1
|
Hevesi Z, Hökfelt T, Harkany T. Neuropeptides: The Evergreen Jack-of-All-Trades in Neuronal Circuit Development and Regulation. Bioessays 2025; 47:e202400238. [PMID: 39723681 PMCID: PMC11848124 DOI: 10.1002/bies.202400238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/09/2024] [Accepted: 12/12/2024] [Indexed: 12/28/2024]
Abstract
Neuropeptides are key modulators of adult neurocircuits, balancing their sensitivity to both excitation and inhibition, and fine-tuning fast neurotransmitter action under physiological conditions. Here, we reason that transient increases in neuropeptide availability and action exist during brain development for synapse maturation, selection, and maintenance. We discuss fundamental concepts of neuropeptide signaling at G protein-coupled receptors (GPCRs), with a particular focus on how signaling at neuropeptide GPCRs could underpin neuronal morphogenesis. We use galanin, a 29/30 amino acid-long neuropeptide, as an example for its retrograde release from the dendrites of thalamic neurons to impact the selection and wiring of sensory afferents originating at the trigeminal nucleus through galanin receptor 1 (GalR1) engagement. Thus, we suggest novel roles for neuropeptides, expressed transiently or permanently during both pre- and postnatal neuronal circuit development, with potentially life-long effects on circuit layout and ensuing behavioral operations.
Collapse
Affiliation(s)
- Zsofia Hevesi
- Department of Molecular NeurosciencesCenter for Brain ResearchMedical University of ViennaViennaAustria
| | - Tomas Hökfelt
- Department of NeuroscienceKarolinska InstitutetSolnaSweden
| | - Tibor Harkany
- Department of Molecular NeurosciencesCenter for Brain ResearchMedical University of ViennaViennaAustria
- Department of NeuroscienceKarolinska InstitutetSolnaSweden
| |
Collapse
|
2
|
Ratna DD, Francis TC. Extrinsic and intrinsic control of striatal cholinergic interneuron activity. Front Mol Neurosci 2025; 18:1528419. [PMID: 40018010 PMCID: PMC11865219 DOI: 10.3389/fnmol.2025.1528419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 01/31/2025] [Indexed: 03/01/2025] Open
Abstract
The striatum is an integrated component of the basal ganglia responsible for associative learning and response. Besides the presence of the most abundant γ-aminobutyric acid (GABA-ergic) medium spiny neurons (MSNs), the striatum also contains distributed populations of cholinergic interneurons (ChIs), which bidirectionally communicate with many of these neuronal subtypes. Despite their sparse distribution, ChIs provide the largest source of acetylcholine (ACh) to striatal cells, have a prominent level of arborization and activity, and are potent modulators of striatal output and play prominent roles in plasticity underlying associative learning and reinforcement. Deviations from this tonic activity, including phasic bursts or pauses caused by region-selective excitatory input, neuromodulator, or neuropeptide release can exert strong influences on intrinsic activity and synaptic plasticity via diverse receptor signaling. Recent studies and new tools have allowed improved identification of factors driving or suppressing cholinergic activity, including peptides. This review aims to outline our current understanding of factors that control tonic and phasic ChI activity, specifically focusing on how neuromodulators and neuropeptides interact to facilitate or suppress phasic ChI responses underlying learning and plasticity.
Collapse
Affiliation(s)
| | - Tanner Chase Francis
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
3
|
Nässel DR. A brief history of insect neuropeptide and peptide hormone research. Cell Tissue Res 2025; 399:129-159. [PMID: 39653844 PMCID: PMC11787221 DOI: 10.1007/s00441-024-03936-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 11/14/2024] [Indexed: 02/02/2025]
Abstract
This review briefly summarizes 50 years of research on insect neuropeptide and peptide hormone (collectively abbreviated NPH) signaling, starting with the sequencing of proctolin in 1975. The first 25 years, before the sequencing of the Drosophila genome, were characterized by efforts to identify novel NPHs by biochemical means, mapping of their distribution in neurons, neurosecretory cells, and endocrine cells of the intestine. Functional studies of NPHs were predominantly dealing with hormonal aspects of peptides and many employed ex vivo assays. With the annotation of the Drosophila genome, and more specifically of the NPHs and their receptors in Drosophila and other insects, a new era followed. This started with matching of NPH ligands to orphan receptors, and studies to localize NPHs with improved detection methods. Important advances were made with introduction of a rich repertoire of innovative molecular genetic approaches to localize and interfere with expression or function of NPHs and their receptors. These methods enabled cell- or circuit-specific interference with NPH signaling for in vivo assays to determine roles in behavior and physiology, imaging of neuronal activity, and analysis of connectivity in peptidergic circuits. Recent years have seen a dramatic increase in reports on the multiple functions of NPHs in development, physiology and behavior. Importantly, we can now appreciate the pleiotropic functions of NPHs, as well as the functional peptidergic "networks" where state dependent NPH signaling ensures behavioral plasticity and systemic homeostasis.
Collapse
Affiliation(s)
- Dick R Nässel
- Department of Zoology, Stockholm University, S-10691, Stockholm, Sweden.
| |
Collapse
|
4
|
Kalkan ÖF, Şahin Z, Aktaş O, Yildirim A, Aydin Abidin S, Özyaşar AF, Uzun İ, Abidin İ. Central administration of p234, kisspeptin antagonist, but not kisspeptin-10, reduces the power of epileptiform activity and slow EEG waves in male rats. Neurol Res 2025; 47:139-146. [PMID: 39838653 DOI: 10.1080/01616412.2025.2456293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 01/12/2025] [Indexed: 01/23/2025]
Abstract
INTRODUCTION We aimed to investigate the effects of central kisspeptin-10 and p234 administration on basal brain activity and epilepsy-like conditions induced by 4-aminopyridine (4-AP), as well as their roles in the electrocorticogram (ECoG) power spectrum and EEG waves. METHODS Thirty-five male Wistar rats were divided into five groups: sham,4-AP (2.5 mg/kg i.p.), kisspeptin-10 post-treatment (200 pmoli.c.v.), p234 post-treatment (1 nmol i.c.v.), and p234 pre-treatment (1 nmol i.c.v.). We performed 70 minutes of recordings (10 min baseline) for all groups under ketamine/xylazine (90/10 mg/kg) anesthesia. In the post-treatment groups, kisspeptin-10 or p234 injections were administered 20 minutes after epilepsy induction. In the pre-treatment group, p234 was injected after baseline recordings. Following a 20-minute pre-treatment period, 4-AP was administered. RESULTS 4-AP alone induced epileptiform activity in all animals, reaching apeak after 30 minutes. Neither kisspeptin-10 nor p234 post-treatment affected 4-AP-induced epileptiform activity (p > 0.05). However, p234 pre-treatment reduced epileptiform activity (p < 0.05). Additionally, kisspeptin-10 did not alter the spectral analysis of the EEG bands or the power of the ECoG (p > 0.05). In contrast, p234 reduced the power of the ECoG and the slow bands (delta and theta) (p < 0.05). CONCLUSION We conclude that p234 pre-treatment has an inhibitory effect on neuronal excitability and epileptiform activity in the neocortex.
Collapse
Affiliation(s)
- Ömer Faruk Kalkan
- Faculty of Medicine, Department of Physiology, Karadeniz Technical University, Trabzon, Turkey
| | - Zafer Şahin
- Faculty of Medicine, Department of Physiology, Karadeniz Technical University, Trabzon, Turkey
| | - Osman Aktaş
- Faculty of Medicine, Department of Physiology, Karadeniz Technical University, Trabzon, Turkey
| | - Abdulhamit Yildirim
- Maçka Vocational School, Laboratory and Veterinary Assistance Services, Karadeniz Technical University, Trabzon, Turkey
| | - Selcen Aydin Abidin
- Faculty of Medicine, Department of Biophysics, Karadeniz Technical University, Trabzon, Turkey
| | - Ali Faruk Özyaşar
- Faculty of Medicine, Department of Anatomy, Karadeniz Technical University, Trabzon, Turkey
| | - İbrahim Uzun
- Maçka Vocational School, Laboratory and Veterinary Assistance Services, Karadeniz Technical University, Trabzon, Turkey
| | - İsmail Abidin
- Faculty of Medicine, Department of Biophysics, Karadeniz Technical University, Trabzon, Turkey
| |
Collapse
|
5
|
Xia X, Li Y. A high-performance GRAB sensor reveals differences in the dynamics and molecular regulation between neuropeptide and neurotransmitter release. Nat Commun 2025; 16:819. [PMID: 39827209 PMCID: PMC11743212 DOI: 10.1038/s41467-025-56129-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025] Open
Abstract
The co-existence and co-transmission of neuropeptides and small molecule neurotransmitters within individual neuron represent a fundamental characteristic observed across various species. However, the differences regarding their in vivo spatiotemporal dynamics and underlying molecular regulation remain poorly understood. Here, we develop a GPCR-activation-based (GRAB) sensor for detecting short neuropeptide F (sNPF) with high sensitivity and spatiotemporal resolution. Furthermore, we investigate the in vivo dynamics and molecular regulation differences between sNPF and acetylcholine (ACh) from the same neurons. Interestingly, our findings reveal distinct spatiotemporal dynamics in the release of sNPF and ACh. Notably, our results indicate that distinct synaptotagmins (Syt) are involved in these two processes, as Syt7 and Sytα for sNPF release, while Syt1 for ACh release. Thus, this high-performance GRAB sensor provides a robust tool for studying neuropeptide release and shedding insights into the unique release dynamics and molecular regulation that distinguish neuropeptides from small molecule neurotransmitters.
Collapse
Affiliation(s)
- Xiju Xia
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
- Academy for Advanced Interdisciplinary Studies (AAIS), and Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program (PTN), Peking University, Beijing, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China.
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China.
- Academy for Advanced Interdisciplinary Studies (AAIS), and Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program (PTN), Peking University, Beijing, China.
- Chinese Institute for Brain Research, Beijing, China.
| |
Collapse
|
6
|
Gilani M, Abak N, Saberian M. Genetic-epigenetic-neuropeptide associations in mood and anxiety disorders: Toward personalized medicine. Pharmacol Biochem Behav 2024; 245:173897. [PMID: 39424200 DOI: 10.1016/j.pbb.2024.173897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/29/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
Mood and anxiety disorders are complex psychiatric conditions shaped by the multifactorial interplay of genetic, epigenetic, and neuropeptide factors. This review aims to elucidate the intricate interactions among these factors and their potential in advancing personalized medicine. We examine the genetic underpinnings, emphasizing key heritability studies and specific gene associations. The role of epigenetics is discussed, focusing on how environmental factors can modify gene expression and contribute to these disorders. Neuropeptides, including substance P, CRF, AVP, NPY, galanin, and kisspeptin, are evaluated for their involvement in mood regulation and their potential as therapeutic targets. Additionally, we address the emerging role of the gut microbiome in modulating neuropeptide activity and its connection to mood disorders. This review integrates findings from genetic, epigenetic, and neuropeptide research, offering a comprehensive overview of their collective impact on mood and anxiety disorders. By highlighting novel insights and potential clinical applications, we underscore the importance of a multi-omics approach in developing personalized treatment strategies. Future research directions are proposed to address existing knowledge gaps and translate these findings into clinical practice. Our review provides a fresh perspective on the pathophysiology of mood and anxiety disorders, paving the way for more effective and individualized therapies.
Collapse
Affiliation(s)
- Maryam Gilani
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Niloofar Abak
- Department of Hematology, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Mostafa Saberian
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Farmer GE, Cunningham JT. Spontaneous and evoked angiotensin II sniffer cell activity in the lamina terminalis in vitro. Am J Physiol Regul Integr Comp Physiol 2024; 327:R486-R496. [PMID: 39133776 PMCID: PMC11563642 DOI: 10.1152/ajpregu.00227.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 07/23/2024] [Accepted: 08/08/2024] [Indexed: 10/11/2024]
Abstract
Angiotensin II (ANG II) has been shown to have central nervous system effects. Although tissue renin-angiotensin systems (RAS) have been demonstrated in multiple tissues, the existence of a brain RAS is still a matter of debate. These studies test for angiotensin release from brain slices prepared from adult male Sprague-Dawley rats and male and female renin knock-out rats using Chinese hamster ovary cells modified to express both the angiotensin II type 1 receptor and a fluorescent calcium indicator. Sniffer cells were placed on the slices and calcium transients were measured from those located on or adjacent to the median preoptic nucleus with and without stimulation of the subfornical organ. Bath application of tetrodotoxin (1 µM) significantly attenuated spontaneous events while abolishing evoked sniffer cell activity. Bath application of dl-AP4 (10 µM, glutamatergic antagonist) did not affect either spontaneous or evoked release. Incubating the slices with fluorocitrate to inactive astrocytes did not influence sniffer cell activity in the MnPO. Pharmacological experiments indicate that ANG II release is largely both renin (aliskiren 10 µM) and ACE-1 (captopril 100 µM) dependent. However, experiments with brain slices prepared from male and female Renin knock-out rats suggest that alternative synthetic pathways may exist. Finally, these studies demonstrate that increases in ANG II release are observed following 7 days of chronic intermittent hypoxia. These studies suggest the existence of a tissue-specific RAS in the brain that involves canonical and alternative ANG II synthetic pathways and is upregulated in an animal model of sleep apnea.NEW & NOTEWORTHY These studies used Chinese hamster ovary cells that were cloned to express an angiotensin receptor (At1ra) and a calcium indicator (R-GECO) to detect the release of angiotensin from brain slices containing the lamina terminalis of rats. Some of the experiments use tissue from renin knockout rats. The results support the existence of an angiotensin system in the brain that may involve alternative synthetic pathways and is upregulated by intermittent hypoxia.
Collapse
Affiliation(s)
- George E Farmer
- Department of Physiology and Anatomy, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, United States
| | - J Thomas Cunningham
- Department of Physiology and Anatomy, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, United States
| |
Collapse
|
8
|
Samandari‐Bahraseman MR, Esmaeilzadeh‐Salestani K, Dogani M, Khaleghdoust B, Hatami N, Esmaeili‐Mahani S, Elyasi L, Loit E, Harro J. Antidepressant- and Anxiolytic-Like Effect of the Froriepia subpinnata Extract in the Rat: Neurochemical Correlates. Brain Behav 2024; 14:e70171. [PMID: 39607287 PMCID: PMC11603432 DOI: 10.1002/brb3.70171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 09/16/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND The study aims to explore the potential antianxiety effect of Froriepia subpinnata, a native plant in northern Iran, and it is considered an antiflatulent, appetizing, antiseptic, antispasmodic, and diuretic. Despite its widespread use in diets and its reputation for calming effects, no prior research has specifically investigated its antianxiety properties. METHODS Rats were subjected to a variety of stressors for 24 days. Rats were treated with the F. subpinnata extract (100, 200, and 400 mg/kg, orally) for 14 days starting from the 10th day of stress. Then behavioral tests (elevated plus-maze, open field, sucrose preference, Morris water maze, passive avoidance) were examined. Real-time PCR was used to investigate changes in the expression of candidate genes of stress response and memory. Oxidative stress markers and corticosterone levels in serum were also measured. RESULTS Chronic stress reduced performance in a variety of tests of anxiety and memory, and treatment with the F. subpinnata extract dose-dependently improved the behavioral deficits caused by chronic stress. At the dose of 200 mg/kg, the F. subpinnata extract mitigated the effect of stress on the expression of several genes, such as those encoding dopamine D1 and D2 receptors, glutamate NMDA, and AMPA receptor subunits (Grin1 and Gria1, respectively), glucocorticoid and mineralocorticoid receptors, cholecystokinin (CCK) and CCKB receptor, neuropeptide Y, and the GABAA receptor alpha2 subunit. Also, the expression of two genes, TrkB and BDNF, was significantly affected by the extract, demonstrating meaningful decreasing changes. Furthermore, treatment with the extract led to a decrease in oxidative stress and an elevation in cortisol levels in stressed animals. CONCLUSION In this study, we provide the first evidence of the antistress and antianxiety effects of F. subpinnata extract, along with its potential procognitive impact on memory.
Collapse
Affiliation(s)
- Mohammad R. Samandari‐Bahraseman
- Department of Biology, Faculty of SciencesShahid Bahonar University of KermanKermanIran
- Varjavand Kesht Kariman, Limited Liability CompanyKermanIran
| | - Keyvan Esmaeilzadeh‐Salestani
- Chair of Crop Science and Plant Biology, Institute of Agricultural and Environmental SciencesEstonian University of Life SciencesTartuEstonia
- Institute of TechnologyUniversity of TartuTartuEstonia
| | - Manijeh Dogani
- Department of Biology, Faculty of SciencesShahid Bahonar University of KermanKermanIran
| | - Banafsheh Khaleghdoust
- Chair of Crop Science and Plant Biology, Institute of Agricultural and Environmental SciencesEstonian University of Life SciencesTartuEstonia
| | - Nima Hatami
- Department of Endodontic DentistryKerman University of Medical SciencesKermanIran
| | - Saeed Esmaeili‐Mahani
- Department of Biology, Faculty of SciencesShahid Bahonar University of KermanKermanIran
| | - Leila Elyasi
- Neuroscience Research Center, Department of Anatomy, Faculty of MedicineGolestan University of Medical ScienceGorganIran
| | - Evelin Loit
- Chair of Crop Science and Plant Biology, Institute of Agricultural and Environmental SciencesEstonian University of Life SciencesTartuEstonia
| | - Jaanus Harro
- Division of Neuropsychopharmacology, Institute of ChemistryUniversity of TartuTartuEstonia
| |
Collapse
|
9
|
Dong C, Gowrishankar R, Jin Y, He XJ, Gupta A, Wang H, Sayar-Atasoy N, Flores RJ, Mahe K, Tjahjono N, Liang R, Marley A, Or Mizuno G, Lo DK, Sun Q, Whistler JL, Li B, Gomes I, Von Zastrow M, Tejeda HA, Atasoy D, Devi LA, Bruchas MR, Banghart MR, Tian L. Unlocking opioid neuropeptide dynamics with genetically encoded biosensors. Nat Neurosci 2024; 27:1844-1857. [PMID: 39009835 PMCID: PMC11374718 DOI: 10.1038/s41593-024-01697-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/05/2024] [Indexed: 07/17/2024]
Abstract
Neuropeptides are ubiquitous in the nervous system. Research into neuropeptides has been limited by a lack of experimental tools that allow for the precise dissection of their complex and diverse dynamics in a circuit-specific manner. Opioid peptides modulate pain, reward and aversion and as such have high clinical relevance. To illuminate the spatiotemporal dynamics of endogenous opioid signaling in the brain, we developed a class of genetically encoded fluorescence sensors based on kappa, delta and mu opioid receptors: κLight, δLight and µLight, respectively. We characterized the pharmacological profiles of these sensors in mammalian cells and in dissociated neurons. We used κLight to identify electrical stimulation parameters that trigger endogenous opioid release and the spatiotemporal scale of dynorphin volume transmission in brain slices. Using in vivo fiber photometry in mice, we demonstrated the utility of these sensors in detecting optogenetically driven opioid release and observed differential opioid release dynamics in response to fearful and rewarding conditions.
Collapse
Affiliation(s)
- Chunyang Dong
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Davis, CA, USA
| | - Raajaram Gowrishankar
- Center for the Neurobiology of Addiction, Pain, and Emotion, Departments of Anesthesiology and Pharmacology, University of Washington, Seattle, WA, USA
| | - Yihan Jin
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Davis, CA, USA
| | - Xinyi Jenny He
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Achla Gupta
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Huikun Wang
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Nilüfer Sayar-Atasoy
- Department of Neuroscience and Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Rodolfo J Flores
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Karan Mahe
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Davis, CA, USA
| | - Nikki Tjahjono
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Davis, CA, USA
| | - Ruqiang Liang
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Davis, CA, USA
| | - Aaron Marley
- Department of Pharmacology, University of California San Francisco, San Francisco, CA, USA
| | - Grace Or Mizuno
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Davis, CA, USA
| | - Darren K Lo
- College of Biological Sciences, University of California Davis, Davis, CA, USA
| | - Qingtao Sun
- Cold Spring Harbor Laboratory, New York, NY, USA
| | | | - Bo Li
- Cold Spring Harbor Laboratory, New York, NY, USA
| | - Ivone Gomes
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Mark Von Zastrow
- Department of Pharmacology, University of California San Francisco, San Francisco, CA, USA
| | - Hugo A Tejeda
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Deniz Atasoy
- Department of Neuroscience and Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Lakshmi A Devi
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Michael R Bruchas
- Center for the Neurobiology of Addiction, Pain, and Emotion, Departments of Anesthesiology and Pharmacology, University of Washington, Seattle, WA, USA.
| | - Matthew R Banghart
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA.
| | - Lin Tian
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Davis, CA, USA.
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA.
| |
Collapse
|
10
|
Messanvi F, Visocky V, Senneca C, Berkun K, Taori M, Bradley SP, Wang H, Tejeda H, Chudasama Y. Galanin receptor 1 expressing neurons in hippocampal-prefrontal circuitry modulate goal directed attention and impulse control. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.29.605653. [PMID: 39131306 PMCID: PMC11312591 DOI: 10.1101/2024.07.29.605653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
While amino acid neurotransmitters are the main chemical messengers in the brain, they are co-expressed with neuropeptides which are increasingly recognized as modulators of cognitive pathways. For example, the neuropeptide galanin has been implicated in a wide range of pathological conditions in which frontal and temporal structures are compromised. In a recent study in rats, we discovered that direct pharmacological stimulation of galanin receptor type 1 (GalR1) in the ventral prefrontal cortex (vPFC) and ventral hippocampus (vHC) led to opposing effects on attention and impulse control behavior. In the present study, we investigate how subtypes of neurons expressing GalR1 in these two areas differentially contribute to these behaviors. We first establish that GalR1 is predominantly expressed in glutamatergic neurons in both the vPFC and HC. We develop a novel viral approach to gain genetic access to GalR1-expressing neurons and demonstrate that optogenetic excitation of GalR1 expressing neurons in the vPFC, but not vHC, selectively disrupts attention in a complex behavioral task. Finally, using fiber photometry, we measure the bulk calcium dynamics in GalR1-expressing neurons during the same task to demonstrate opposing activity in vPFC and vHC. These results are consistent with our previous work demonstrating differential behavioral effects induced by GalR1 activating in vPFC and vHC. These results indicate the distinct neuromodulatory and behavioral contributions of galanin mediated by subclasses of neurons in the hippocampal and prefrontal circuitry.
Collapse
Affiliation(s)
- Fany Messanvi
- Section on Behavioral Neuroscience, National Institute of Mental Health, Bethesda, MD, USA
| | - Vladimir Visocky
- Section on Behavioral Neuroscience, National Institute of Mental Health, Bethesda, MD, USA
| | - Carolyn Senneca
- Section on Behavioral Neuroscience, National Institute of Mental Health, Bethesda, MD, USA
| | - Kathleen Berkun
- Section on Behavioral Neuroscience, National Institute of Mental Health, Bethesda, MD, USA
| | - Maansi Taori
- Section on Behavioral Neuroscience, National Institute of Mental Health, Bethesda, MD, USA
| | - Sean P Bradley
- Rodent Behavioral Core, National Institute of Mental Health, Bethesda, MD, USA
| | - Huikun Wang
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, Bethesda, MD, USA
| | - Hugo Tejeda
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, Bethesda, MD, USA
| | - Yogita Chudasama
- Section on Behavioral Neuroscience, National Institute of Mental Health, Bethesda, MD, USA
- Rodent Behavioral Core, National Institute of Mental Health, Bethesda, MD, USA
| |
Collapse
|
11
|
Seitz J, Lahaye E, Andreani NA, Thomas B, Takhlidjt S, Chartrel N, Herpertz-Dahlmann B, Baines JF, Fetissov SO. Long-Term Dynamics of Serum α-MSH and α-MSH-Binding Immunoglobulins with a Link to Gut Microbiota Composition in Patients with Anorexia Nervosa. Neuroendocrinology 2024; 114:907-920. [PMID: 38852579 PMCID: PMC11460951 DOI: 10.1159/000539316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 05/06/2024] [Indexed: 06/11/2024]
Abstract
INTRODUCTION Immunoglobulins (Ig) reactive with α-melanocyte-stimulating hormone (α-MSH), an anorexigenic neuropeptide, are present in humans and were previously associated with eating disorders. In this longitudinal study involving patients with anorexia nervosa (AN), we determined whether α-MSH in serum is bound to IgG and analyzed long-term dynamics of both α-MSH peptide and α-MSH-reactive Ig in relation to changes in BMI and gut microbiota composition. METHODS The study included 64 adolescents with a restrictive form of AN, whose serum samples were collected at hospital admission, discharge, and during a 1-year follow-up visit and 41 healthy controls, all females. RESULTS We found that in both study groups, approximately 40% of serum α-MSH was reversibly bound to IgG and that levels of α-MSH-reactive IgG but not of α-MSH peptide in patients with AN were low at hospital admission but recovered 1 year later. Total IgG levels were also low at admission. Moreover, BMI-standard deviation score correlated positively with α-MSH IgG in both groups studied but negatively with α-MSH peptide only in controls. Significant correlations between the abundance of specific bacterial taxa in the gut microbiota and α-MSH peptide and IgG levels were found in both study groups, but they were more frequent in controls. CONCLUSION We conclude that IgG in the blood plays a role as an α-MSH-binding protein, whose characteristics are associated with BMI in both patients with AN and controls. Furthermore, the study suggests that low production of α-MSH-reactive IgG during the starvation phase in patients with AN may be related to altered gut microbiota composition.
Collapse
Affiliation(s)
- Jochen Seitz
- Department of Child and Adolescent Psychiatry, Psychotherapy and Psychosomatics, University Hospital, RWTH University Aachen, Aachen, Germany
- Department of Child and Adolescent Psychiatry, Psychotherapy and Psychosomatics, LVR-University Hospital, Essen, Germany
| | - Emilie Lahaye
- Regulatory Peptides – Energy Metabolism and Motivated Behavior Team, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Inserm UMR1239, University of Rouen Normandie, Rouen, France
| | - Nadia Andrea Andreani
- Guest Group Evolutionary Medicine, Max Planck Institute for Evolutionary Biology, Plön, Germany
- Section of Evolutionary Medicine, Institute for Experimental Medicine, Kiel University, Kiel, Germany
| | - Benjamin Thomas
- Regulatory Peptides – Energy Metabolism and Motivated Behavior Team, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Inserm UMR1239, University of Rouen Normandie, Rouen, France
| | - Saloua Takhlidjt
- Regulatory Peptides – Energy Metabolism and Motivated Behavior Team, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Inserm UMR1239, University of Rouen Normandie, Rouen, France
| | - Nicolas Chartrel
- Regulatory Peptides – Energy Metabolism and Motivated Behavior Team, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Inserm UMR1239, University of Rouen Normandie, Rouen, France
| | - Beate Herpertz-Dahlmann
- Department of Child and Adolescent Psychiatry, Psychotherapy and Psychosomatics, University Hospital, RWTH University Aachen, Aachen, Germany
| | - John F. Baines
- Guest Group Evolutionary Medicine, Max Planck Institute for Evolutionary Biology, Plön, Germany
- Section of Evolutionary Medicine, Institute for Experimental Medicine, Kiel University, Kiel, Germany
| | - Sergueï O. Fetissov
- Regulatory Peptides – Energy Metabolism and Motivated Behavior Team, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Inserm UMR1239, University of Rouen Normandie, Rouen, France
| |
Collapse
|
12
|
Xia X, Li Y. A new GRAB sensor reveals differences in the dynamics and molecular regulation between neuropeptide and neurotransmitter release. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.22.595424. [PMID: 38826473 PMCID: PMC11142204 DOI: 10.1101/2024.05.22.595424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
The co-existence and co-transmission of neuropeptides and small molecule neurotransmitters in the same neuron is a fundamental aspect of almost all neurons across various species. However, the differences regarding their in vivo spatiotemporal dynamics and underlying molecular regulation remain poorly understood. Here, we developed a GPCR-activation-based (GRAB) sensor for detecting short neuropeptide F (sNPF) with high sensitivity and spatiotemporal resolution. Furthermore, we explore the differences of in vivo dynamics and molecular regulation between sNPF and acetylcholine (ACh) from the same neurons. Interestingly, the release of sNPF and ACh shows different spatiotemporal dynamics. Notably, we found that distinct synaptotagmins (Syt) are involved in these two processes, as Syt7 and Sytα for sNPF release, while Syt1 for ACh release. Thus, this new GRAB sensor provides a powerful tool for studying neuropeptide release and providing new insights into the distinct release dynamics and molecular regulation between neuropeptides and small molecule neurotransmitters.
Collapse
Affiliation(s)
- Xiju Xia
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
- Academy for Advanced Interdisciplinary Studies (AAIS), and Peking University–Tsinghua University–National Institute of Biological Sciences Joint Graduate Program (PTN), Peking University, Beijing, 100871, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
- Academy for Advanced Interdisciplinary Studies (AAIS), and Peking University–Tsinghua University–National Institute of Biological Sciences Joint Graduate Program (PTN), Peking University, Beijing, 100871, China
- Chinese Institute for Brain Research, Beijing 102206, China
| |
Collapse
|
13
|
Agoston DV. Of artificial intelligence, machine learning, and the human brain. Celebrating Miklos Palkovits' 90th birthday. Front Neuroanat 2024; 18:1374864. [PMID: 38764486 PMCID: PMC11099251 DOI: 10.3389/fnana.2024.1374864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 03/25/2024] [Indexed: 05/21/2024] Open
Affiliation(s)
- Denes V. Agoston
- Department of Anatomy, Physiology and Genetics, Uniformed Services University, Bethesda, MD, United States
| |
Collapse
|
14
|
Murdock MH, Yang CY, Sun N, Pao PC, Blanco-Duque C, Kahn MC, Kim T, Lavoie NS, Victor MB, Islam MR, Galiana F, Leary N, Wang S, Bubnys A, Ma E, Akay LA, Sneve M, Qian Y, Lai C, McCarthy MM, Kopell N, Kellis M, Piatkevich KD, Boyden ES, Tsai LH. Multisensory gamma stimulation promotes glymphatic clearance of amyloid. Nature 2024; 627:149-156. [PMID: 38418876 PMCID: PMC10917684 DOI: 10.1038/s41586-024-07132-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 01/25/2024] [Indexed: 03/02/2024]
Abstract
The glymphatic movement of fluid through the brain removes metabolic waste1-4. Noninvasive 40 Hz stimulation promotes 40 Hz neural activity in multiple brain regions and attenuates pathology in mouse models of Alzheimer's disease5-8. Here we show that multisensory gamma stimulation promotes the influx of cerebrospinal fluid and the efflux of interstitial fluid in the cortex of the 5XFAD mouse model of Alzheimer's disease. Influx of cerebrospinal fluid was associated with increased aquaporin-4 polarization along astrocytic endfeet and dilated meningeal lymphatic vessels. Inhibiting glymphatic clearance abolished the removal of amyloid by multisensory 40 Hz stimulation. Using chemogenetic manipulation and a genetically encoded sensor for neuropeptide signalling, we found that vasoactive intestinal peptide interneurons facilitate glymphatic clearance by regulating arterial pulsatility. Our findings establish novel mechanisms that recruit the glymphatic system to remove brain amyloid.
Collapse
Affiliation(s)
- Mitchell H Murdock
- Department of Brain and Cognitive Sciences and the Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Cheng-Yi Yang
- Department of Brain and Cognitive Sciences and the Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Na Sun
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ping-Chieh Pao
- Department of Brain and Cognitive Sciences and the Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Cristina Blanco-Duque
- Department of Brain and Cognitive Sciences and the Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Martin C Kahn
- Department of Brain and Cognitive Sciences and the Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - TaeHyun Kim
- Department of Brain and Cognitive Sciences and the Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nicolas S Lavoie
- Department of Brain and Cognitive Sciences and the Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Matheus B Victor
- Department of Brain and Cognitive Sciences and the Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Md Rezaul Islam
- Department of Brain and Cognitive Sciences and the Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Fabiola Galiana
- Department of Brain and Cognitive Sciences and the Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Noelle Leary
- Department of Brain and Cognitive Sciences and the Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sidney Wang
- Department of Brain and Cognitive Sciences and the Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Adele Bubnys
- Department of Brain and Cognitive Sciences and the Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Emily Ma
- Department of Brain and Cognitive Sciences and the Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Leyla A Akay
- Department of Brain and Cognitive Sciences and the Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Madison Sneve
- Departments of Biological Engineering and Brain and Cognitive Sciences, McGovern Institute, Cambridge, MA, USA
- Koch Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yong Qian
- Departments of Biological Engineering and Brain and Cognitive Sciences, McGovern Institute, Cambridge, MA, USA
- Koch Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Cuixin Lai
- School of Life Sciences, Westlake University, Westlake Laboratory of Life Sciences and Biomedicine, and Westlake Institute for Advanced Study, Hangzhou, China
| | - Michelle M McCarthy
- Department of Mathematics and Statistics, Boston University, Boston, MA, USA
| | - Nancy Kopell
- Department of Mathematics and Statistics, Boston University, Boston, MA, USA
| | - Manolis Kellis
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kiryl D Piatkevich
- Departments of Biological Engineering and Brain and Cognitive Sciences, McGovern Institute, Cambridge, MA, USA
- Koch Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
- School of Life Sciences, Westlake University, Westlake Laboratory of Life Sciences and Biomedicine, and Westlake Institute for Advanced Study, Hangzhou, China
| | - Edward S Boyden
- Departments of Biological Engineering and Brain and Cognitive Sciences, McGovern Institute, Cambridge, MA, USA
- Koch Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Li-Huei Tsai
- Department of Brain and Cognitive Sciences and the Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
15
|
Cerri DH, Albaugh DL, Walton LR, Katz B, Wang TW, Chao THH, Zhang W, Nonneman RJ, Jiang J, Lee SH, Etkin A, Hall CN, Stuber GD, Shih YYI. Distinct neurochemical influences on fMRI response polarity in the striatum. Nat Commun 2024; 15:1916. [PMID: 38429266 PMCID: PMC10907631 DOI: 10.1038/s41467-024-46088-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 02/13/2024] [Indexed: 03/03/2024] Open
Abstract
The striatum, known as the input nucleus of the basal ganglia, is extensively studied for its diverse behavioral roles. However, the relationship between its neuronal and vascular activity, vital for interpreting functional magnetic resonance imaging (fMRI) signals, has not received comprehensive examination within the striatum. Here, we demonstrate that optogenetic stimulation of dorsal striatal neurons or their afferents from various cortical and subcortical regions induces negative striatal fMRI responses in rats, manifesting as vasoconstriction. These responses occur even with heightened striatal neuronal activity, confirmed by electrophysiology and fiber-photometry. In parallel, midbrain dopaminergic neuron optogenetic modulation, coupled with electrochemical measurements, establishes a link between striatal vasodilation and dopamine release. Intriguingly, in vivo intra-striatal pharmacological manipulations during optogenetic stimulation highlight a critical role of opioidergic signaling in generating striatal vasoconstriction. This observation is substantiated by detecting striatal vasoconstriction in brain slices after synthetic opioid application. In humans, manipulations aimed at increasing striatal neuronal activity likewise elicit negative striatal fMRI responses. Our results emphasize the necessity of considering vasoactive neurotransmission alongside neuronal activity when interpreting fMRI signal.
Collapse
Affiliation(s)
- Domenic H Cerri
- Center for Animal MRI, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Daniel L Albaugh
- Center for Animal MRI, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Lindsay R Walton
- Center for Animal MRI, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Brittany Katz
- Center for Animal MRI, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Tzu-Wen Wang
- Center for Animal MRI, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Tzu-Hao Harry Chao
- Center for Animal MRI, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Weiting Zhang
- Center for Animal MRI, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Randal J Nonneman
- Center for Animal MRI, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jing Jiang
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Sung-Ho Lee
- Center for Animal MRI, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Amit Etkin
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Alto Neuroscience, Los Altos, CA, USA
| | - Catherine N Hall
- Sussex Neuroscience, University of Sussex, Falmer, United Kingdom
- School of Psychology, University of Sussex, Falmer, United Kingdom
| | - Garret D Stuber
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
- Department of Pharmacology, University of Washington, Seattle, WA, USA
| | - Yen-Yu Ian Shih
- Center for Animal MRI, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Biomedical Research Imaging Center, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Neurology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
16
|
Abramova O, Zorkina Y, Pavlov K, Ushakova V, Morozova A, Zubkov E, Pavlova O, Storozheva Z, Gurina O, Chekhonin V. Chronic Ultrasound Prenatal Stress Altered the Brain's Neurochemical Systems in Newborn Rats. Neural Plast 2024; 2024:3829941. [PMID: 39290524 PMCID: PMC11407898 DOI: 10.1155/2024/3829941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 12/25/2023] [Accepted: 01/27/2024] [Indexed: 09/19/2024] Open
Abstract
Prenatal stress (PS) affects the development and functioning of the central nervous system, but the exact mechanisms underpinning this effect have not been pinpointed yet. A promising model of PS is one based on chronic exposure of pregnant rodents to variable-frequency ultrasound (US PS), as it mimics the PS with a psychic nature that most adequately captures the human stressors in modern society. The aim of this study was to investigate the effects of US PS on the brain neurotransmitter, neuropeptide, and neurotrophic systems of newborn Wistar rats. We determined the concentration of neurotransmitters and their metabolites (serotonin, HIAA, dopamine, DOPAC, and norepinephrine), neuropeptides (α-MSH, β-endorphin, neurotensin, oxytocin, and substance P), and the neurotrophin brain-derived neurotrophic factor (BDNF) in rat brain tissues by HPLC-ED, ELISA, and multiplex ELISA. Correlation analysis and principal component analysis (PCA) were used to get a sense of the relationship between the biochemical parameters of the brain. The results demonstrated that US PS increases the concentration of serotonin (p=0.004) and DOPAC (p=0.04) in the hippocampus has no effect on the neurotransmitter systems of the frontal cortex, reduces the concentration of BDNF in the entirety of the brain of males (p=0.008), and increases the neuropeptides α-MSH (p=0.02), β-endorphin (p=0.01), oxytocin (p=0.008), and substance P (p < 0.001) in the entire brain. A degree of complexity in the neurotransmitter system network in the frontal cortex and network change in the hippocampus after exposure to US PS have been observed. PCA revealed a similar pattern of neurotransmitter system interactions in the frontal cortex and hippocampus in males and females after exposure to US PS. We suggest that US PS can alter neurodevelopment, which is mediated by changes in the studied neurochemical systems that thus affect the behavioral phenotype in animals.
Collapse
Affiliation(s)
- Olga Abramova
- Department of Basic and Applied Neurobiology, V. Serbsky National Medical Research Centre of Psychiatry and Narcology, Moscow, Russia
| | - Yana Zorkina
- Department of Basic and Applied Neurobiology, V. Serbsky National Medical Research Centre of Psychiatry and Narcology, Moscow, Russia
| | - Konstantin Pavlov
- Department of Basic and Applied Neurobiology, V. Serbsky National Medical Research Centre of Psychiatry and Narcology, Moscow, Russia
| | - Valeria Ushakova
- Department of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Anna Morozova
- Mental-Health Clinic No. 1 Named After N.A. Alekseev, Zagorodnoe Highway 2, Moscow 115191, Russia
| | - Eugene Zubkov
- Department of Basic and Applied Neurobiology, V. Serbsky National Medical Research Centre of Psychiatry and Narcology, Moscow, Russia
| | - Olga Pavlova
- Department of Basic and Applied Neurobiology, V. Serbsky National Medical Research Centre of Psychiatry and Narcology, Moscow, Russia
| | - Zinaida Storozheva
- Laboratory of Functional Neurochemistry, P. K. Anokhin Institute of Normal Physiology, Moscow, Russia
| | - Olga Gurina
- Department of Basic and Applied Neurobiology, V. Serbsky National Medical Research Centre of Psychiatry and Narcology, Moscow, Russia
| | - Vladimir Chekhonin
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
17
|
Barde S, Aguila J, Zhong W, Solarz A, Mei I, Prud'homme J, Palkovits M, Turecki G, Mulder J, Uhlén M, Nagy C, Mechawar N, Hedlund E, Hökfelt T. Substance P, NPY, CCK and their receptors in five brain regions in major depressive disorder with transcriptomic analysis of locus coeruleus neurons. Eur Neuropsychopharmacol 2024; 78:54-63. [PMID: 37931511 DOI: 10.1016/j.euroneuro.2023.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/11/2023] [Accepted: 09/20/2023] [Indexed: 11/08/2023]
Abstract
Major depressive disorder (MDD) is a serious disease and a burden to patients, families and society. Rodent experiments and human studies suggest that several neuropeptide systems are involved in mood regulation. The aim of this study is two-fold: (i) to monitor, with qPCR, transcript levels of the substance P/tachykinin (TAC), NPY and CCK systems in bulk samples from control and suicide subjects, targeting five postmortem brain regions including locus coeruleus (LC); and (ii) to analyse expression of neuropeptide family transcripts in LC neurons of 'normal' postmortem brains by using laser capture microdissection with Smart-Seq2 RNA sequencing. qPCR revealed distinct regional expression patterns in male and female controls with higher levels for the TAC system in the dorsal raphe nucleus and LC, versus higher transcripts levels of the NPY and CCK systems in prefrontal cortex. In suicide patients, TAC, TAC receptors and a few NPY family transcript levels were increased mainly in prefrontal cortex and LC. The second study on 'normal' noradrenergic LC neurons revealed expression of transcripts for GAL, NPY, TAC1, CCK, and TACR1 and many other peptides (e.g. Cerebellin4 and CARTPT) and receptors (e.g. Adcyap1R1 and GPR173). These data and our previous results on suicide brains indicates that the tachykinin and galanin systems may be valid targets for developing antidepressant medicines. Moreover, the perturbation of neuropeptide systems in MDD patients, and the detection of further neuropeptide and receptor transcripts in LC, shed new light on signalling in noradrenergic LC neurons and on mechanisms possibly associated with mood disorders.
Collapse
Affiliation(s)
- Swapnali Barde
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Julio Aguila
- Department of Biochemistry and Biophysics, Stockholm University, 106 91, Stockholm, Sweden; Department of Cell and Molecular Biology, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Wen Zhong
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, 11428, Sweden
| | - Anna Solarz
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Irene Mei
- Department of Biochemistry and Biophysics, Stockholm University, 106 91, Stockholm, Sweden
| | - Josee Prud'homme
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada; Department of Psychiatry, McGill University, Montréal, QC, Canada
| | - Miklos Palkovits
- The Hungarian Academy of Sciences, Budapest, Hungary and Human Brain Tissue Bank and Laboratory, Semmelweis University, H-1085, Budapest, Hungary
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada; Department of Psychiatry, McGill University, Montréal, QC, Canada
| | - Jan Mulder
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Mathias Uhlén
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, 11428, Sweden
| | - Corina Nagy
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada; Department of Psychiatry, McGill University, Montréal, QC, Canada
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada; Department of Psychiatry, McGill University, Montréal, QC, Canada
| | - Eva Hedlund
- Department of Biochemistry and Biophysics, Stockholm University, 106 91, Stockholm, Sweden; Department of Cell and Molecular Biology, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Tomas Hökfelt
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
18
|
Wen YX, Fan LY, Yang AY, Zhang YC, Xu C, Wang ZH, Xie WJ, Lu Y, Zhang XY, Zhu JN, Sun A, Li L, Zhang QP. Oxytocinergic neurons, but not oxytocin, are crucial for male penile erection. Neuropharmacology 2023; 235:109576. [PMID: 37164226 DOI: 10.1016/j.neuropharm.2023.109576] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/04/2023] [Accepted: 05/07/2023] [Indexed: 05/12/2023]
Abstract
The cumulative evidence suggests that oxytocin is involved in the male sexual behaviors. However, no significant sexual impairments were observed in oxytocin gene knock-out (KO) mice, suggesting that oxytocin is not necessary for sexual behavior in male mice. To better understand the role of oxytocin in male erection, two types of oxytocin gene KO mice were created. In the first type, the oxytocin gene was deleted in the zygote, while in the second type, the oxytocin gene was mutated in adulthood by injecting the CRISPR/Cas9 AAVs. The results showed that disrupting the oxytocin gene at either the embryonic or adult stage did not affect erection, indicating that oxytocin is not necessary for penile erection. Pharmacologically, injecting oxytocin receptor agonist Carbetocin into the VTA of the oxytocin gene KO mice still evoked penile erection. By employing the Oxt-Ires-Cre mice, we found that specifically activating oxytocinergic neurons through chemogenetics strongly induced penile erection, while inhibiting these neurons blocked the erection responses. Furthermore, ablating PVN oxytocinergic neurons abolished the male erection response. In conclusion, although the neuropeptide oxytocin is not essential for male erection, the activity of oxytocinergic neurons is required. Our results might reflect the redundancy in the central nerve system in the sense that many signals contribute to the activation of oxytocinergic neurons to evoke penile erection during sexual behaviors.
Collapse
Affiliation(s)
- Yu-Xiang Wen
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), NJU Institute of AI Biomedicine and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, Institute for Brain Sciences, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Lin-Yao Fan
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), NJU Institute of AI Biomedicine and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, Institute for Brain Sciences, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - An-Yong Yang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), NJU Institute of AI Biomedicine and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, Institute for Brain Sciences, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Yan-Chufei Zhang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), NJU Institute of AI Biomedicine and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, Institute for Brain Sciences, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Chang Xu
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), NJU Institute of AI Biomedicine and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, Institute for Brain Sciences, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Zi-Hui Wang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), NJU Institute of AI Biomedicine and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, Institute for Brain Sciences, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Wen-Jiong Xie
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), NJU Institute of AI Biomedicine and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, Institute for Brain Sciences, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Yang Lu
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), NJU Institute of AI Biomedicine and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, Institute for Brain Sciences, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Xiao-Yang Zhang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), NJU Institute of AI Biomedicine and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, Institute for Brain Sciences, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Jing-Ning Zhu
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), NJU Institute of AI Biomedicine and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, Institute for Brain Sciences, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Anyang Sun
- Laboratory of Neurodegenerative Diseases & Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, 201318, China.
| | - Liang Li
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), NJU Institute of AI Biomedicine and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, Institute for Brain Sciences, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China.
| | - Qi-Peng Zhang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), NJU Institute of AI Biomedicine and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, Institute for Brain Sciences, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China.
| |
Collapse
|
19
|
Priest MF, Freda SN, Rieth IJ, Badong D, Dumrongprechachan V, Kozorovitskiy Y. Peptidergic and functional delineation of the Edinger-Westphal nucleus. Cell Rep 2023; 42:112992. [PMID: 37594894 PMCID: PMC10512657 DOI: 10.1016/j.celrep.2023.112992] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 06/15/2023] [Accepted: 07/31/2023] [Indexed: 08/20/2023] Open
Abstract
Many neuronal populations that release fast-acting excitatory and inhibitory neurotransmitters in the brain also contain slower-acting neuropeptides. These facultative peptidergic cell types are common, but it remains uncertain whether neurons that solely release peptides exist. Our fluorescence in situ hybridization, genetically targeted electron microscopy, and electrophysiological characterization suggest that most neurons of the non-cholinergic, centrally projecting Edinger-Westphal nucleus in mice are obligately peptidergic. We further show, using anterograde projection mapping, monosynaptic retrograde tracing, angled-tip fiber photometry, and chemogenetic modulation and genetically targeted ablation in conjunction with canonical assays for anxiety, that this peptidergic population activates in response to loss of motor control and promotes anxiety responses. Together, these findings elucidate an integrative, ethologically relevant role for the Edinger-Westphal nucleus and functionally align the nucleus with the periaqueductal gray, where it resides. This work advances our understanding of peptidergic modulation of anxiety and provides a framework for future investigations of peptidergic systems.
Collapse
Affiliation(s)
- Michael F Priest
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Sara N Freda
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Isabelle J Rieth
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Deanna Badong
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Vasin Dumrongprechachan
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA
| | - Yevgenia Kozorovitskiy
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA.
| |
Collapse
|
20
|
Caramia M, Romanov RA, Syderomenos S, Hevesi Z, Zhao M, Krasniakova M, Xu ZQD, Harkany T, Hökfelt TGM. Neuronal diversity of neuropeptide signaling, including galanin, in the mouse locus coeruleus. Proc Natl Acad Sci U S A 2023; 120:e2222095120. [PMID: 37487094 PMCID: PMC10401028 DOI: 10.1073/pnas.2222095120] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 06/15/2023] [Indexed: 07/26/2023] Open
Abstract
The locus coeruleus (LC) is a small nucleus in the pons from which ascending and descending projections innervate major parts of the central nervous system. Its major transmitter is norepinephrine (NE). This system is evolutionarily conserved, including in humans, and its functions are associated with wakefulness and related to disorders, such as depression. Here, we performed single-cell ribonucleic acid-sequencing (RNA-seq) to subdivide neurons in the LC (24 clusters in total) into 3 NE, 17 glutamate, and 5 γ-aminobutyric acid (GABA) subtypes, and to chart their neuropeptide, cotransmitter, and receptor profiles. We found that NE neurons expressed at least 19 neuropeptide transcripts, notably galanin (Gal) but not Npy, and >30 neuropeptide receptors. Among the galanin receptors, Galr1 was expressed in ~19% of NE neurons, as was also confirmed by in situ hybridization. Unexpectedly, Galr1 was highly expressed in GABA neurons surrounding the NE ensemble. Patch-clamp electrophysiology and cell-type-specific Ca2+-imaging using GCaMP6s revealed that a GalR1 agonist inhibits up to ~35% of NE neurons. This effect is direct and does not rely on feed-forward GABA inhibition. Our results define a role for the galanin system in NE functions, and a conceptual framework for the action of many other peptides and their receptors.
Collapse
Affiliation(s)
- Martino Caramia
- Department of Neuroscience, Biomedicum, Karolinska Institutet, Stockholm17177, Sweden
| | - Roman A. Romanov
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna1090, Austria
| | - Spyridon Syderomenos
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna1090, Austria
| | - Zsofia Hevesi
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna1090, Austria
| | - Ming Zhao
- Department of Neuroscience, Biomedicum, Karolinska Institutet, Stockholm17177, Sweden
| | - Marharyta Krasniakova
- Department of Neuroscience, Biomedicum, Karolinska Institutet, Stockholm17177, Sweden
| | - Zhi-Qing David Xu
- Department of Neurobiology, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing100069, China
- Department of Pathology, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing100069, China
| | - Tibor Harkany
- Department of Neuroscience, Biomedicum, Karolinska Institutet, Stockholm17177, Sweden
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna1090, Austria
| | - Tomas G. M. Hökfelt
- Department of Neuroscience, Biomedicum, Karolinska Institutet, Stockholm17177, Sweden
| |
Collapse
|
21
|
Yu H, Ni P, Zhao L, Tian Y, Li M, Li X, Wei W, Wei J, Deng W, Du X, Wang Q, Guo W, Ma X, Coid J, Li T. Decreased plasma neuropeptides in first-episode schizophrenia, bipolar disorder, major depressive disorder: associations with clinical symptoms and cognitive function. Front Psychiatry 2023; 14:1180720. [PMID: 37275985 PMCID: PMC10235770 DOI: 10.3389/fpsyt.2023.1180720] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/02/2023] [Indexed: 06/07/2023] Open
Abstract
Background There is an urgent need to identify differentiating and disease-monitoring biomarkers of schizophrenia, bipolar disorders (BD), and major depressive disorders (MDD) to improve treatment and management. Methods We recruited 54 first-episode schizophrenia (FES) patients, 52 BD patients, 35 MDD patients, and 54 healthy controls from inpatient and outpatient clinics. α-Melanocyte Stimulating Hormone (α-MSH), β-endorphin, neurotensin, orexin-A, oxytocin, and substance P were investigated using quantitative multiplex assay method. Psychotic symptoms were measured using the Brief Psychiatric Rating Scale (BPRS) and Positive and Negative Syndrome Scale (PANSS), manic symptoms using the Young Mania Rating Scale (YMRS), and depressive symptoms using 17 item-Hamilton Depression Rating Scale (HAMD). We additionally measured cognitive function by using a battery of tests given to all participants. Results α-MSH, neurotensin, orexin-A, oxytocin, and substance P were decreased in the three patient groups compared with controls. Neurotensin outperformed all biomarkers in differentiating patient groups from controls. There were no significant differences for 6 neuropeptides in their ability to differentiate between the three patient groups. Higher neurotensin was associated with better executive function across the entire sample. Lower oxytocin and higher substance p were associated with more psychotic symptoms in FES and BD groups. β-endorphin was associated with early morning wakening symptom in all three patient groups. Conclusion Our research shows decreased circulating neuropeptides have the potential to differentiate severe mental illnesses from controls. These neuropeptides are promising treatment targets for improving clinical symptoms and cognitive function in FES, BD, and MDD.
Collapse
Affiliation(s)
- Hua Yu
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Peiyan Ni
- The Psychiatric Laboratory and Mental Health Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Liansheng Zhao
- The Psychiatric Laboratory and Mental Health Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yang Tian
- The Psychiatric Laboratory and Mental Health Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Mingli Li
- The Psychiatric Laboratory and Mental Health Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaojing Li
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wei Wei
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jinxue Wei
- The Psychiatric Laboratory and Mental Health Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wei Deng
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiangdong Du
- Suzhou Psychiatry Hospital, Affiliated Guangji Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qiang Wang
- The Psychiatric Laboratory and Mental Health Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wanjun Guo
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaohong Ma
- The Psychiatric Laboratory and Mental Health Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jeremy Coid
- The Psychiatric Laboratory and Mental Health Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tao Li
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
22
|
Værøy H, Takhlidjt S, Cherifi Y, Lahaye E, Chartrel N, Fetissov SO. Blood Levels of Neuropeptide 26RFa in Relation to Anxiety and Aggressive Behavior in Humans-An Exploratory Study. Brain Sci 2023; 13:brainsci13020237. [PMID: 36831780 PMCID: PMC9954400 DOI: 10.3390/brainsci13020237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/27/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
26RFa, also referred to as QRFP, is a hypothalamic neuropeptide mainly known for its role in the regulation of appetite and glucose metabolism. Its possible relevance to emotional regulation is largely unexplored. To address this, in the present exploratory study, we analyzed the plasma concentrations of 26RFa in humans characterized by different levels of anxiety and aggressive behavior. For this purpose, the study included 13 prison inmates who have committed violent crimes and 19 age-matched healthy men from the general population as controls. Anxiety, depression and aggressive behavior were evaluated in both groups using standard questionnaires. The inmate group was characterized by increased aggression and anxiety compared to the controls. We found that the mean plasma levels of 26RFa did not significantly differ between the inmates and the controls. However, several high outliers were present only in the inmate group. The plasma levels of 26RFa correlated positively with the anxiety scores in all the studied subjects and controls. After removing the high outliers in the inmate group, positive correlations of 26RFa with anxiety and a subscale of hostility in the aggression scale were also recorded in this group. No significant correlations of 26RFa with depression scores or other parameters of aggressive behavior were found. Thus, the present results did not support an involvement of 26RFa in aggressive behavior in humans but pointed to a link between this neuropeptide and anxiety. Nevertheless, considering the exploratory nature of the present study, this conclusion should be verified in a larger cohort, including the clinical degree of anxiety.
Collapse
Affiliation(s)
- Henning Værøy
- Department of Psychiatric Research, Akershus University Hospital, N-1478 Nordbyhagen, Norway
| | - Saloua Takhlidjt
- Regulatory Peptides-Energy Metabolism and Motivated Behavior Team, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Inserm UMR1239, University of Rouen Normandie, 76000 Rouen, France
| | - Yamina Cherifi
- Regulatory Peptides-Energy Metabolism and Motivated Behavior Team, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Inserm UMR1239, University of Rouen Normandie, 76000 Rouen, France
| | - Emilie Lahaye
- Regulatory Peptides-Energy Metabolism and Motivated Behavior Team, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Inserm UMR1239, University of Rouen Normandie, 76000 Rouen, France
| | - Nicolas Chartrel
- Regulatory Peptides-Energy Metabolism and Motivated Behavior Team, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Inserm UMR1239, University of Rouen Normandie, 76000 Rouen, France
| | - Serguei O. Fetissov
- Regulatory Peptides-Energy Metabolism and Motivated Behavior Team, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Inserm UMR1239, University of Rouen Normandie, 76000 Rouen, France
- Correspondence:
| |
Collapse
|
23
|
Martinez Damonte V, Pomrenze MB, Manning CE, Casper C, Wolfden AL, Malenka RC, Kauer JA. Somatodendritic Release of Cholecystokinin Potentiates GABAergic Synapses Onto Ventral Tegmental Area Dopamine Cells. Biol Psychiatry 2023; 93:197-208. [PMID: 35961792 PMCID: PMC9976994 DOI: 10.1016/j.biopsych.2022.06.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 06/01/2022] [Accepted: 06/10/2022] [Indexed: 11/02/2022]
Abstract
BACKGROUND Neuropeptides are contained in nearly every neuron in the central nervous system and can be released not only from nerve terminals but also from somatodendritic sites. Cholecystokinin (CCK), among the most abundant neuropeptides in the brain, is expressed in the majority of midbrain dopamine neurons. Despite this high expression, CCK function within the ventral tegmental area (VTA) is not well understood. METHODS We confirmed CCK expression in VTA dopamine neurons through immunohistochemistry and in situ hybridization and detected optogenetically induced CCK release using an enzyme-linked immunosorbent assay. To investigate whether CCK modulates VTA circuit activity, we used whole-cell patch clamp recordings in mouse brain slices. We infused CCK locally in vivo and tested food intake and locomotion in fasted mice. We also used in vivo fiber photometry to measure Ca2+ transients in dopamine neurons during feeding. RESULTS Here we report that VTA dopamine neurons release CCK from somatodendritic regions, where it triggers long-term potentiation of GABAergic (gamma-aminobutyric acidergic) synapses. The somatodendritic release occurs during trains of optogenetic stimuli or prolonged but modest depolarization and is dependent on synaptotagmin-7 and T-type Ca2+ channels. Depolarization-induced long-term potentiation is blocked by a CCK2 receptor antagonist and mimicked by exogenous CCK. Local infusion of CCK in vivo inhibits food consumption and decreases distance traveled in an open field test. Furthermore, intra-VTA-infused CCK reduced dopamine cell Ca2+ signals during food consumption after an overnight fast and was correlated with reduced food intake. CONCLUSIONS Our experiments introduce somatodendritic neuropeptide release as a previously unknown feedback regulator of VTA dopamine cell excitability and dopamine-related behaviors.
Collapse
|
24
|
Miranda L, Bordes J, Gasperoni S, Lopez JP. Increasing resolution in stress neurobiology: from single cells to complex group behaviors. Stress 2023; 26:2186141. [PMID: 36855966 DOI: 10.1080/10253890.2023.2186141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/02/2023] Open
Abstract
Stress can have severe psychological and physiological consequences. Thus, inappropriate regulation of the stress response is linked to the etiology of mood and anxiety disorders. The generation and implementation of preclinical animal models represent valuable tools to explore and characterize the mechanisms underlying the pathophysiology of stress-related psychiatric disorders and the development of novel pharmacological strategies. In this commentary, we discuss the strengths and limitations of state-of-the-art molecular and computational advances employed in stress neurobiology research, with a focus on the ever-increasing spatiotemporal resolution in cell biology and behavioral science. Finally, we share our perspective on future directions in the fields of preclinical and human stress research.
Collapse
Affiliation(s)
- Lucas Miranda
- Department of Statistical Genetics, Max Planck Institute of Psychiatry, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany
| | - Joeri Bordes
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
| | - Serena Gasperoni
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Juan Pablo Lopez
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
25
|
Ncube D, Tallafuss A, Serafin J, Bruckner J, Farnsworth DR, Miller AC, Eisen JS, Washbourne P. A conserved transcriptional fingerprint of multi-neurotransmitter neurons necessary for social behavior. BMC Genomics 2022; 23:675. [PMID: 36175871 PMCID: PMC9523972 DOI: 10.1186/s12864-022-08879-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 09/02/2022] [Indexed: 11/11/2022] Open
Abstract
Background An essential determinant of a neuron’s functionality is its neurotransmitter phenotype. We previously identified a defined subpopulation of cholinergic neurons required for social orienting behavior in zebrafish. Results We transcriptionally profiled these neurons and discovered that they are capable of synthesizing both acetylcholine and GABA. We also established a constellation of transcription factors and neurotransmitter markers that can be used as a “transcriptomic fingerprint” to recognize a homologous neuronal population in another vertebrate. Conclusion Our results suggest that this transcriptomic fingerprint and the cholinergic-GABAergic neuronal subtype that it defines are evolutionarily conserved. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08879-w.
Collapse
Affiliation(s)
- Denver Ncube
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA
| | - Alexandra Tallafuss
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA
| | - Jen Serafin
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA
| | - Joseph Bruckner
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA
| | - Dylan R Farnsworth
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA
| | - Adam C Miller
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA
| | - Judith S Eisen
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA
| | - Philip Washbourne
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA.
| |
Collapse
|
26
|
Dissel S, Klose MK, van Swinderen B, Cao L, Ford M, Periandri EM, Jones JD, Li Z, Shaw PJ. Sleep-promoting neurons remodel their response properties to calibrate sleep drive with environmental demands. PLoS Biol 2022; 20:e3001797. [PMID: 36173939 PMCID: PMC9521806 DOI: 10.1371/journal.pbio.3001797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 08/16/2022] [Indexed: 01/29/2023] Open
Abstract
Falling asleep at the wrong time can place an individual at risk of immediate physical harm. However, not sleeping degrades cognition and adaptive behavior. To understand how animals match sleep need with environmental demands, we used live-brain imaging to examine the physiological response properties of the dorsal fan-shaped body (dFB) following interventions that modify sleep (sleep deprivation, starvation, time-restricted feeding, memory consolidation) in Drosophila. We report that dFB neurons change their physiological response-properties to dopamine (DA) and allatostatin-A (AstA) in response to different types of waking. That is, dFB neurons are not simply passive components of a hard-wired circuit. Rather, the dFB neurons intrinsically regulate their response to the activity from upstream circuits. Finally, we show that the dFB appears to contain a memory trace of prior exposure to metabolic challenges induced by starvation or time-restricted feeding. Together, these data highlight that the sleep homeostat is plastic and suggests an underlying mechanism.
Collapse
Affiliation(s)
- Stephane Dissel
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
- * E-mail: (SD); (PJS)
| | - Markus K. Klose
- University of Pittsburgh School of Medicine, Department of Pharmacology & Chemical Biology, Pittsburgh, Pennsylvania, United States of America
| | - Bruno van Swinderen
- Queensland Brain Institute, The University of Queensland, St Lucia, Australia
| | - Lijuan Cao
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Melanie Ford
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Erica M. Periandri
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Joseph D. Jones
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
| | - Zhaoyi Li
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Paul J. Shaw
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail: (SD); (PJS)
| |
Collapse
|
27
|
Jin S, Maddern XJ, Campbell EJ, Lawrence AJ. Examining ventral subiculum and basolateral amygdala projections to the nucleus accumbens shell: Differential expression of VGLuT1, VGLuT2 and VGaT in the rat. Neurosci Lett 2022; 788:136858. [PMID: 36038028 DOI: 10.1016/j.neulet.2022.136858] [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: 07/15/2022] [Revised: 08/18/2022] [Accepted: 08/23/2022] [Indexed: 12/01/2022]
Abstract
Projections to the striatum are well-identified. For example, in the ventral striatum, two major inputs to the medial nucleus accumbens shell include the ventral subiculum and basolateral amygdala. However, the chemical phenotype(s) of these projection neurons remain unclear. In this study, we examined amygdalostriatal and corticostriatal connectivity in rats using injections of the retrograde tracer cholera toxin b into the nucleus accumbens shell. To determine the neurotransmitter identity of projection neurons, we combined retrograde tracing with RNAscope in-situ hybridization, using mRNA probes against vesicular transporters associated with glutamatergic (VGluT1 - Slc17a7, VGluT2 - Slc17a6) or GABAergic (VGaT - Slc32a1) neurotransmission. Confocal imaging was used to examine vesicular transporter mRNA expression in the ventral subiculum and basolateral amygdala inputs to the nucleus accumbens shell. Both projections contained mostly VGluT1-expressing neurons. Interestingly, almost a quarter of ventral subiculum to nucleus accumbens shell projections co-expressed VGluT1 and VGluT2 compared to a relatively small number (∼3%) that were co-expressed in basolateral amygdala to nucleus accumbens shell afferents. However, almost a quarter of basolateral amygdala to nucleus accumbens shell projections were VGaT-positive. These findings highlight the diverse proportions of glutamatergic and GABAergic afferents in two major projections to the nucleus accumbens shell and raise important questions for functional studies.
Collapse
Affiliation(s)
- Shubo Jin
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3010, Australia; Florey Department of Neuroscience and Mental Health, The University of Melbourne, Victoria 3010, Australia
| | - Xavier J Maddern
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3010, Australia; Florey Department of Neuroscience and Mental Health, The University of Melbourne, Victoria 3010, Australia
| | - Erin J Campbell
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3010, Australia; Florey Department of Neuroscience and Mental Health, The University of Melbourne, Victoria 3010, Australia.
| | - Andrew J Lawrence
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3010, Australia; Florey Department of Neuroscience and Mental Health, The University of Melbourne, Victoria 3010, Australia.
| |
Collapse
|
28
|
Eiden LE, Hernández VS, Jiang SZ, Zhang L. Neuropeptides and small-molecule amine transmitters: cooperative signaling in the nervous system. Cell Mol Life Sci 2022; 79:492. [PMID: 35997826 PMCID: PMC11072502 DOI: 10.1007/s00018-022-04451-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 06/15/2022] [Accepted: 06/23/2022] [Indexed: 12/17/2022]
Abstract
Neuropeptides are expressed in cell-specific patterns throughout mammalian brain. Neuropeptide gene expression has been useful for clustering neurons by phenotype, based on single-cell transcriptomics, and for defining specific functional circuits throughout the brain. How neuropeptides function as first messengers in inter-neuronal communication, in cooperation with classical small-molecule amine transmitters (SMATs) is a current topic of systems neurobiology. Questions include how neuropeptides and SMATs cooperate in neurotransmission at the molecular, cellular and circuit levels; whether neuropeptides and SMATs always co-exist in neurons; where neuropeptides and SMATs are stored in the neuron, released from the neuron and acting, and at which receptors, after release; and how neuropeptides affect 'classical' transmitter function, both directly upon co-release, and indirectly, via long-term regulation of gene transcription and neuronal plasticity. Here, we review an extensive body of data about the distribution of neuropeptides and their receptors, their actions after neuronal release, and their function based on pharmacological and genetic loss- and gain-of-function experiments, that addresses these questions, fundamental to understanding brain function, and development of neuropeptide-based, and potentially combinatorial peptide/SMAT-based, neurotherapeutics.
Collapse
Affiliation(s)
- Lee E Eiden
- Section On Molecular Neuroscience, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 49 Convent Drive, Room 5A38, Bethesda, MD, 20892, USA.
| | - Vito S Hernández
- Department of Physiology, School of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Sunny Z Jiang
- Section On Molecular Neuroscience, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 49 Convent Drive, Room 5A38, Bethesda, MD, 20892, USA
| | - Limei Zhang
- Department of Physiology, School of Medicine, National Autonomous University of Mexico, Mexico City, Mexico.
| |
Collapse
|
29
|
Abstract
The neuropeptide system encompasses the most diverse family of neurotransmitters, but their expression, cellular localization, and functional role in the human brain have received limited attention. Here, we study human postmortem samples from prefrontal cortex (PFC), a key brain region, and employ RNA sequencing and RNAscope methods integrated with published single-cell data. Our aim is to characterize the distribution of peptides and their receptors in 17 PFC subregions and to explore their role in chemical signaling. The results suggest that the well-established anatomical and functional heterogeneity of human PFC is also reflected in the expression pattern of the neuropeptides. Our findings support ongoing efforts from academia and pharmaceutical companies to explore the potential of neuropeptide receptors as targets for drug development. Human prefrontal cortex (hPFC) is a complex brain region involved in cognitive and emotional processes and several psychiatric disorders. Here, we present an overview of the distribution of the peptidergic systems in 17 subregions of hPFC and three reference cortices obtained by microdissection and based on RNA sequencing and RNAscope methods integrated with published single-cell transcriptomics data. We detected expression of 60 neuropeptides and 60 neuropeptide receptors in at least one of the hPFC subregions. The results reveal that the peptidergic landscape in PFC consists of closely located and functionally different subregions with unique peptide/transmitter–related profiles. Neuropeptide-rich PFC subregions were identified, encompassing regions from anterior cingulate cortex/orbitofrontal gyrus. Furthermore, marked differences in gene expression exist between different PFC regions (>5-fold; cocaine and amphetamine–regulated transcript peptide) as well as between PFC regions and reference regions, for example, for somatostatin and several receptors. We suggest that the present approach allows definition of, still hypothetical, microcircuits exemplified by glutamatergic neurons expressing a peptide cotransmitter either as an agonist (hypocretin/orexin) or antagonist (galanin). Specific neuropeptide receptors have been identified as possible targets for neuronal afferents and, interestingly, peripheral blood-borne peptide hormones (leptin, adiponectin, gastric inhibitory peptide, glucagon-like peptides, and peptide YY). Together with other recent publications, our results support the view that neuropeptide systems may play an important role in hPFC and underpin the concept that neuropeptide signaling helps stabilize circuit connectivity and fine-tune/modulate PFC functions executed during health and disease.
Collapse
|
30
|
Rosen JB, Schulkin J. Hyperexcitability: From Normal Fear to Pathological Anxiety and Trauma. Front Syst Neurosci 2022; 16:727054. [PMID: 35993088 PMCID: PMC9387392 DOI: 10.3389/fnsys.2022.727054] [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: 06/17/2021] [Accepted: 06/20/2022] [Indexed: 11/16/2022] Open
Abstract
Hyperexcitability in fear circuits is suggested to be important for development of pathological anxiety and trauma from adaptive mechanisms of fear. Hyperexcitability is proposed to be due to acquired sensitization in fear circuits that progressively becomes more severe over time causing changing symptoms in early and late pathology. We use the metaphor and mechanisms of kindling to examine gains and losses in function of one excitatory and one inhibitory neuropeptide, corticotrophin releasing factor and somatostatin, respectively, to explore this sensitization hypothesis. We suggest amygdala kindling induced hyperexcitability, hyper-inhibition and loss of inhibition provide clues to mechanisms for hyperexcitability and progressive changes in function initiated by stress and trauma.
Collapse
Affiliation(s)
- Jeffrey B. Rosen
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, United States
- *Correspondence: Jeffrey B. Rosen,
| | - Jay Schulkin
- School of Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
31
|
Cholecystokinin B receptor gene polymorphism (rs2941026) is associated with anxious personality and suicidal thoughts in a longitudinal study. Acta Neuropsychiatr 2022; 34:201-211. [PMID: 34924075 DOI: 10.1017/neu.2021.41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVES Cholecystokinin is a neuropeptide with a role in the neurobiology of adaptive behaviour that is implicated in anxiety disorders, while the underlying mechanisms currently remain insufficiently explained. The rs2941026 variation in the cholecystokinin B receptor gene has previously been associated with trait anxiety. Our aim was to investigate associations between the CCKB receptor gene polymorphism rs2941026 with anxiety, personality, depressiveness and suicidality in a longitudinal study of late adolescence and early adulthood. METHODS We used reports on trait and state anxiety, depressiveness and suicidal thoughts, as well as Affective Neuroscience Personality Scales, from the two birth cohorts of the Estonian Children Personality, Behaviour and Health Study. We measured associations between the CCKBR gene rs2941026 and anxiety-related phenotypes both longitudinally and cross-sectionally at ages 15, 18, 25 and 33. RESULTS Homozygosity for both alleles of the CCKBR rs2941026 was associated with higher trait and state anxiety in the longitudinal analysis. Cross-sectional comparisons were statistically significant at ages 18 and 25 for trait anxiety and at ages 25 and 33 for state anxiety. Higher depressiveness and suicidal thoughts were associated with the A/A genotype at age 18. Additionally, homozygosity for the A-allele was related to higher FEAR and SADNESS in the Affective Neuroscience Personality Scales. The genotype effects were more apparent in females, who displayed higher levels of negative affect overall. CONCLUSIONS CCKBR genotype is persistently associated with negative affect in adolescence and young adulthood. The association of the CCKBR rs2941026 genotype with anxiety-related phenotypes is more pronounced in females.
Collapse
|
32
|
Control of lymph node activity by direct local innervation. Trends Neurosci 2022; 45:704-712. [PMID: 35820971 DOI: 10.1016/j.tins.2022.06.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/25/2022] [Accepted: 06/15/2022] [Indexed: 11/24/2022]
Abstract
The nervous system detects environmental and internal stimuli and relays this information to immune cells via neurotransmitters and neuropeptides. This is essential to respond appropriately to immunogenic threats and to support system homeostasis. Lymph nodes (LNs) act as sentinels where adaptive immune responses are generated. They are richly innervated by peripheral sympathetic and sensory nerves, which are responsible for the local secretion of neurotransmitters by sympathetic fibers, such as norepinephrine, and neuropeptides by sensory fibers, including calcitonin gene-related peptide (CGRP) and substance P. Additionally, time-of-day-dependent oscillations in nerve activity are associated with differential immune responses, suggesting a potential role for neuroimmune interactions in coordinating immunity in a circadian fashion. Here, we discuss how LN activity is controlled by local innervation.
Collapse
|
33
|
Nässel DR, Zandawala M. Endocrine cybernetics: neuropeptides as molecular switches in behavioural decisions. Open Biol 2022; 12:220174. [PMID: 35892199 PMCID: PMC9326288 DOI: 10.1098/rsob.220174] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Plasticity in animal behaviour relies on the ability to integrate external and internal cues from the changing environment and hence modulate activity in synaptic circuits of the brain. This context-dependent neuromodulation is largely based on non-synaptic signalling with neuropeptides. Here, we describe select peptidergic systems in the Drosophila brain that act at different levels of a hierarchy to modulate behaviour and associated physiology. These systems modulate circuits in brain regions, such as the central complex and the mushroom bodies, which supervise specific behaviours. At the top level of the hierarchy there are small numbers of large peptidergic neurons that arborize widely in multiple areas of the brain to orchestrate or modulate global activity in a state and context-dependent manner. At the bottom level local peptidergic neurons provide executive neuromodulation of sensory gain and intrinsically in restricted parts of specific neuronal circuits. The orchestrating neurons receive interoceptive signals that mediate energy and sleep homeostasis, metabolic state and circadian timing, as well as external cues that affect food search, aggression or mating. Some of these cues can be triggers of conflicting behaviours such as mating versus aggression, or sleep versus feeding, and peptidergic neurons participate in circuits, enabling behaviour choices and switches.
Collapse
Affiliation(s)
- Dick R. Nässel
- Department of Zoology, Stockholm University, 10691 Stockholm, Sweden
| | - Meet Zandawala
- Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, University of Würzburg, Am Hubland Würzburg 97074, Germany
| |
Collapse
|
34
|
Piwowarczyk-Nowak A, Pałasz A, Suszka-Świtek A, Della Vecchia A, Grajoszek A, Krzystanek M, Worthington JJ. Escitalopram alters local expression of noncanonical stress-related neuropeptides in the rat brain via NPS receptor signaling. Pharmacol Rep 2022; 74:637-653. [PMID: 35653031 DOI: 10.1007/s43440-022-00374-z] [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: 02/13/2022] [Revised: 05/12/2022] [Accepted: 05/16/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Neuropeptide S (NPS) is a multifunctional regulatory factor that exhibits a potent anxiolytic activity in animal models. However, there are no reports dealing with the potential molecular relationships between the anxiolytic activity of selective serotonin reuptake inhibitors (SSRIs) and NPS signaling, especially in the context of novel stress-related neuropeptides action. The present work therefore focused on gene expression of novel stress neuropeptides in the rat brain after acute treatment with escitalopram and in combination with neuropeptide S receptor (NPSR) blockade. METHODS Studies were carried out on adult, male Sprague-Dawley rats that were divided into five groups: animals injected with saline (control) and experimental rats treated with escitalopram (at single dose 10 mg/kg daily), escitalopram and SHA-68, a selective NPSR antagonist (at a single dose of 40 mg/kg), SHA-68 alone and corresponding vehicle (solvent SHA-68) control. To measure anxiety-like behavior and locomotor activity the open field test was performed. All individuals were killed under anaesthesia and the whole brain was excised. Total mRNA was isolated from homogenized samples of the amygdala, hippocampus, hypothalamus, thalamus, cerebellum, and brainstem. Real-time PCR was used for estimation of related NPS, NPSR, neuromedin U (NMU), NMU receptor 2 (NMUR2) and nesfatin-1 precursor nucleobindin-2 (NUCB2) gene expression. RESULTS Acute escitalopram administration affects the local expression of the examined neuropeptides mRNA in a varied manner depending on brain location. An increase in NPSR and NUCB2 mRNA expression in the hypothalamus and brainstem was abolished by SHA-68 coadministration, while NMU mRNA expression was upregulated after NPSR blockade in the hippocampus and cerebellum. CONCLUSIONS The pharmacological effects of escitalopram may be connected with local NPSR-related alterations in NPS/NMU/NMUR2 and nesfatin-1 gene expression at the level of selected rat brain regions. A novel alternative mode of SSRI action can be therefore cautiously proposed.
Collapse
Affiliation(s)
- Aneta Piwowarczyk-Nowak
- Department of Anatomy, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Medyków 18, 40-752, Katowice, Poland
| | - Artur Pałasz
- Department of Histology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Medyków 18, 40-752, Katowice, Poland.
| | - Aleksandra Suszka-Świtek
- Department of Histology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, ul. Medyków 18, 40-752, Katowice, Poland
| | - Alessandra Della Vecchia
- Section of Psychiatry, Department of Clinical and Experimental Medicine, University of Pisa, 67, Via Roma, 56100, Pisa, Italy
| | - Aniela Grajoszek
- Department for Experimental Medicine, Medical University of Silesia, ul. Medyków 4, 40-752, Katowice, Poland
| | - Marek Krzystanek
- Department of Psychiatry and Psychotherapy, Faculty of Medical Sciences in Katowice, Clinic of Psychiatric Rehabilitation, Medical University of Silesia, ul. Ziolowa 45/47, 40-635, Katowice, Poland
| | - John J Worthington
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, LA1 4YQ, UK
| |
Collapse
|
35
|
Smith SJ, von Zastrow M. A Molecular Landscape of Mouse Hippocampal Neuromodulation. Front Neural Circuits 2022; 16:836930. [PMID: 35601530 PMCID: PMC9120848 DOI: 10.3389/fncir.2022.836930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/30/2022] [Indexed: 12/23/2022] Open
Abstract
Adaptive neuronal circuit function requires a continual adjustment of synaptic network parameters known as “neuromodulation.” This process is now understood to be based primarily on the binding of myriad secreted “modulatory” ligands such as dopamine, serotonin and the neuropeptides to G protein-coupled receptors (GPCRs) that, in turn, regulate the function of the ion channels that establish synaptic weights and membrane excitability. Many of the basic molecular mechanisms of neuromodulation are now known, but the organization of neuromodulation at a network level is still an enigma. New single-cell RNA sequencing data and transcriptomic neurotaxonomies now offer bright new lights to shine on this critical “dark matter” of neuroscience. Here we leverage these advances to explore the cell-type-specific expression of genes encoding GPCRs, modulatory ligands, ion channels and intervening signal transduction molecules in mouse hippocampus area CA1, with the goal of revealing broad outlines of this well-studied brain structure’s neuromodulatory network architecture.
Collapse
Affiliation(s)
- Stephen J Smith
- Allen Institute for Brain Science, Seattle, WA, United States
- *Correspondence: Stephen J Smith,
| | - Mark von Zastrow
- Departments of Psychiatry and Pharmacology, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
36
|
Behl T, Madaan P, Sehgal A, Singh S, Makeen HA, Albratty M, Alhazmi HA, Meraya AM, Bungau S. Demystifying the Neuroprotective Role of Neuropeptides in Parkinson's Disease: A Newfangled and Eloquent Therapeutic Perspective. Int J Mol Sci 2022; 23:4565. [PMID: 35562956 PMCID: PMC9099669 DOI: 10.3390/ijms23094565] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/17/2022] [Accepted: 04/18/2022] [Indexed: 02/07/2023] Open
Abstract
Parkinson's disease (PD) refers to one of the eminently grievous, preponderant, tortuous nerve-cell-devastating ailments that markedly impacts the dopaminergic (DArgic) nerve cells of the midbrain region, namely the substantia nigra pars compacta (SN-PC). Even though the exact etiopathology of the ailment is yet indefinite, the existing corroborations have suggested that aging, genetic predisposition, and environmental toxins tremendously influence the PD advancement. Additionally, pathophysiological mechanisms entailed in PD advancement encompass the clumping of α-synuclein inside the lewy bodies (LBs) and lewy neurites, oxidative stress, apoptosis, neuronal-inflammation, and abnormalities in the operation of mitochondria, autophagy lysosomal pathway (ALP), and ubiquitin-proteasome system (UPS). The ongoing therapeutic approaches can merely mitigate the PD-associated manifestations, but until now, no therapeutic candidate has been depicted to fully arrest the disease advancement. Neuropeptides (NPs) are little, protein-comprehending additional messenger substances that are typically produced and liberated by nerve cells within the entire nervous system. Numerous NPs, for instance, substance P (SP), ghrelin, neuropeptide Y (NPY), neurotensin, pituitary adenylate cyclase-activating polypeptide (PACAP), nesfatin-1, and somatostatin, have been displayed to exhibit consequential neuroprotection in both in vivo and in vitro PD models via suppressing apoptosis, cytotoxicity, oxidative stress, inflammation, autophagy, neuronal toxicity, microglia stimulation, attenuating disease-associated manifestations, and stimulating chondriosomal bioenergetics. The current scrutiny is an effort to illuminate the neuroprotective action of NPs in various PD-experiencing models. The authors carried out a methodical inspection of the published work procured through reputable online portals like PubMed, MEDLINE, EMBASE, and Frontier, by employing specific keywords in the subject of our article. Additionally, the manuscript concentrates on representing the pathways concerned in bringing neuroprotective action of NPs in PD. In sum, NPs exert substantial neuroprotection through regulating paramount pathways indulged in PD advancement, and consequently, might be a newfangled and eloquent perspective in PD therapy.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, India; (P.M.); (A.S.); (S.S.)
| | - Piyush Madaan
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, India; (P.M.); (A.S.); (S.S.)
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, India; (P.M.); (A.S.); (S.S.)
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, India; (P.M.); (A.S.); (S.S.)
| | - Hafiz A. Makeen
- Pharmacy Practice Research Unit, Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (H.A.M.); (A.M.M.)
| | - Mohammed Albratty
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (M.A.); (H.A.A.)
| | - Hassan A. Alhazmi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (M.A.); (H.A.A.)
- Substance Abuse and Toxicology Research Center, Jazan University, Jazan 45142, Saudi Arabia
| | - Abdulkarim M. Meraya
- Pharmacy Practice Research Unit, Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (H.A.M.); (A.M.M.)
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
- Doctoral School of Biomedical Sciences, University of Oradea, 410028 Oradea, Romania
| |
Collapse
|
37
|
Watts AG, Kanoski SE, Sanchez-Watts G, Langhans W. The physiological control of eating: signals, neurons, and networks. Physiol Rev 2022; 102:689-813. [PMID: 34486393 PMCID: PMC8759974 DOI: 10.1152/physrev.00028.2020] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/30/2021] [Indexed: 02/07/2023] Open
Abstract
During the past 30 yr, investigating the physiology of eating behaviors has generated a truly vast literature. This is fueled in part by a dramatic increase in obesity and its comorbidities that has coincided with an ever increasing sophistication of genetically based manipulations. These techniques have produced results with a remarkable degree of cell specificity, particularly at the cell signaling level, and have played a lead role in advancing the field. However, putting these findings into a brain-wide context that connects physiological signals and neurons to behavior and somatic physiology requires a thorough consideration of neuronal connections: a field that has also seen an extraordinary technological revolution. Our goal is to present a comprehensive and balanced assessment of how physiological signals associated with energy homeostasis interact at many brain levels to control eating behaviors. A major theme is that these signals engage sets of interacting neural networks throughout the brain that are defined by specific neural connections. We begin by discussing some fundamental concepts, including ones that still engender vigorous debate, that provide the necessary frameworks for understanding how the brain controls meal initiation and termination. These include key word definitions, ATP availability as the pivotal regulated variable in energy homeostasis, neuropeptide signaling, homeostatic and hedonic eating, and meal structure. Within this context, we discuss network models of how key regions in the endbrain (or telencephalon), hypothalamus, hindbrain, medulla, vagus nerve, and spinal cord work together with the gastrointestinal tract to enable the complex motor events that permit animals to eat in diverse situations.
Collapse
Affiliation(s)
- Alan G Watts
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Scott E Kanoski
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Graciela Sanchez-Watts
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, Eidgenössische Technische Hochschule-Zürich, Schwerzenbach, Switzerland
| |
Collapse
|
38
|
Yang K, Liu T, Wang Z, Liu J, Shen Y, Pan X, Wen R, Xie H, Ruan Z, Tan Z, Chen Y, Guo A, Liu H, Han H, Di Z, Zhang K. Classifying Drosophila Olfactory Projection Neuron Boutons by Quantitative Analysis of Electron Microscopic Reconstruction. iScience 2022; 25:104180. [PMID: 35494235 PMCID: PMC9038572 DOI: 10.1016/j.isci.2022.104180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 01/25/2022] [Accepted: 03/29/2022] [Indexed: 11/29/2022] Open
Affiliation(s)
- Kai Yang
- School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
- BNU-BUCM Hengqin Innovation Institute of Science and Technology, Zhuhai, Guangdong 518057, China
| | - Tong Liu
- International Academic Center of Complex Systems, Advanced Institute of Natural Sciences, Beijing Normal University at Zhuhai, Zhuhai, Guangdong 519087, China
| | - Ze Wang
- International Academic Center of Complex Systems, Advanced Institute of Natural Sciences, Beijing Normal University at Zhuhai, Zhuhai, Guangdong 519087, China
| | - Jing Liu
- Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Yuxinyao Shen
- Huitong College, Beijing Normal University at Zhuhai, Zhuhai, Guangdong 519087, China
| | - Xinyi Pan
- Huitong College, Beijing Normal University at Zhuhai, Zhuhai, Guangdong 519087, China
| | - Ruyi Wen
- Huitong College, Beijing Normal University at Zhuhai, Zhuhai, Guangdong 519087, China
| | - Haotian Xie
- Huitong College, Beijing Normal University at Zhuhai, Zhuhai, Guangdong 519087, China
| | - Zhaoxuan Ruan
- Huitong College, Beijing Normal University at Zhuhai, Zhuhai, Guangdong 519087, China
| | - Zixiao Tan
- Huitong College, Beijing Normal University at Zhuhai, Zhuhai, Guangdong 519087, China
| | - Yingying Chen
- Huitong College, Beijing Normal University at Zhuhai, Zhuhai, Guangdong 519087, China
| | - Aike Guo
- International Academic Center of Complex Systems, Advanced Institute of Natural Sciences, Beijing Normal University at Zhuhai, Zhuhai, Guangdong 519087, China
- Huitong College, Beijing Normal University at Zhuhai, Zhuhai, Guangdong 519087, China
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - He Liu
- International Academic Center of Complex Systems, Advanced Institute of Natural Sciences, Beijing Normal University at Zhuhai, Zhuhai, Guangdong 519087, China
| | - Hua Han
- Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Zengru Di
- International Academic Center of Complex Systems, Advanced Institute of Natural Sciences, Beijing Normal University at Zhuhai, Zhuhai, Guangdong 519087, China
| | - Ke Zhang
- International Academic Center of Complex Systems, Advanced Institute of Natural Sciences, Beijing Normal University at Zhuhai, Zhuhai, Guangdong 519087, China
- Corresponding author
| |
Collapse
|
39
|
De-Miguel FF. The Thermodynamically Expensive Contribution of Three Calcium Sources to Somatic Release of Serotonin. Int J Mol Sci 2022; 23:1495. [PMID: 35163419 PMCID: PMC8836226 DOI: 10.3390/ijms23031495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/28/2021] [Accepted: 12/30/2021] [Indexed: 02/01/2023] Open
Abstract
The soma, dendrites and axon of neurons may display calcium-dependent release of transmitters and peptides. Such release is named extrasynaptic for occurring in absence of synaptic structures. This review describes the cooperative actions of three calcium sources on somatic exocytosis. Emphasis is given to the somatic release of serotonin by the classical leech Retzius neuron, which has allowed detailed studies on the fine steps from excitation to exocytosis. Trains of action potentials induce transmembrane calcium entry through L-type channels. For action potential frequencies above 5 Hz, summation of calcium transients on individual action potentials activates the second calcium source: ryanodine receptors produce calcium-induced calcium release. The resulting calcium tsunami activates mitochondrial ATP synthesis to fuel transport of vesicles to the plasma membrane. Serotonin that is released maintains a large-scale exocytosis by activating the third calcium source: serotonin autoreceptors coupled to phospholipase C promote IP3 production. Activated IP3 receptors in peripheral endoplasmic reticulum release calcium that promotes vesicle fusion. The Swiss-clock workings of the machinery for somatic exocytosis has a striking disadvantage. The essential calcium-releasing endoplasmic reticulum near the plasma membrane hinders the vesicle transport, drastically reducing the thermodynamic efficiency of the ATP expenses and elevating the energy cost of release.
Collapse
Affiliation(s)
- Francisco F De-Miguel
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
40
|
Muthu SJ, Lakshmanan G, Seppan P. Influence of Testosterone depletion on Neurotrophin-4 in Hippocampal synaptic plasticity and its effects on learning and memory. Dev Neurosci 2022; 44:102-112. [PMID: 35086088 DOI: 10.1159/000522201] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 01/22/2022] [Indexed: 11/19/2022] Open
Abstract
Sex steroids are neuromodulators that play a crucial role in learning, memory, and synaptic plasticity, providing circuit flexibility and dynamic functional connectivity in mammals. Previous studies indicate that testosterone is crucial for neuronal functions and required further investigation on various frontiers. However, it is surprising to note that studies on testosterone-induced NT-4 expression and its influence on synaptic plasticity and learning and memory moderation are scanty. The present study is focused on analyzing the localized influence of neurotrophin-4 (NT4) on hippocampal synaptic plasticity and associated moderation in learning and memory under testosterone deprivation. Adult Wistar albino rats were randomly divided into various groups, control (Cont), orchidectomy (ORX), orchidectomy + testosterone supplementation (ORX+T) and control + testosterone (Cont+T). After two weeks, the serum testosterone level was undetectable in ORX rats. The behavioural assessment showed a decline in the learning ability of ORX rats with increased working and reference memory errors in the behavioural assessment in the 8-arm radial maze. The mRNA and protein expressions of NT-4 and androgen receptors were significantly reduced in the ORX group. In addition, there was a decrease in the number of neuronal dendrites in Golgi-Cox staining. These changes were not seen in ORX+T rats with improved learning behaviour. Indicating that testosterone exerts its protective effect on hippocampal synaptic plasticity through androgen receptor-dependent neurotrophin-4 regulation in learning and memory upgrade.
Collapse
Affiliation(s)
- Sakthi Jothi Muthu
- Department of Anatomy, Dr. Arcot Lakshmanasamy Mudaliar Postgraduate Institute of Basic Medical Sciences, University of Madras, Chennai, India
| | - Ganesh Lakshmanan
- Department of Anatomy, Dr. Arcot Lakshmanasamy Mudaliar Postgraduate Institute of Basic Medical Sciences, University of Madras, Chennai, India
| | - Prakash Seppan
- Department of Anatomy, Dr. Arcot Lakshmanasamy Mudaliar Postgraduate Institute of Basic Medical Sciences, University of Madras, Chennai, India
| |
Collapse
|
41
|
Baraniuk JN. Review of the Midbrain Ascending Arousal Network Nuclei and Implications for Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS), Gulf War Illness (GWI) and Postexertional Malaise (PEM). Brain Sci 2022; 12:132. [PMID: 35203896 PMCID: PMC8870178 DOI: 10.3390/brainsci12020132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/10/2022] [Accepted: 01/14/2022] [Indexed: 12/10/2022] Open
Abstract
Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS and Gulf War Illness (GWI) share features of post-exertional malaise (PEM), exertional exhaustion, or postexertional symptom exacerbation. In a two-day model of PEM, submaximal exercise induced significant changes in activation of the dorsal midbrain during a high cognitive load working memory task (Washington 2020) (Baraniuk this issue). Controls had no net change. However, ME/CFS had increased activity after exercise, while GWI had significantly reduced activity indicating differential responses to exercise and pathological mechanisms. These data plus findings of the midbrain and brainstem atrophy in GWI inspired a review of the anatomy and physiology of the dorsal midbrain and isthmus nuclei in order to infer dysfunctional mechanisms that may contribute to disease pathogenesis and postexertional malaise. The nuclei of the ascending arousal network were addressed. Midbrain and isthmus nuclei participate in threat assessment, awareness, attention, mood, cognition, pain, tenderness, sleep, thermoregulation, light and sound sensitivity, orthostatic symptoms, and autonomic dysfunction and are likely to contribute to the symptoms of postexertional malaise in ME/CFS and GWI.
Collapse
Affiliation(s)
- James N Baraniuk
- Department of Medicine, Georgetown University, Washington, DC 20057, USA
| |
Collapse
|
42
|
Bakalar D, Sweat S, Drossel G, Jiang SZ, Samal BS, Stroth N, Xu W, Zhang L, Zhang H, Eiden LE. Relationships between constitutive and acute gene regulation, and physiological and behavioral responses, mediated by the neuropeptide PACAP. Psychoneuroendocrinology 2022; 135:105447. [PMID: 34741979 PMCID: PMC8900973 DOI: 10.1016/j.psyneuen.2021.105447] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 09/10/2021] [Accepted: 10/08/2021] [Indexed: 01/03/2023]
Abstract
Since the advent of gene knock-out technology in 1987, insight into the role(s) of neuropeptides in centrally- and peripherally-mediated physiological regulation has been gleaned by examining altered physiological functioning in mammals, predominantly mice, after genetic editing to produce animals deficient in neuropeptides or their cognate G-protein coupled receptors (GPCRs). These results have complemented experiments involving infusion of neuropeptide agonists or antagonists systemically or into specific brain regions. Effects of gene loss are often interpreted as indicating that the peptide and its receptor(s) are required for the physiological or behavioral responses elicited in wild-type mice at the time of experimental examination. These interpretations presume that peptide/peptide receptor gene deletion affects only the expression of the peptide/receptor itself, and therefore impacts physiological events only at the time at which the experiment is conducted. A way to support 'real-time' interpretations of neuropeptide gene knock-out is to demonstrate that the wild-type transcriptome, except for the deliberately deleted gene(s), in tissues of interest, is preserved in the knock-out mouse. Here, we show that there is a cohort of genes (constitutively PACAP-Regulated Genes, or cPRGs) whose basal expression is affected by constitutive knock-out of the Adcyap1 gene in C57Bl6/N mice, and additional genes whose expression in response to physiological challenge, in adults, is altered or impaired in the absence of PACAP expression (acutely PACAP-Regulated Genes, or aPRGs). Distinguishing constitutive and acute transcriptomic effects of neuropeptide deficiency on physiological function and behavior in mice reveals alternative mechanisms of action, and changing functions of neuropeptides, throughout the lifespan.
Collapse
Affiliation(s)
- Dana Bakalar
- Section on Molecular Neuroscience, National Institute of Mental Heath - Intramural Research Program, Bethesda, MD. NIH, USA
| | - Sean Sweat
- Section on Molecular Neuroscience, National Institute of Mental Heath - Intramural Research Program, Bethesda, MD. NIH, USA
| | - Gunner Drossel
- Section on Molecular Neuroscience, National Institute of Mental Heath - Intramural Research Program, Bethesda, MD. NIH, USA
| | - Sunny Z. Jiang
- Section on Molecular Neuroscience, National Institute of Mental Heath - Intramural Research Program, Bethesda, MD. NIH, USA
| | - Babru S. Samal
- Section on Molecular Neuroscience, National Institute of Mental Heath - Intramural Research Program, Bethesda, MD. NIH, USA
| | - Nikolas Stroth
- Section on Molecular Neuroscience, National Institute of Mental Heath - Intramural Research Program, Bethesda, MD. NIH, USA
| | - Wenqin Xu
- Section on Molecular Neuroscience, National Institute of Mental Heath - Intramural Research Program, Bethesda, MD. NIH, USA
| | - Limei Zhang
- Section on Molecular Neuroscience, National Institute of Mental Heath - Intramural Research Program, Bethesda, MD. NIH, USA,Department of Physiology, Autonomous National University of Mexico (UNAM) Medical School, Mexico City, Mexico
| | - Haiying Zhang
- Section on Molecular Neuroscience, National Institute of Mental Heath - Intramural Research Program, Bethesda, MD. NIH, USA
| | - Lee E. Eiden
- Section on Molecular Neuroscience, National Institute of Mental Heath - Intramural Research Program, Bethesda, MD. NIH, USA,Correspondence Lee E. Eiden, Ph.D., Section on Molecular Neuroscience, National Institute of Mental Heath – Intramural Research Program, Bethesda, MD. NIH, USA, Phone: +13014964110,
| |
Collapse
|
43
|
Marcoli M, Agnati LF, Franco R, Cortelli P, Anderlini D, Guidolin D, Cervetto C, Maura G. Modulating brain integrative actions as a new perspective on pharmacological approaches to neuropsychiatric diseases. Front Endocrinol (Lausanne) 2022; 13:1038874. [PMID: 36699033 PMCID: PMC9868467 DOI: 10.3389/fendo.2022.1038874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 12/20/2022] [Indexed: 01/11/2023] Open
Abstract
A critical aspect of drug development in the therapy of neuropsychiatric diseases is the "Target Problem", that is, the selection of a proper target after not simply the etiopathological classification but rather the detection of the supposed structural and/or functional alterations in the brain networks. There are novel ways of approaching the development of drugs capable of overcoming or at least reducing the deficits without triggering deleterious side effects. For this purpose, a model of brain network organization is needed, and the main aspects of its integrative actions must also be established. Thus, to this aim we here propose an updated model of the brain as a hyper-network in which i) the penta-partite synapses are suggested as key nodes of the brain hyper-network and ii) interacting cell surface receptors appear as both decoders of signals arriving to the network and targets of central nervous system diseases. The integrative actions of the brain networks follow the "Russian Doll organization" including the micro (i.e., synaptic) and nano (i.e., molecular) levels. In this scenario, integrative actions result primarily from protein-protein interactions. Importantly, the macromolecular complexes arising from these interactions often have novel structural binding sites of allosteric nature. Taking G protein-coupled receptors (GPCRs) as potential targets, GPCRs heteromers offer a way to increase the selectivity of pharmacological treatments if proper allosteric drugs are designed. This assumption is founded on the possible selectivity of allosteric interventions on G protein-coupled receptors especially when organized as "Receptor Mosaics" at penta-partite synapse level.
Collapse
Affiliation(s)
- Manuela Marcoli
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), Pisa, Italy
- Center of Excellence for Biomedical Research, University of Genova, Genova, Italy
- *Correspondence: Manuela Marcoli, ; Luigi F. Agnati,
| | - Luigi F. Agnati
- Department of Biomedical, Metabolic Sciences and Neuroscience, University of Modena and Reggio Emilia, Modena, Italy
- *Correspondence: Manuela Marcoli, ; Luigi F. Agnati,
| | - Rafael Franco
- CiberNed Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain
- Molecular Neurobiology laboratory, Department of Biochemistry and Molecular Biomedicine. Universitat de Barcelona, Barcelona, Spain
- School of Chemistry, Universitat de Barcelona, Barcelona, Spain
| | - Pietro Cortelli
- Department of Biomedical and NeuroMotor Sciences (DIBINEM), Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Deanna Anderlini
- Centre for Sensorimotor Performance, The University of Queensland, Brisbane, QLD, Australia
| | - Diego Guidolin
- Department of Neuroscience, University of Padova, Padova, Italy
| | - Chiara Cervetto
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), Pisa, Italy
| | - Guido Maura
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy
| |
Collapse
|
44
|
Spexin Promotes the Proliferation and Differentiation of C2C12 Cells In Vitro—The Effect of Exercise on SPX and SPX Receptor Expression in Skeletal Muscle In Vivo. Genes (Basel) 2021; 13:genes13010081. [PMID: 35052420 PMCID: PMC8774514 DOI: 10.3390/genes13010081] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/15/2021] [Accepted: 12/27/2021] [Indexed: 01/04/2023] Open
Abstract
SPX (spexin) and its receptors GalR2 and GalR3 (galanin receptor subtype 2 and galanin receptor subtype 3) play an important role in the regulation of lipid and carbohydrate metabolism in human and animal fat tissue. However, little is still known about the role of this peptide in the metabolism of muscle. The aim of this study was to determine the impact of SPX on the metabolism, proliferation and differentiation of the skeletal muscle cell line C2C12. Moreover, we determined the effect of exercise on the SPX transduction pathway in mice skeletal muscle. We found that increased SPX, acting via GalR2 and GalR3 receptors, and ERK1/2 phosphorylation stimulated the proliferation of C2C12 cells (p < 0.01). We also noted that SPX stimulated the differentiation of C2C12 by increasing mRNA and protein levels of differentiation markers Myh, myogenin and MyoD (p < 0.01). SPX consequently promoted myoblast fusion into the myotubule (p < 0.01). Moreover, we found that, in the first stage (after 2 days) of myocyte differentiation, GalR2 and GalR3 were involved, whereas in the last stage (day six), the effect of SPX was mediated by the GalR3 isoform. We also noted that exercise stimulated SPX and GalR2 expression in mice skeletal muscle as well as an increase in SPX concentration in blood serum. These new insights may contribute to a better understanding of the role of SPX in the metabolism of skeletal muscle.
Collapse
|
45
|
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: 32] [Impact Index Per Article: 8.0] [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
|
46
|
Smith SJ. Transcriptomic evidence for dense peptidergic networks within forebrains of four widely divergent tetrapods. Curr Opin Neurobiol 2021; 71:100-109. [PMID: 34775262 DOI: 10.1016/j.conb.2021.09.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/21/2021] [Accepted: 09/27/2021] [Indexed: 12/15/2022]
Abstract
The primary function common to every neuron is communication with other neurons. Such cell-cell signaling can take numerous forms, including fast synaptic transmission and slower neuromodulation via secreted messengers, such as neuropeptides, dopamine, and many other diffusible small molecules. Individual neurons are quite diverse, however, in all particulars of both synaptic and neuromodulatory communication. Neuron classification schemes have therefore proven very useful in exploring the emergence of network function, behavior, and cognition from the communication functions of individual neurons. Recently published single-cell mRNA sequencing data and corresponding transcriptomic neuron classifications from turtle, songbird, mouse, and human provide evidence for a long evolutionary history and adaptive significance of localized peptidergic signaling. Across all four species, sets of at least twenty orthologous cognate pairs of neuropeptide precursor protein and receptor genes are expressed in individually sparse but heavily overlapping patterns suggesting that all forebrain neuron types are densely interconnected by local peptidergic signals.
Collapse
|
47
|
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: 44] [Impact Index Per Article: 11.0] [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
|
48
|
Foster SL, Galaj E, Karne SL, Ferré S, Weinshenker D. Cell-type specific expression and behavioral impact of galanin and GalR1 in the locus coeruleus during opioid withdrawal. Addict Biol 2021; 26:e13037. [PMID: 33768673 PMCID: PMC8376771 DOI: 10.1111/adb.13037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/15/2021] [Indexed: 12/14/2022]
Abstract
The neuropeptide galanin is reported to attenuate opioid withdrawal symptoms, potentially by reducing neuronal hyperactivity in the noradrenergic locus coeruleus (LC) via galanin receptor 1 (GalR1). We evaluated this mechanism by using RNAscope in situ hybridization to characterize GalR1 mRNA distribution in the dorsal pons and to compare galanin and GalR1 mRNA expression in tyrosine hydroxylase-positive (TH+) LC cells at baseline and following chronic morphine or precipitated withdrawal. We then used genetically altered mouse lines and pharmacology to test whether noradrenergic galanin (NE-Gal) modulates withdrawal symptoms. RNAscope revealed that, while GalR1 signal was evident in the dorsal pons, 80.7% of the signal was attributable to TH- neurons outside the LC. Galanin and TH mRNA were abundant in LC cells at baseline and were further increased by withdrawal, whereas low basal GalR1 mRNA expression was unaltered by chronic morphine or withdrawal. Naloxone-precipitated withdrawal symptoms in mice lacking NE-Gal (GalcKO-Dbh ) were largely similar to WT littermates, indicating that loss of NE-Gal does not exacerbate withdrawal. Complementary experiments using NE-Gal overexpressor mice (NE-Gal OX) and systemic administration of the galanin receptor agonist galnon revealed that increasing galanin signaling also failed to alter behavioral withdrawal, while suppressing noradrenergic transmission with the alpha-2 adrenergic receptor agonist clonidine attenuated multiple symptoms. These results indicate that galanin does not acutely attenuate precipitated opioid withdrawal via an LC-specific mechanism, which has important implications for the general role of galanin in regulation of somatic and affective opioid responses and LC activity.
Collapse
Affiliation(s)
- Stephanie L. Foster
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Ewa Galaj
- Addiction Biology Unit, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland, USA
| | - Saumya L. Karne
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - David Weinshenker
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA,Correspondence: David Weinshenker, PhD, Department of Human Genetics, 615 Michael St, Whitehead 301, Atlanta, GA 30322, , Fax: (404) 727-3949
| |
Collapse
|
49
|
Trueta C. An analytical method to measure the contribution of clear synaptic and dense-core peri-synaptic vesicles to neurotransmitter release from synaptic terminals with two classes of secretory vesicles. MethodsX 2021; 8:101374. [PMID: 34430270 PMCID: PMC8374493 DOI: 10.1016/j.mex.2021.101374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 04/25/2021] [Indexed: 12/03/2022] Open
Abstract
Two types of secretory vesicles co-exist at some presynaptic terminals. Clear synaptic vesicles (CSV) release their contents at the synaptic active zone, upon single impulses, while dense-core vesicles (DCV) usually release their contents in the periphery of the terminal upon repetitive stimulation. Part of the transmitter released by DCV diffuses to produce paracrine effects, and part of it reaches the postsynaptic terminal, adding its effect to that of synaptic release. This article presents an analytical method to separate the contribution of CSV and DCV to the postsynaptic responses, based on the kinetics of postsynaptic currents (PSCs). Since stimulation with single presynaptic impulses usually triggers release only from CSV, the kinetics of the resulting PSC can be used as a template to model the postsynaptic response to release from CSV during stimulation trains, accounting for the variations in the amplitude of PSCs due to short-term synaptic plasticity. Subtraction of this model simulation to the total recorded PSC renders the response to DCV peri‑synaptic release, which has slower kinetics. The method can be further simplified by measuring only the amplitudes of the PSC peaks for synaptic release and the integral of the current for peri‑synaptic release.The postsynaptic current in response to presynaptic release from clear synaptic vesicles is modeled using the kinetics of the PSC in response to single impulses. The model synaptic response is subtracted from the total recorded PSC to obtain the response to peri‑synaptic release from dense-core vesicles.
Collapse
|
50
|
Jiang M, Zhao B, Luo S, Wang Q, Chu Y, Chen T, Mao X, Liu Y, Wang Y, Jiang X, Wei DQ, Xiong Y. NeuroPpred-Fuse: an interpretable stacking model for prediction of neuropeptides by fusing sequence information and feature selection methods. Brief Bioinform 2021; 22:6350884. [PMID: 34396388 DOI: 10.1093/bib/bbab310] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 07/01/2021] [Accepted: 07/18/2021] [Indexed: 12/13/2022] Open
Abstract
Neuropeptides acting as signaling molecules in the nervous system of various animals play crucial roles in a wide range of physiological functions and hormone regulation behaviors. Neuropeptides offer many opportunities for the discovery of new drugs and targets for the treatment of neurological diseases. In recent years, there have been several data-driven computational predictors of various types of bioactive peptides, but the relevant work about neuropeptides is little at present. In this work, we developed an interpretable stacking model, named NeuroPpred-Fuse, for the prediction of neuropeptides through fusing a variety of sequence-derived features and feature selection methods. Specifically, we used six types of sequence-derived features to encode the peptide sequences and then combined them. In the first layer, we ensembled three base classifiers and four feature selection algorithms, which select non-redundant important features complementarily. In the second layer, the output of the first layer was merged and fed into logistic regression (LR) classifier to train the model. Moreover, we analyzed the selected features and explained the feasibility of the selected features. Experimental results show that our model achieved 90.6% accuracy and 95.8% AUC on the independent test set, outperforming the state-of-the-art models. In addition, we exhibited the distribution of selected features by these tree models and compared the results on the training set to that on the test set. These results fully showed that our model has a certain generalization ability. Therefore, we expect that our model would provide important advances in the discovery of neuropeptides as new drugs for the treatment of neurological diseases.
Collapse
Affiliation(s)
- Mingming Jiang
- State Key Laboratory of Microbial Metabolism, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Bowen Zhao
- State Key Laboratory of Microbial Metabolism, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shenggan Luo
- State Key Laboratory of Microbial Metabolism, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qiankun Wang
- State Key Laboratory of Microbial Metabolism, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yanyi Chu
- State Key Laboratory of Microbial Metabolism, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Tianhang Chen
- State Key Laboratory of Microbial Metabolism, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xueying Mao
- State Key Laboratory of Microbial Metabolism, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yatong Liu
- State Key Laboratory of Microbial Metabolism, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yanjing Wang
- State Key Laboratory of Microbial Metabolism, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xue Jiang
- State Key Laboratory of Microbial Metabolism, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Dong-Qing Wei
- State Key Laboratory of Microbial Metabolism, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yi Xiong
- State Key Laboratory of Microbial Metabolism, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|