding, and drinking, but the specific role of LK is not yet known. This review summarizes Drosophila data on embryonic lineages of LK neurons, functional roles of individual LK neuron types, interactions with other peptidergic systems, and orchestrating functions of LK.
Collapse
Affiliation(s)
- Dick R Nässel
- Department of Zoology, Stockholm University, S-10691 Stockholm, Sweden
| |
Collapse
|
69
|
Nässel DR, Wu SF. Leucokinins: Multifunctional Neuropeptides and Hormones in Insects and Other Invertebrates. Int J Mol Sci 2021; 22:1531. [PMID: 33546414 PMCID: PMC7913504 DOI: 10.3390/ijms22041531] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 01/28/2021] [Accepted: 01/28/2021] [Indexed: 12/27/2022] Open
Abstract
Leucokinins (LKs) constitute a neuropeptide family first discovered in a cockroach and later identified in numerous insects and several other invertebrates. The LK receptors are only distantly related to other known receptors. Among insects, there are many examples of species where genes encoding LKs and their receptors are absent. Furthermore, genomics has revealed that LK signaling is lacking in several of the invertebrate phyla and in vertebrates. In insects, the number and complexity of LK-expressing neurons vary, from the simple pattern in the Drosophila larva where the entire CNS has 20 neurons of 3 main types, to cockroaches with about 250 neurons of many different types. Common to all studied insects is the presence or 1-3 pairs of LK-expressing neurosecretory cells in each abdominal neuromere of the ventral nerve cord, that, at least in some insects, regulate secretion in Malpighian tubules. This review summarizes the diverse functional roles of LK signaling in insects, as well as other arthropods and mollusks. These functions include regulation of ion and water homeostasis, feeding, sleep-metabolism interactions, state-dependent memory formation, as well as modulation of gustatory sensitivity and nociception. Other functions are implied by the neuronal distribution of LK, but remain to be investigated.
Collapse
Affiliation(s)
- Dick R. Nässel
- Department of Zoology, Stockholm University, S-10691 Stockholm, Sweden
| | - Shun-Fan Wu
- College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China;
| |
Collapse
|
70
|
Rainey AN, Fukui SM, Mark K, King HM, Blitz DM. Intrinsic sources of tachykinin-related peptide in the thoracic ganglion mass of the crab, Cancer borealis. Gen Comp Endocrinol 2021; 302:113688. [PMID: 33275935 DOI: 10.1016/j.ygcen.2020.113688] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/27/2020] [Accepted: 11/28/2020] [Indexed: 11/30/2022]
Abstract
Neuropeptides comprise the largest class of neural and neuroendocrine signaling molecules. Vertebrate tachykinins (TKs) and the structurally-related invertebrate tachykinin-related peptides (TRPs) together form the largest neuropeptide superfamily, with a number of conserved neural and neuroendocrine functions across species. Arthropods, including crustaceans, have provided many insights into neuropeptide signaling and function. Crustacean tachykinin-related peptide occurs in endocrine organs and cells and in two of the major crustacean CNS components, the supraoesophageal ganglion ("brain") and the stomatogastric nervous system. However, little is known about TRP sources in the remaining major CNS component, the thoracic ganglion mass (TGM). To gain further insight into the function of this peptide, we aimed to identify intrinsic TRP sources in the TGM of the Jonah crab, Cancer borealis. We first adapted a clearing protocol to improve TRP immunoreactivity specifically in the TGM, which is a dense, fused mass of multiple ganglia in short-bodied crustaceans such as Cancer species of crabs. We verified that the clearing protocol avoided distortion of cell body morphology yet increased visibility of TRP immunoreactivity. Using confocal microscopy, we found TRP-immunoreactive (TRP-IR) axon tracts running the length of the TGM, TRP-IR neuropil in all ganglia, and approximately 110 TRP-IR somata distributed throughout the TGM, within and between ganglia. These somata likely represent both neural and neuroendocrine sources of TRP. Thus, there are many potential intrinsic sources of TRP in the TGM that are positioned to regulate behaviors such as food intake, locomotion, respiration, and reproduction.
Collapse
Affiliation(s)
- Amanda N Rainey
- Department of Biology and Center for Neuroscience, Miami University, Oxford, OH 45056, United States
| | - Stephanie M Fukui
- Department of Biology and Center for Neuroscience, Miami University, Oxford, OH 45056, United States
| | - Katie Mark
- Department of Biology and Center for Neuroscience, Miami University, Oxford, OH 45056, United States
| | - Hailey M King
- Department of Biology and Center for Neuroscience, Miami University, Oxford, OH 45056, United States
| | - Dawn M Blitz
- Department of Biology and Center for Neuroscience, Miami University, Oxford, OH 45056, United States.
| |
Collapse
|
71
|
Nässel DR, Zandawala M. Hormonal axes in Drosophila: regulation of hormone release and multiplicity of actions. Cell Tissue Res 2020; 382:233-266. [PMID: 32827072 PMCID: PMC7584566 DOI: 10.1007/s00441-020-03264-z] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/20/2020] [Indexed: 12/16/2022]
Abstract
Hormones regulate development, as well as many vital processes in the daily life of an animal. Many of these hormones are peptides that act at a higher hierarchical level in the animal with roles as organizers that globally orchestrate metabolism, physiology and behavior. Peptide hormones can act on multiple peripheral targets and simultaneously convey basal states, such as metabolic status and sleep-awake or arousal across many central neuronal circuits. Thereby, they coordinate responses to changing internal and external environments. The activity of neurosecretory cells is controlled either by (1) cell autonomous sensors, or (2) by other neurons that relay signals from sensors in peripheral tissues and (3) by feedback from target cells. Thus, a hormonal signaling axis commonly comprises several components. In mammals and other vertebrates, several hormonal axes are known, such as the hypothalamic-pituitary-gonad axis or the hypothalamic-pituitary-thyroid axis that regulate reproduction and metabolism, respectively. It has been proposed that the basic organization of such hormonal axes is evolutionarily old and that cellular homologs of the hypothalamic-pituitary system can be found for instance in insects. To obtain an appreciation of the similarities between insect and vertebrate neurosecretory axes, we review the organization of neurosecretory cell systems in Drosophila. Our review outlines the major peptidergic hormonal pathways known in Drosophila and presents a set of schemes of hormonal axes and orchestrating peptidergic systems. The detailed organization of the larval and adult Drosophila neurosecretory systems displays only very basic similarities to those in other arthropods and vertebrates.
Collapse
Affiliation(s)
- Dick R. Nässel
- Department of Zoology, Stockholm University, Stockholm, Sweden
| | - Meet Zandawala
- Department of Neuroscience, Brown University, Providence, RI USA
| |
Collapse
|
72
|
Colton GF, Cook AP, Nusbaum MP. Different microcircuit responses to comparable input from one versus both copies of an identified projection neuron. J Exp Biol 2020; 223:jeb228114. [PMID: 32820029 PMCID: PMC7648612 DOI: 10.1242/jeb.228114] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 08/13/2020] [Indexed: 12/19/2022]
Abstract
Neuronal inputs to microcircuits are often present as multiple copies of apparently equivalent neurons. Thus far, however, little is known regarding the relative influence on microcircuit output of activating all or only some copies of such an input. We examine this issue in the crab (Cancer borealis) stomatogastric ganglion, where the gastric mill (chewing) microcircuit is activated by modulatory commissural neuron 1 (MCN1), a bilaterally paired modulatory projection neuron. Both MCN1s contain the same co-transmitters, influence the same gastric mill microcircuit neurons, can drive the biphasic gastric mill rhythm, and are co-activated by all identified MCN1-activating pathways. Here, we determine whether the gastric mill microcircuit response is equivalent when stimulating one or both MCN1s under conditions where the pair are matched to collectively fire at the same overall rate and pattern as single MCN1 stimulation. The dual MCN1 stimulations elicited more consistently coordinated rhythms, and these rhythms exhibited longer phases and cycle periods. These different outcomes from single and dual MCN1 stimulation may have resulted from the relatively modest, and equivalent, firing rate of the gastric mill neuron LG (lateral gastric) during each matched set of stimulations. The LG neuron-mediated, ionotropic inhibition of the MCN1 axon terminals is the trigger for the transition from the retraction to protraction phase. This LG neuron influence on MCN1 was more effective during the dual stimulations, where each MCN1 firing rate was half that occurring during the matched single stimulations. Thus, equivalent individual- and co-activation of a class of modulatory projection neurons does not necessarily drive equivalent microcircuit output.
Collapse
Affiliation(s)
- Gabriel F Colton
- Department of Neuroscience, 211 Clinical Research Building, 415 Curie Boulevard, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aaron P Cook
- Department of Neuroscience, 211 Clinical Research Building, 415 Curie Boulevard, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael P Nusbaum
- Department of Neuroscience, 211 Clinical Research Building, 415 Curie Boulevard, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
73
|
Di Virgilio F, Jacobson KA, Williams M. Geoffrey Burnstock - An accidental pharmacologist. Biochem Pharmacol 2020; 187:114300. [PMID: 33203518 DOI: 10.1016/j.bcp.2020.114300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 11/29/2022]
Abstract
Geoffrey Burnstock, the founder of the field of purinergic signaling research passed away in Melbourne, Australia on June 3rd, 2020, at the age of 91. With his death, the world of biomedical research lost one of its most passionate, creative and unconventional thought leaders. He was an inspiration to the many researchers he interacted with for more than 50 years and a frequent irritation to those in the administrative establishment. Geoff never considered himself a pharmacologist having being trained as a zoologist and becoming an autonomic neurophysiologist based on his evolving interests in systems and disease-related research. By the end of his life he had: published some 1550 papers; been cited more than 125,000 times; had an h-index of 156 and had supervised over 100 Ph.D. students. His indelible legacy, based on a holistic, data-based, multidisciplinary, unconventional "outside the box" approach to research was reflected in two of the seminal findings in late 20th century biomedical research: the purinergic neurotransmitter hypothesis and the concept of co-neurotransmission, both of which were initially received by his peers with considerable skepticism that at times verged on disdain. Nonetheless, while raising hackles and threatening the status quo, Geoff persevered and prevailed, becoming a mentor for several generations of biomedical researchers. In this review we provide a joint perspective on Geoff Burnstock's legacy in research.
Collapse
Affiliation(s)
| | - Kenneth A Jacobson
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, Bethesda, MD, United States
| | - Michael Williams
- Department of Biological Chemistry and Pharmacology, College of Medicine, Ohio State University, Columbus, OH, United States.
| |
Collapse
|
74
|
Duhart JM, Herrero A, de la Cruz G, Ispizua JI, Pírez N, Ceriani MF. Circadian Structural Plasticity Drives Remodeling of E Cell Output. Curr Biol 2020; 30:5040-5048.e5. [PMID: 33065014 DOI: 10.1016/j.cub.2020.09.057] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/21/2020] [Accepted: 09/17/2020] [Indexed: 12/15/2022]
Abstract
Behavioral outputs arise as a result of highly regulated yet flexible communication among neurons. The Drosophila circadian network includes 150 neurons that dictate the temporal organization of locomotor activity; under light-dark (LD) conditions, flies display a robust bimodal pattern. The pigment-dispersing factor (PDF)-positive small ventral lateral neurons (sLNv) have been linked to the generation of the morning activity peak (the "M cells"), whereas the Cryptochrome (CRY)-positive dorsal lateral neurons (LNds) and the PDF-negative sLNv are necessary for the evening activity peak (the "E cells") [1, 2]. While each group directly controls locomotor output pathways [3], an interplay between them along with a third dorsal cluster (the DN1ps) is necessary for the correct timing of each peak and for adjusting behavior to changes in the environment [4-7]. M cells set the phase of roughly half of the circadian neurons (including the E cells) through PDF [5, 8-10]. Here, we show the existence of synaptic input provided by the evening oscillator onto the M cells. Both structural and functional approaches revealed that E-to-M cell connectivity changes across the day, with higher excitatory input taking place before the day-to-night transition. We identified two different neurotransmitters, acetylcholine and glutamate, released by E cells that are relevant for robust circadian output. Indeed, we show that acetylcholine is responsible for the excitatory input from E cells to M cells, which show preferential responsiveness to acetylcholine during the evening. Our findings provide evidence of an excitatory feedback between circadian clusters and unveil an important plastic remodeling of the E cells' synaptic connections.
Collapse
Affiliation(s)
- José M Duhart
- Laboratorio de Genética del Comportamiento, Fundación Instituto Leloir, IIBBA-CONICET, Av. Patricias Argentinas 435, Buenos Aires 1405-BWE, Argentina
| | - Anastasia Herrero
- Laboratorio de Genética del Comportamiento, Fundación Instituto Leloir, IIBBA-CONICET, Av. Patricias Argentinas 435, Buenos Aires 1405-BWE, Argentina
| | - Gabriel de la Cruz
- Laboratorio de Genética del Comportamiento, Fundación Instituto Leloir, IIBBA-CONICET, Av. Patricias Argentinas 435, Buenos Aires 1405-BWE, Argentina
| | - Juan I Ispizua
- Laboratorio de Genética del Comportamiento, Fundación Instituto Leloir, IIBBA-CONICET, Av. Patricias Argentinas 435, Buenos Aires 1405-BWE, Argentina
| | - Nicolás Pírez
- Laboratorio de Genética del Comportamiento, Fundación Instituto Leloir, IIBBA-CONICET, Av. Patricias Argentinas 435, Buenos Aires 1405-BWE, Argentina
| | - M Fernanda Ceriani
- Laboratorio de Genética del Comportamiento, Fundación Instituto Leloir, IIBBA-CONICET, Av. Patricias Argentinas 435, Buenos Aires 1405-BWE, Argentina.
| |
Collapse
|
75
|
Modulatory Roles of ATP and Adenosine in Cholinergic Neuromuscular Transmission. Int J Mol Sci 2020; 21:ijms21176423. [PMID: 32899290 PMCID: PMC7503321 DOI: 10.3390/ijms21176423] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/29/2020] [Accepted: 09/01/2020] [Indexed: 12/23/2022] Open
Abstract
A review of the data on the modulatory action of adenosine 5’-triphosphate (ATP), the main co-transmitter with acetylcholine, and adenosine, the final ATP metabolite in the synaptic cleft, on neuromuscular transmission is presented. The effects of these endogenous modulators on pre- and post-synaptic processes are discussed. The contribution of purines to the processes of quantal and non-quantal secretion of acetylcholine into the synaptic cleft, as well as the influence of the postsynaptic effects of ATP and adenosine on the functioning of cholinergic receptors, are evaluated. As usual, the P2-receptor-mediated influence is minimal under physiological conditions, but it becomes very important in some pathophysiological situations such as hypothermia, stress, or ischemia. There are some data demonstrating the same in neuromuscular transmission. It is suggested that the role of endogenous purines is primarily to provide a safety factor for the efficiency of cholinergic neuromuscular transmission.
Collapse
|
76
|
Parajuli LK, Wako K, Maruo S, Kakuta S, Koike M. Unique synaptic topography of crest-type synapses in the interpeduncular nucleus. Biochem Biophys Res Commun 2020; 530:130-135. [PMID: 32828274 DOI: 10.1016/j.bbrc.2020.06.046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 06/10/2020] [Indexed: 12/21/2022]
Abstract
Neurons in the central nervous system display a great diversity of synaptic architecture. While much of our knowledge on the excitatory synapse morphology derives from the prototypical asymmetric synapses, little has been studied about the atypical crest-type synapse that exists in the restricted brain regions. Here, we used focused ion beam scanning electron microscopy (FIB/SEM) to image a neuropil volume of interpeduncular nucleus (IPN) and manually reconstructed several dendrites to obtain an insight about the topography and quantitative features of crest synapses. Three-dimensional reconstruction showed numerous U-shaped structures protruding from the IPN dendrites. On either faces of the U-shaped structure, a pair of crest synapses are aligned in parallel such that there exists a positive correlation between the postsynaptic density (PSD) area of synapses that participate in pair formation. Interestingly, mitochondria are excluded from the site of crest synapses. Several presynaptic axons run through the hollow, cylindrical space of the U-shape grooves such that the plasma membrane of the axon and the dendrite are organized in a tight opposition without any intervening glial membrane. Unlike the peculiar dendritic morphology, IPN neurons possess typical somatic morphology with an oval, centrally located nucleus. In conclusion, our data reveals a hitherto unknown unique topographical feature of crest synapses in the IPN.
Collapse
Affiliation(s)
- Laxmi Kumar Parajuli
- Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan.
| | - Ken Wako
- Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Suiki Maruo
- Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Soichiro Kakuta
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan; Laboratory of Morphology and Image Analysis, Research Support Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Masato Koike
- Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan; Advanced Research Institute for Health Science, Juntendo University, Tokyo, 113-8421, Japan.
| |
Collapse
|
77
|
Smith SJ, Hawrylycz M, Rossier J, Sümbül U. New light on cortical neuropeptides and synaptic network plasticity. Curr Opin Neurobiol 2020; 63:176-188. [PMID: 32679509 DOI: 10.1016/j.conb.2020.04.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 04/11/2020] [Accepted: 04/13/2020] [Indexed: 01/14/2023]
Abstract
Neuropeptides, members of a large and evolutionarily ancient family of proteinaceous cell-cell signaling molecules, are widely recognized as extremely potent regulators of brain function and behavior. At the cellular level, neuropeptides are known to act mainly via modulation of ion channel and synapse function, but functional impacts emerging at the level of complex cortical synaptic networks have resisted mechanistic analysis. New findings from single-cell RNA-seq transcriptomics now illuminate intricate patterns of cortical neuropeptide signaling gene expression and new tools now offer powerful molecular access to cortical neuropeptide signaling. Here we highlight some of these new findings and tools, focusing especially on prospects for experimental and theoretical exploration of peptidergic and synaptic networks interactions underlying cortical function and plasticity.
Collapse
Affiliation(s)
- Stephen J Smith
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA, USA.
| | - Michael Hawrylycz
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA, USA
| | - Jean Rossier
- Neuroscience Paris Seine, Sorbonne Université, Paris, France
| | - Uygar Sümbül
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA, USA
| |
Collapse
|
78
|
Role of the Serotonin Receptor 7 in Brain Plasticity: From Development to Disease. Int J Mol Sci 2020; 21:ijms21020505. [PMID: 31941109 PMCID: PMC7013427 DOI: 10.3390/ijms21020505] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/08/2020] [Accepted: 01/10/2020] [Indexed: 12/18/2022] Open
Abstract
Our knowledge on the plastic functions of the serotonin (5-HT) receptor subtype 7 (5-HT7R) in the brain physiology and pathology have advanced considerably in recent years. A wealth of data show that 5-HT7R is a key player in the establishment and remodeling of neuronal cytoarchitecture during development and in the mature brain, and its dysfunction is linked to neuropsychiatric and neurodevelopmental diseases. The involvement of this receptor in synaptic plasticity is further demonstrated by data showing that its activation allows the rescue of long-term potentiation (LTP) and long-term depression (LTD) deficits in various animal models of neurodevelopmental diseases. In addition, it is becoming clear that the 5-HT7R is involved in inflammatory intestinal diseases, modulates the function of immune cells, and is likely to play a role in the gut-brain axis. In this review, we will mainly focus on recent findings on this receptor’s role in the structural and synaptic plasticity of the mammalian brain, although we will also illustrate novel aspects highlighted in gastrointestinal (GI) tract and immune system.
Collapse
|
79
|
Presynaptic MAST kinase controls opposing postsynaptic responses to convey stimulus valence in Caenorhabditis elegans. Proc Natl Acad Sci U S A 2020; 117:1638-1647. [PMID: 31911469 PMCID: PMC6983413 DOI: 10.1073/pnas.1909240117] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Animals need to quickly extract the valence information of sensory stimulus and assess whether the stimulus is attractive or aversive. Deciphering the molecular and circuit mechanisms that determine the stimulus valence is fundamental to understand how the nervous system generates the animal behaviors. Here we report that the AFD thermosensory neurons of C. elegans evoke in its postsynaptic AIY interneurons opposing neuronal responses that correlate with the valence of thermal stimuli. The C. elegans homologs of MAST kinase, Stomatin, and Diacylglycerol kinase function in AFD and regulate the opposing AIY responses. Our results further suggest that the alteration between excitatory and inhibitory AIY responses is mediated by controlling the balance of two opposing signals released from the AFD neurons. Presynaptic plasticity is known to modulate the strength of synaptic transmission. However, it remains unknown whether regulation in presynaptic neurons can evoke excitatory and inhibitory postsynaptic responses. We report here that the Caenorhabditis elegans homologs of MAST kinase, Stomatin, and Diacylglycerol kinase act in a thermosensory neuron to elicit in its postsynaptic neuron an excitatory or inhibitory response that correlates with the valence of thermal stimuli. By monitoring neural activity of the valence-coding interneuron in freely behaving animals, we show that the alteration between excitatory and inhibitory responses of the interneuron is mediated by controlling the balance of two opposing signals released from the presynaptic neuron. These alternative transmissions further generate opposing behavioral outputs necessary for the navigation on thermal gradients. Our findings suggest that valence-encoding interneuronal activity is determined by a presynaptic mechanism whereby MAST kinase, Stomatin, and Diacylglycerol kinase influence presynaptic outputs.
Collapse
|
80
|
Incognito AV, Jendzjowsky NG. The organization of the sympathetic nervous system: shining new light on historic views. J Physiol 2019; 597:5327-5329. [PMID: 31535387 DOI: 10.1113/jp278898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Anthony V Incognito
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Nicholas G Jendzjowsky
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
81
|
Molecular and Cellular Mechanisms Underlying Somatostatin-Based Signaling in Two Model Neural Networks, the Retina and the Hippocampus. Int J Mol Sci 2019; 20:ijms20102506. [PMID: 31117258 PMCID: PMC6566141 DOI: 10.3390/ijms20102506] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/16/2019] [Accepted: 05/20/2019] [Indexed: 02/06/2023] Open
Abstract
Neural inhibition plays a key role in determining the specific computational tasks of different brain circuitries. This functional "braking" activity is provided by inhibitory interneurons that use different neurochemicals for signaling. One of these substances, somatostatin, is found in several neural networks, raising questions about the significance of its widespread occurrence and usage. Here, we address this issue by analyzing the somatostatinergic system in two regions of the central nervous system: the retina and the hippocampus. By comparing the available information on these structures, we identify common motifs in the action of somatostatin that may explain its involvement in such diverse circuitries. The emerging concept is that somatostatin-based signaling, through conserved molecular and cellular mechanisms, allows neural networks to operate correctly.
Collapse
|