1
|
Asahina K, Zelikowsky M. Comparative Perspectives on Neuropeptide Function and Social Isolation. Biol Psychiatry 2025; 97:942-952. [PMID: 39892690 PMCID: PMC12048258 DOI: 10.1016/j.biopsych.2025.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 01/07/2025] [Accepted: 01/25/2025] [Indexed: 02/04/2025]
Abstract
Chronic social isolation alters behavior across animal species. Genetic model organisms such as mice and flies provide crucial insight into the molecular and physiological effects of social isolation on brain cells and circuits. Here, we comparatively review recent findings regarding the function of conserved neuropeptides in social isolation in mice and flies. Analogous functions of 3 classes of neuropeptides-tachykinins, cholecystokinins, and neuropeptide Y/F-in the two model organisms suggest that these molecules may be involved in modulating behavioral changes induced by social isolation across a wider range of species, including humans. Comparative approaches armed with tools to dissect neuropeptidergic function can lead to an integrated understanding of the impacts of social isolation on brain circuits and behavior.
Collapse
Affiliation(s)
- Kenta Asahina
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California.
| | - Moriel Zelikowsky
- Department of Neurobiology, School of Medicine, The University of Utah, Salt Lake City, Utah
| |
Collapse
|
2
|
Dong J, Chen M, van Weering JRT, Li KW, Smit AB, Toonen RF, Verhage M. Rab10 regulates neuropeptide release by maintaining Ca 2+ homeostasis and protein synthesis. eLife 2025; 13:RP94930. [PMID: 40172954 PMCID: PMC11964448 DOI: 10.7554/elife.94930] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025] Open
Abstract
Dense core vesicles (DCVs) transport and release various neuropeptides and neurotrophins that control diverse brain functions, but the DCV secretory pathway remains poorly understood. Here, we tested a prediction emerging from invertebrate studies about the crucial role of the intracellular trafficking GTPase Rab10, by assessing DCV exocytosis at single-cell resolution upon acute Rab10 depletion in mature mouse hippocampal neurons, to circumvent potential confounding effects of Rab10's established role in neurite outgrowth. We observed a significant inhibition of DCV exocytosis in Rab10-depleted neurons, whereas synaptic vesicle exocytosis was unaffected. However, rather than a direct involvement in DCV trafficking, this effect was attributed to two ER-dependent processes, ER-regulated intracellular Ca2+ dynamics, and protein synthesis. Gene Ontology analysis of differentially expressed proteins upon Rab10 depletion identified substantial alterations in synaptic and ER/ribosomal proteins, including the Ca2+ pump SERCA2. In addition, ER morphology and dynamics were altered, ER Ca2+ levels were depleted, and Ca2+ homeostasis was impaired in Rab10-depleted neurons. However, Ca2+ entry using a Ca2+ ionophore still triggered less DCV exocytosis. Instead, leucine supplementation, which enhances protein synthesis, largely rescued DCV exocytosis deficiency. We conclude that Rab10 is required for neuropeptide release by maintaining Ca2+ dynamics and regulating protein synthesis. Furthermore, DCV exocytosis appeared more dependent on (acute) protein synthesis than synaptic vesicle exocytosis.
Collapse
Affiliation(s)
- Jian Dong
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) AmsterdamAmsterdamNetherlands
| | - Mian Chen
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) AmsterdamAmsterdamNetherlands
| | - Jan RT van Weering
- Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research (CNCR), University Medical Center AmsterdamAmsterdamNetherlands
| | - Ka Wan Li
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) AmsterdamAmsterdamNetherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) AmsterdamAmsterdamNetherlands
| | - Ruud F Toonen
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) AmsterdamAmsterdamNetherlands
| | - Matthijs Verhage
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) AmsterdamAmsterdamNetherlands
- Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research (CNCR), University Medical Center AmsterdamAmsterdamNetherlands
| |
Collapse
|
3
|
Nässel DR. What Drosophila can tell us about state-dependent peptidergic signaling in insects. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2025; 179:104275. [PMID: 39956367 DOI: 10.1016/j.ibmb.2025.104275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/12/2025] [Accepted: 02/12/2025] [Indexed: 02/18/2025]
Abstract
Plasticity in animal behavior and physiology is largely due to modulatory and regulatory signaling with neuropeptides and peptide hormones (collectively abbreviated NPHs). The NPHs constitute a very large and versatile group of signaling substances that partake at different regulatory levels in most daily activities of an organism. This review summarizes key principles in NPH actions in the brain and in interorgan signaling, with focus on Drosophila. NPHs are produced by neurons, neurosecretory cells (NSCs) and other endocrine cells in NPH-specific and stereotypic patterns. Most of the NPHs have multiple (pleiotropic) functions and target several different neuronal circuits and/or peripheral tissues. Such divergent NPH signaling ensures orchestration of behavior and physiology in state-dependent manners. Conversely, many neurons, circuits, NSCs, or other cells, are targeted by multiple NPHs. This convergent signaling commonly conveys various signals reporting changes in the external and internal environment to central neurons/circuits. As an example of wider functional convergence, 26 different Drosophila NPHs act at many different levels to regulate food search and feeding. Convergence is also seen in hormonal regulation of peripheral functions. For instance, multiple NPHs target renal tubules to ensure osmotic homeostasis. Interestingly, several of the same osmoregulatory NPHs also regulate feeding, metabolism and stress. However, for some NPHs the cellular distribution and functions suggests multiple unrelated functions that are restricted to specific circuits. Thus, NPH signaling follows distinct patterns for each specific NPH, but taken together they form overlapping networks that modulate behavior and physiology.
Collapse
Affiliation(s)
- Dick R Nässel
- Department of Zoology, Stockholm University, S-10691, Stockholm, Sweden.
| |
Collapse
|
4
|
Antal M. Molecular Anatomy of Synaptic and Extrasynaptic Neurotransmission Between Nociceptive Primary Afferents and Spinal Dorsal Horn Neurons. Int J Mol Sci 2025; 26:2356. [PMID: 40076973 PMCID: PMC11900602 DOI: 10.3390/ijms26052356] [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: 12/04/2024] [Revised: 02/26/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
Sensory signals generated by peripheral nociceptors are transmitted by peptidergic and nonpeptidergic nociceptive primary afferents to the superficial spinal dorsal horn, where their central axon terminals establish synaptic contacts with secondary sensory spinal neurons. In the case of suprathreshold activation, the axon terminals release glutamate into the synaptic cleft and stimulate postsynaptic spinal neurons by activating glutamate receptors located on the postsynaptic membrane. When overexcitation is evoked by peripheral inflammation, neuropathy or pruritogens, peptidergic nociceptive axon terminals may corelease various neuropeptides, neurotrophins and endomorphin, together with glutamate. However, in contrast to glutamate, neuropeptides, neurotrophins and endomorphin are released extrasynaptically. They diffuse from the site of release and modulate the function of spinal neurons via volume transmission, activating specific extrasynaptic receptors. Thus, the released neuropeptides, neurotrophins and endomorphin may evoke excitation, disinhibition or inhibition in various spinal neuronal populations, and together with glutamate, induce overall overexcitation, called central sensitization. In addition, the synaptic and extrasynaptic release of neurotransmitters is subjected to strong retrograde control mediated by various retrogradely acting transmitters, messengers, and their presynaptic receptors. Moreover, the composition of this complex chemical apparatus is heavily dependent on the actual patterns of nociceptive primary afferent activation in the periphery. This review provides an overview of the complexity of this signaling apparatus, how nociceptive primary afferents can activate secondary sensory spinal neurons via synaptic and volume transmission in the superficial spinal dorsal horn, and how these events can be controlled by presynaptic mechanisms.
Collapse
Affiliation(s)
- Miklós Antal
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| |
Collapse
|
5
|
Dereli AS, Apaire A, El Tahry R. Sudden Unexpected Death in Epilepsy: Central Respiratory Chemoreception. Int J Mol Sci 2025; 26:1598. [PMID: 40004062 PMCID: PMC11855741 DOI: 10.3390/ijms26041598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/03/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Sudden unexpected death in epilepsy (SUDEP) is a critical concern for individuals suffering from epilepsy, with respiratory dysfunction playing a significant role in its pathology. Fatal seizures are often characterized by central apnea and hypercapnia (elevated CO2 levels), indicating a failure in ventilatory control. Research has shown that both human epilepsy patients and animal models exhibit a reduced hypercapnic ventilatory response in the interictal (non-seizure) period, suggesting an impaired ability to regulate breathing in response to high CO2 levels. This review examines the role of central chemoreceptors-specifically the retrotrapezoid nucleus, raphe nuclei, nucleus tractus solitarius, locus coeruleus, and hypothalamus in this pathology. These structures are critical for sensing CO2 and maintaining respiratory homeostasis. Emerging evidence also implicates neuropeptidergic pathways within these chemoreceptive regions in SUDEP. Neuropeptides like galanin, pituitary adenylate cyclase-activating peptide (PACAP), orexin, somatostatin, and bombesin-like peptides may modulate chemosensitivity and respiratory function, potentially exacerbating respiratory failure during seizures. Understanding the mechanisms linking central chemoreception, respiratory control, and neuropeptidergic signaling is essential to developing targeted interventions to reduce the risk of SUDEP in epilepsy patients.
Collapse
Affiliation(s)
- Ayse S. Dereli
- Clinical Neuroscience, Institute of Neuroscience (IoNS), Université Catholique de Louvain, 1200 Brussels, Belgium; (A.A.); (R.E.T.)
| | - Auriane Apaire
- Clinical Neuroscience, Institute of Neuroscience (IoNS), Université Catholique de Louvain, 1200 Brussels, Belgium; (A.A.); (R.E.T.)
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), WEL Research Institute, 1300 Wavre, Belgium
| | - Riem El Tahry
- Clinical Neuroscience, Institute of Neuroscience (IoNS), Université Catholique de Louvain, 1200 Brussels, Belgium; (A.A.); (R.E.T.)
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), WEL Research Institute, 1300 Wavre, Belgium
- Center for Refractory Epilepsy, Department of Neurology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| |
Collapse
|
6
|
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
|
7
|
Zhang L, Sun L, Song G, Wang B, Cui Y, Liu F, Li Y, Wang Z. Genome-Wide Identification and Expression of Neuropeptides and Their Expression Patterns After RNAi of CHH Genes in Pacific White Shrimp Litopenaeus vannamei. BIOLOGY 2024; 13:1038. [PMID: 39765705 PMCID: PMC11673531 DOI: 10.3390/biology13121038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025]
Abstract
Neuropeptides are pivotal in regulating a broad spectrum of developmental, physiological, and behavioral processes throughout the life cycle of crustaceans. In this comprehensive study, we utilized a multiomics approach to characterize neuropeptide precursors and to assess the expression profiles of neuropeptide-encoding genes across various tissues and developmental stages in the Pacific white shrimp, Litopenaeus vannamei. Additionally, we explored the differential expression of neuropeptide genes in the eyestalk before and after the RNA interference-mediated suppression of crustacean hyperglycemic hormone (CHH) and vitellogenesis-inhibiting hormone (VIH) gene expression. Our study identified a total of 125 neuropeptide-encoding genes in L. vannamei, with 54 of these genes previously uncharacterized in the genome. Notably, certain neuropeptide-encoding gene families showed significant expansion, as demonstrated by the discovery of 10 adipokinetic hormone/corazonin-like peptide (ACP) genes, 55 CHH superfamily genes, and 13 pigment-dispersing hormone (PDH) genes. Alternative splicing was also found to play a crucial role in generating functionally diverse neuropeptides; for example, the agatoxin and calcitonin genes undergo alternative splicing that leads to the production of three distinct agatoxin neuropeptides and two distinct calcitonin neuropeptides, respectively. Neuropeptide genes are predominantly expressed in neuroendocrine tissues, including the eyestalk, cerebral ganglia, thoracic ganglia, and ventral ganglia. During the embryonic development of L. vannamei, with the exception of the molt-inhibiting hormone (MIH) gene, all monitored genes display minimal expression from the zygote stage through to the larval in membrane (Lim) stage. In contrast, the majority of these genes exhibit a steady uptick in expression from the nauplius stage onwards, culminating in the post-larval stage. Furthermore, comparative transcriptomic analysis of the eyestalk revealed that the expression of the majority of neuropeptide genes was downregulated following the suppression of CHH and VIH gene expression. This downregulation was significantly associated with the enrichment of pathways related to amino acid metabolism and hormone synthesis. The findings of this study provide valuable insights for future research aimed at elucidating the role of neuropeptides in regulating physiological functions in L. vannamei, potentially leading to advancements in shrimp aquaculture practices.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Zhongkai Wang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China
| |
Collapse
|
8
|
Kim DI, Kang SJ, Jhang J, Jo YS, Park S, Ye M, Pyeon GH, Im GH, Kim SG, Han S. Encoding opposing valences through frequency-dependent transmitter switching in single peptidergic neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.09.622790. [PMID: 39574736 PMCID: PMC11581014 DOI: 10.1101/2024.11.09.622790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Peptidergic neurons often co-express fast transmitters and neuropeptides in separate vesicles with distinct release properties. However, the release dynamics of each transmitter in various contexts have not been fully understood in behaving animals. Here, we demonstrate that calcitonin gene-related peptide (CGRP) neurons in the external lateral subdivision of the parabrachial nucleus (CGRPPBel) encode opposing valence via differential release, rather than co-release, of glutamate and neuropeptides, according to firing rate. Glutamate is released preferentially at lower firing rates with minimal release at higher firing rates, whereas neuropeptides are released at higher firing rates, resulting in frequency-dependent switching of transmitters. Aversive stimuli evoke high frequency responses with accompanying neuropeptide release to encode negative valence, whereas appetitive stimuli evoke low frequency responses with glutamate release to encode positive valence. Our study reveals a previously unknown capability of single CGRPPBel neurons to bidirectionally encode valence via frequency-dependent differential release of transmitters in vivo.
Collapse
Affiliation(s)
- Dong-Il Kim
- Peptide Biology Laboratory, The Salk Institute for Biological Studies; La Jolla, CA 92037, USA
| | - Sukjae J. Kang
- Peptide Biology Laboratory, The Salk Institute for Biological Studies; La Jolla, CA 92037, USA
| | - Jinho Jhang
- Peptide Biology Laboratory, The Salk Institute for Biological Studies; La Jolla, CA 92037, USA
| | - Yong S. Jo
- School of Psychology, Korea University; Seoul, Republic of Korea
| | - Seahyung Park
- Peptide Biology Laboratory, The Salk Institute for Biological Studies; La Jolla, CA 92037, USA
| | - Mao Ye
- Peptide Biology Laboratory, The Salk Institute for Biological Studies; La Jolla, CA 92037, USA
| | - Gyeong Hee Pyeon
- School of Psychology, Korea University; Seoul, Republic of Korea
| | - Geun-Ho Im
- Center for Neuroscience Imaging Research, Institute for Basic Science; Suwon, Republic of Korea
| | - Seong-Gi Kim
- Center for Neuroscience Imaging Research, Institute for Basic Science; Suwon, Republic of Korea
- Department of Biomedical Engineering, Sungkyunkwan University; Suwon, Republic of Korea
| | - Sung Han
- Peptide Biology Laboratory, The Salk Institute for Biological Studies; La Jolla, CA 92037, USA
- Center for Neuroscience Imaging Research, Institute for Basic Science; Suwon, Republic of Korea
- Department of Biomedical Engineering, Sungkyunkwan University; Suwon, Republic of Korea
| |
Collapse
|
9
|
Aoki I, Golinelli L, Dunkel E, Bhat S, Bassam E, Beets I, Gottschalk A. Hierarchical regulation of functionally antagonistic neuropeptides expressed in a single neuron pair. Nat Commun 2024; 15:9504. [PMID: 39489735 PMCID: PMC11532408 DOI: 10.1038/s41467-024-53899-7] [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: 12/19/2023] [Accepted: 10/22/2024] [Indexed: 11/05/2024] Open
Abstract
Neuronal communication involves small-molecule transmitters, gap junctions, and neuropeptides. While neurons often express multiple neuropeptides, our understanding of the coordination of their actions and their mutual interactions remains limited. Here, we demonstrate that two neuropeptides, NLP-10 and FLP-1, released from the same interneuron pair, AVKL/R, exert antagonistic effects on locomotion speed in Caenorhabditis elegans. NLP-10 accelerates locomotion by activating the G protein-coupled receptor NPR-35 on premotor interneurons that promote forward movement. Notably, we establish that NLP-10 is crucial for the aversive response to mechanical and noxious light stimuli. Conversely, AVK-derived FLP-1 slows down locomotion by suppressing the secretion of NLP-10 from AVK, through autocrine feedback via activation of its receptor DMSR-7 in AVK neurons. Our findings suggest that peptidergic autocrine motifs, exemplified by the interaction between NLP-10 and FLP-1, might represent a widespread mechanism in nervous systems across species. These mutual functional interactions among peptidergic co-transmitters could fine-tune brain activity.
Collapse
Affiliation(s)
- Ichiro Aoki
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue-Strasse 15, D-60438, Frankfurt, Germany.
- Department of Biochemistry, Chemistry and Pharmacy, Institute of Biophysical Chemistry, Goethe University, Frankfurt, Germany.
| | | | - Eva Dunkel
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue-Strasse 15, D-60438, Frankfurt, Germany
- Department of Biochemistry, Chemistry and Pharmacy, Institute of Biophysical Chemistry, Goethe University, Frankfurt, Germany
| | - Shripriya Bhat
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue-Strasse 15, D-60438, Frankfurt, Germany
| | - Erschad Bassam
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue-Strasse 15, D-60438, Frankfurt, Germany
| | - Isabel Beets
- Department of Biology, KU Leuven, Leuven, Belgium
| | - Alexander Gottschalk
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue-Strasse 15, D-60438, Frankfurt, Germany.
- Department of Biochemistry, Chemistry and Pharmacy, Institute of Biophysical Chemistry, Goethe University, Frankfurt, Germany.
| |
Collapse
|
10
|
Fan H, Liang X, Tang Y. Neuroscience in peripheral cancers: tumors hijacking nerves and neuroimmune crosstalk. MedComm (Beijing) 2024; 5:e784. [PMID: 39492832 PMCID: PMC11527832 DOI: 10.1002/mco2.784] [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: 05/21/2024] [Revised: 09/25/2024] [Accepted: 09/25/2024] [Indexed: 11/05/2024] Open
Abstract
Cancer neuroscience is an emerging field that investigates the intricate relationship between the nervous system and cancer, gaining increasing recognition for its importance. The central nervous system governs the development of the nervous system and directly affects brain tumors, and the peripheral nervous system (PNS) shapes the tumor microenvironment (TME) of peripheral tumors. Both systems are crucial in cancer initiation and progression, with recent studies revealing a more intricate role of the PNS within the TME. Tumors not only invade nerves but also persuade them through remodeling to further promote malignancy, creating a bidirectional interaction between nerves and cancers. Notably, immune cells also contribute to this communication, forming a triangular relationship that influences protumor inflammation and the effectiveness of immunotherapy. This review delves into the intricate mechanisms connecting the PNS and tumors, focusing on how various immune cell types influence nerve‒tumor interactions, emphasizing the clinical relevance of nerve‒tumor and nerve‒immune dynamics. By deepening our understanding of the interplay between nerves, cancer, and immune cells, this review has the potential to reshape tumor biology insights, inspire innovative therapies, and improve clinical outcomes for cancer patients.
Collapse
Affiliation(s)
- Hua‐Yang Fan
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Xin‐Hua Liang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Ya‐Ling Tang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Oral PathologyWest China Hospital of StomatologySichuan UniversityChengduChina
| |
Collapse
|
11
|
Kim DI, Park S, Park S, Ye M, Chen JY, Kang SJ, Jhang J, Hunker AC, Zweifel LS, Caron KM, Vaughan JM, Saghatelian A, Palmiter RD, Han S. Presynaptic sensor and silencer of peptidergic transmission reveal neuropeptides as primary transmitters in pontine fear circuit. Cell 2024; 187:5102-5117.e16. [PMID: 39043179 PMCID: PMC11380597 DOI: 10.1016/j.cell.2024.06.035] [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: 01/13/2023] [Revised: 11/17/2023] [Accepted: 06/25/2024] [Indexed: 07/25/2024]
Abstract
Neurons produce and release neuropeptides to communicate with one another. Despite their importance in brain function, circuit-based mechanisms of peptidergic transmission are poorly understood, primarily due to the lack of tools for monitoring and manipulating neuropeptide release in vivo. Here, we report the development of two genetically encoded tools for investigating peptidergic transmission in behaving mice: a genetically encoded large dense core vesicle (LDCV) sensor that detects presynaptic neuropeptide release and a genetically encoded silencer that specifically degrades neuropeptides inside LDCVs. Using these tools, we show that neuropeptides, not glutamate, encode the unconditioned stimulus in the parabrachial-to-amygdalar threat pathway during Pavlovian threat learning. We also show that neuropeptides play important roles in encoding positive valence and suppressing conditioned threat response in the amygdala-to-parabrachial endogenous opioidergic circuit. These results show that our sensor and silencer for presynaptic peptidergic transmission are reliable tools to investigate neuropeptidergic systems in awake, behaving animals.
Collapse
Affiliation(s)
- Dong-Il Kim
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Sekun Park
- Howard Hughes Medical Institute, Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Seahyung Park
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Mao Ye
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Jane Y Chen
- Howard Hughes Medical Institute, Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Sukjae J Kang
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Jinho Jhang
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Avery C Hunker
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Larry S Zweifel
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Kathleen M Caron
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Joan M Vaughan
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Alan Saghatelian
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Richard D Palmiter
- Howard Hughes Medical Institute, Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Sung Han
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon 16419, Republic of Korea; Department of Biomedical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
12
|
Hofbauer B, Zandawala M, Reinhard N, Rieger D, Werner C, Evers JF, Wegener C. The neuropeptide pigment-dispersing factor signals independently of Bruchpilot-labelled active zones in daily remodelled terminals of Drosophila clock neurons. Eur J Neurosci 2024; 59:2665-2685. [PMID: 38414155 DOI: 10.1111/ejn.16294] [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: 06/21/2023] [Revised: 02/06/2024] [Accepted: 02/08/2024] [Indexed: 02/29/2024]
Abstract
The small ventrolateral neurons (sLNvs) are key components of the central clock in the Drosophila brain. They signal via the neuropeptide pigment-dispersing factor (PDF) to align the molecular clockwork of different central clock neurons and to modulate downstream circuits. The dorsal terminals of the sLNvs undergo daily morphological changes that affect presynaptic sites organised by the active zone protein Bruchpilot (BRP), a homolog of mammalian ELKS proteins. However, the role of these presynaptic sites for PDF release is ill-defined. Here, we combined expansion microscopy with labelling of active zones by endogenously tagged BRP to examine the spatial correlation between PDF-containing dense-core vesicles and BRP-labelled active zones. We found that the number of BRP-labelled puncta in the sLNv terminals was similar while their density differed between Zeitgeber time (ZT) 2 and 14. The relative distance between BRP- and PDF-labelled puncta was increased in the morning, around the reported time of PDF release. Spontaneous dense-core vesicle release profiles of sLNvs in a publicly available ssTEM dataset (FAFB) consistently lacked spatial correlation to BRP-organised active zones. RNAi-mediated downregulation of brp and other active zone proteins expressed by the sLNvs did not affect PDF-dependent locomotor rhythmicity. In contrast, down-regulation of genes encoding proteins of the canonical vesicle release machinery, the dense-core vesicle-related protein CADPS, as well as PDF impaired locomotor rhythmicity. Taken together, our study suggests that PDF release from the sLNvs is independent of BRP-organised active zones, while BRP may be redistributed to active zones in a time-dependent manner.
Collapse
Affiliation(s)
- Benedikt Hofbauer
- Biocenter, Theodor-Boveri-Institute, Neurobiology and Genetics, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Meet Zandawala
- Biocenter, Theodor-Boveri-Institute, Neurobiology and Genetics, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
- Department of Biochemistry and Molecular Biology, University of Nevada Reno, Reno, NV, USA
| | - Nils Reinhard
- Biocenter, Theodor-Boveri-Institute, Neurobiology and Genetics, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Dirk Rieger
- Biocenter, Theodor-Boveri-Institute, Neurobiology and Genetics, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Christian Werner
- Biocenter, Theodor-Boveri-Institute, Department of Biotechnology and Biophysics, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Jan Felix Evers
- Centre for organismal studies COS, Universität Heidelberg, Heidelberg, Germany
- Cairn GmbH, Heidelberg, Germany
| | - Christian Wegener
- Biocenter, Theodor-Boveri-Institute, Neurobiology and Genetics, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| |
Collapse
|
13
|
Pop M, Klemke AL, Seidler L, Wernet N, Steudel PL, Baust V, Wohlmann E, Fischer R. Caenorhabditis elegans neuropeptide NLP-27 enhances neurodegeneration and paralysis in an opioid-like manner during fungal infection. iScience 2024; 27:109484. [PMID: 38784855 PMCID: PMC11112505 DOI: 10.1016/j.isci.2024.109484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 10/26/2023] [Accepted: 03/08/2024] [Indexed: 05/25/2024] Open
Abstract
The nervous system of metazoans is involved in host-pathogen interactions to control immune activation. In Caenorhabditis elegans, this includes sleep induction, mediated by neuropeptide-like proteins (NLPs), which increases the chance of survival after wounding. Here we analyzed the role of NLP-27 in the infection of C. elegans with the nematode-trapping fungus Arthrobotrys flagrans. Early responses of C. elegans were the upregulation of nlp-27, the induction of paralysis (sleep), and neurodegeneration of the mechanosensing PVD (Posterior Ventral Process D) neurons. Deletion of nlp-27 reduced neurodegeneration during fungal attack. Induction of nlp-27 was independent of the MAP kinase PMK-1, and expression of nlp-27 in the hypodermis was sufficient to induce paralysis, although NLP-27 was also upregulated in head neurons. NLP-27 contains the pentapeptide YGGYG sequence known to bind the human μ- and κ-type opioid receptors suggesting NLP-27 or peptides thereof act on opioid receptors. The opioid receptor antagonist naloxone shortened the paralysis time like overexpression of NLP-27.
Collapse
Affiliation(s)
- Maria Pop
- Karlsruhe Institute of Technology (KIT) - South Campus, Institute for Applied Biosciences, Department of Microbiology, Fritz-Haber-Weg 4, 76131 Karlsruhe, Germany
| | - Anna-Lena Klemke
- Karlsruhe Institute of Technology (KIT) - South Campus, Institute for Applied Biosciences, Department of Microbiology, Fritz-Haber-Weg 4, 76131 Karlsruhe, Germany
| | - Lena Seidler
- Karlsruhe Institute of Technology (KIT) - South Campus, Institute for Applied Biosciences, Department of Microbiology, Fritz-Haber-Weg 4, 76131 Karlsruhe, Germany
| | - Nicole Wernet
- Karlsruhe Institute of Technology (KIT) - South Campus, Institute for Applied Biosciences, Department of Microbiology, Fritz-Haber-Weg 4, 76131 Karlsruhe, Germany
| | - Pietrina Loredana Steudel
- Karlsruhe Institute of Technology (KIT) - South Campus, Institute for Applied Biosciences, Department of Microbiology, Fritz-Haber-Weg 4, 76131 Karlsruhe, Germany
| | - Vanessa Baust
- Karlsruhe Institute of Technology (KIT) - South Campus, Institute for Applied Biosciences, Department of Microbiology, Fritz-Haber-Weg 4, 76131 Karlsruhe, Germany
| | - Elke Wohlmann
- Karlsruhe Institute of Technology (KIT) - South Campus, Institute for Applied Biosciences, Department of Microbiology, Fritz-Haber-Weg 4, 76131 Karlsruhe, Germany
| | - Reinhard Fischer
- Karlsruhe Institute of Technology (KIT) - South Campus, Institute for Applied Biosciences, Department of Microbiology, Fritz-Haber-Weg 4, 76131 Karlsruhe, Germany
| |
Collapse
|
14
|
Chen D, Yuan Y, Huang Z, Wang Y. LH-Nts Neurons Regulate VTA Calcium Dynamics Via Releasing GABA and Nts. Neurosci Bull 2024; 40:550-552. [PMID: 38416271 PMCID: PMC11004095 DOI: 10.1007/s12264-024-01181-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 12/25/2023] [Indexed: 02/29/2024] Open
Affiliation(s)
- Danni Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
- Key Laboratory of Element Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Yinfeng Yuan
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
- Key Laboratory of Element Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Zhihui Huang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China.
- Key Laboratory of Element Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China.
| | - Yongjie Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China.
- Key Laboratory of Element Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China.
| |
Collapse
|
15
|
Mukherjee M, Mukherjee C, Ghosh V, Jain A, Sadhukhan S, Dagar S, Sahu BS. Endoplasmic reticulum stress impedes regulated secretion by governing key exocytotic and granulogenic molecular switches. J Cell Sci 2024; 137:jcs261257. [PMID: 38348894 DOI: 10.1242/jcs.261257] [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: 04/19/2023] [Accepted: 02/05/2024] [Indexed: 03/20/2024] Open
Abstract
Dense core vesicles (DCVs) and synaptic vesicles are specialised secretory vesicles in neurons and neuroendocrine cells, and abnormal release of their cargo is associated with various pathophysiologies. Endoplasmic reticulum (ER) stress and inter-organellar communication are also associated with disease biology. To investigate the functional status of regulated exocytosis arising from the crosstalk of a stressed ER and DCVs, ER stress was modelled in PC12 neuroendocrine cells using thapsigargin. DCV exocytosis was severely compromised in ER-stressed PC12 cells and was reversed to varying magnitudes by ER stress attenuators. Experiments with tunicamycin, an independent ER stressor, yielded similar results. Concurrently, ER stress also caused impaired DCV exocytosis in insulin-secreting INS-1 cells. Molecular analysis revealed blunted SNAP25 expression, potentially attributed to augmented levels of ATF4, an inhibitor of CREB that binds to the CREB-binding site. The effects of loss of function of ATF4 in ER-stressed cells substantiated this attribution. Our studies revealed severe defects in DCV exocytosis in ER-stressed cells for the first time, mediated by reduced levels of key exocytotic and granulogenic switches regulated via the eIF2α (EIF2A)-ATF4 axis.
Collapse
Affiliation(s)
- Mohima Mukherjee
- National Brain Research Centre, Manesar, Gurgaon, Haryana 122052, India
| | | | - Vinayak Ghosh
- National Brain Research Centre, Manesar, Gurgaon, Haryana 122052, India
| | - Aamna Jain
- National Brain Research Centre, Manesar, Gurgaon, Haryana 122052, India
| | - Souren Sadhukhan
- National Brain Research Centre, Manesar, Gurgaon, Haryana 122052, India
| | - Sushma Dagar
- National Brain Research Centre, Manesar, Gurgaon, Haryana 122052, India
| | | |
Collapse
|
16
|
Sun W, Jin T, Yang H, Li J, Tian Q, Gao J, Peng R, Zhang G, Zhang X. Alterations of serum neuropeptide levels and their relationship to cognitive impairment and psychopathology in male patients with chronic schizophrenia. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2024; 10:3. [PMID: 38172494 PMCID: PMC10851704 DOI: 10.1038/s41537-023-00425-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 12/07/2023] [Indexed: 01/05/2024]
Abstract
Serum neuropeptide levels may be linked to schizophrenia (SCZ) pathogenesis. This study aims to examine the relation between five serum neuropeptide levels and the cognition of patients with treatment-resistant schizophrenia (TRS), chronic stable schizophrenia (CSS), and in healthy controls (HC). Three groups were assessed: 29 TRS and 48 CSS patients who were hospitalized in regional psychiatric hospitals, and 53 HC. After the above participants were enrolled, we examined the Repeatable Battery for the Assessment of Neuropsychological Status (RBANS) and the blood serum levels of α-melanocyte stimulating hormone (α-MSH), β-endorphin (BE), neurotensin (NT), oxytocin (OT) and substance.P (S.P). Psychiatric symptoms in patients with SCZ were assessed with the Positive and Negative Syndrome Scale. SCZ patients performed worse than HC in total score and all subscales of the RBANS. The levels of the above five serum neuropeptides were significantly higher in SCZ than in HC. The levels of OT and S.P were significantly higher in CSS than in TRS patients. The α-MSH levels in TRS patients were significantly and negatively correlated with the language scores of RBANS. However, the BE and NT levels in CSS patients were significantly and positively correlated with the visuospatial/constructional scores of RBANS. Moreover, the interaction effect of NT and BE levels was positively associated with the visuospatial/constructional scores of RBANS. Therefore, abnormally increased serum neuropeptide levels may be associated with the physiology of SCZ, and may cause cognitive impairment and psychiatric symptoms, especially in patients with TRS.
Collapse
Affiliation(s)
- Wenxi Sun
- Suzhou Medical College of Soochow University, Suzhou, 215031, Jiangsu, China
- Psychiatry Department of Suzhou Guangji Hospital, Affiliated Guangji Hospital of Soochow University, Suzhou, 215137, Jiangsu, China
| | - Tingting Jin
- Psychiatry Department of Suzhou Guangji Hospital, Affiliated Guangji Hospital of Soochow University, Suzhou, 215137, Jiangsu, China
| | - Haidong Yang
- Department of Psychiatry, The Fourth People's Hospital of Lianyungang, The Affiliated KangDa College of Nanjing Medical University, Lianyungang, 222003, PR China
| | - Jin Li
- Psychiatry Department of Suzhou Guangji Hospital, Affiliated Guangji Hospital of Soochow University, Suzhou, 215137, Jiangsu, China
| | - Qing Tian
- Psychiatry Department of Suzhou Guangji Hospital, Affiliated Guangji Hospital of Soochow University, Suzhou, 215137, Jiangsu, China
| | - Ju Gao
- Psychiatry Department of Suzhou Guangji Hospital, Affiliated Guangji Hospital of Soochow University, Suzhou, 215137, Jiangsu, China
| | - Ruijie Peng
- Suzhou Medical College of Soochow University, Suzhou, 215031, Jiangsu, China
| | - Guangya Zhang
- Psychiatry Department of Suzhou Guangji Hospital, Affiliated Guangji Hospital of Soochow University, Suzhou, 215137, Jiangsu, China.
| | - Xiaobin Zhang
- Psychiatry Department of Suzhou Guangji Hospital, Affiliated Guangji Hospital of Soochow University, Suzhou, 215137, Jiangsu, China.
| |
Collapse
|
17
|
Liu D, Lin Z, Jia C. NeuroCNN_GNB: an ensemble model to predict neuropeptides based on a convolution neural network and Gaussian naive Bayes. Front Genet 2023; 14:1226905. [PMID: 37576553 PMCID: PMC10414792 DOI: 10.3389/fgene.2023.1226905] [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: 05/22/2023] [Accepted: 06/30/2023] [Indexed: 08/15/2023] Open
Abstract
Neuropeptides contain more chemical information than other classical neurotransmitters and have multiple receptor recognition sites. These characteristics allow neuropeptides to have a correspondingly higher selectivity for nerve receptors and fewer side effects. Traditional experimental methods, such as mass spectrometry and liquid chromatography technology, still need the support of a complete neuropeptide precursor database and the basic characteristics of neuropeptides. Incomplete neuropeptide precursor and information databases will lead to false-positives or reduce the sensitivity of recognition. In recent years, studies have proven that machine learning methods can rapidly and effectively predict neuropeptides. In this work, we have made a systematic attempt to create an ensemble tool based on four convolution neural network models. These baseline models were separately trained on one-hot encoding, AAIndex, G-gap dipeptide encoding and word2vec and integrated using Gaussian Naive Bayes (NB) to construct our predictor designated NeuroCNN_GNB. Both 5-fold cross-validation tests using benchmark datasets and independent tests showed that NeuroCNN_GNB outperformed other state-of-the-art methods. Furthermore, this novel framework provides essential interpretations that aid the understanding of model success by leveraging the powerful Shapley Additive exPlanation (SHAP) algorithm, thereby highlighting the most important features relevant for predicting neuropeptides.
Collapse
Affiliation(s)
- Di Liu
- Information Science and Technology College, Dalian Maritime University, Dalian, China
| | - Zhengkui Lin
- Information Science and Technology College, Dalian Maritime University, Dalian, China
| | - Cangzhi Jia
- School of Science, Dalian Maritime University, Dalian, China
| |
Collapse
|
18
|
Soden ME, Yee JX, Zweifel LS. Circuit coordination of opposing neuropeptide and neurotransmitter signals. Nature 2023; 619:332-337. [PMID: 37380765 PMCID: PMC10947507 DOI: 10.1038/s41586-023-06246-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 05/22/2023] [Indexed: 06/30/2023]
Abstract
Fast-acting neurotransmitters and slow, modulatory neuropeptides are co-released from neurons in the central nervous system, albeit from distinct synaptic vesicles1. The mechanisms of how co-released neurotransmitters and neuropeptides that have opposing actions-for example, stimulatory versus inhibitory-work together to exert control of neural circuit output remain unclear. This has been difficult to resolve owing to the inability to selectively isolate these signalling pathways in a cell- and circuit-specific manner. Here we developed a genetic-based anatomical disconnect procedure that utilizes distinct DNA recombinases to independently facilitate CRISPR-Cas9 mutagenesis2 of neurotransmitter- and neuropeptide-related genes in distinct cell types in two different brain regions simultaneously. We demonstrate that neurons within the lateral hypothalamus that produce the stimulatory neuropeptide neurotensin and the inhibitory neurotransmitter GABA (γ-aminobutyric acid) utilize these signals to coordinately activate dopamine-producing neurons of the ventral tegmental area. We show that GABA release from lateral hypothalamus neurotensin neurons inhibits GABA neurons within the ventral tegmental area, disinhibiting dopamine neurons and causing a rapid rise in calcium, whereas neurotensin directly generates a slow inactivating calcium signal in dopamine neurons that is dependent on the expression of neurotensin receptor 1 (Ntsr1). We further show that these two signals work together to regulate dopamine neuron responses to maximize behavioural responding. Thus, a neurotransmitter and a neuropeptide with opposing signals can act on distinct timescales through different cell types to enhance circuit output and optimize behaviour.
Collapse
Affiliation(s)
- Marta E Soden
- Department of Pharmacology, University of Washington, Seattle, WA, USA.
| | - Joshua X Yee
- Department of Pharmacology, University of Washington, Seattle, WA, USA
| | - Larry S Zweifel
- Department of Pharmacology, University of Washington, Seattle, WA, USA.
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA.
| |
Collapse
|
19
|
Cockx B, Van Bael S, Boelen R, Vandewyer E, Yang H, Le TA, Dalzell JJ, Beets I, Ludwig C, Lee J, Temmerman L. Mass Spectrometry-Driven Discovery of Neuropeptides Mediating Nictation Behavior of Nematodes. Mol Cell Proteomics 2023; 22:100479. [PMID: 36481452 PMCID: PMC9881375 DOI: 10.1016/j.mcpro.2022.100479] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/27/2022] [Accepted: 11/20/2022] [Indexed: 12/12/2022] Open
Abstract
Neuropeptides regulate animal physiology and behavior, making them widely studied targets of functional genetics research. While the field often relies on differential -omics approaches to build hypotheses, no such method exists for neuropeptidomics. It would nonetheless be valuable for studying behaviors suspected to be regulated by neuropeptides, especially when little information is otherwise available. This includes nictation, a phoretic strategy of Caenorhabditis elegans dauers that parallels host-finding strategies of infective juveniles of many pathogenic nematodes. We here developed a targeted peptidomics method for the model organism C. elegans and show that 161 quantified neuropeptides are more abundant in its dauer stage compared with L3 juveniles. Many of these have orthologs in the commercially relevant pathogenic nematode Steinernema carpocapsae, in whose infective juveniles, we identified 126 neuropeptides in total. Through further behavioral genetics experiments, we identify flp-7 and flp-11 as novel regulators of nictation. Our work advances knowledge on the genetics of nictation behavior and adds comparative neuropeptidomics as a tool to functional genetics workflows.
Collapse
Affiliation(s)
- Bram Cockx
- Animal Physiology & Neurobiology, Department of Biology, University of Leuven (KU Leuven), Leuven, Belgium
| | - Sven Van Bael
- Animal Physiology & Neurobiology, Department of Biology, University of Leuven (KU Leuven), Leuven, Belgium
| | - Rose Boelen
- Animal Physiology & Neurobiology, Department of Biology, University of Leuven (KU Leuven), Leuven, Belgium
| | - Elke Vandewyer
- Animal Physiology & Neurobiology, Department of Biology, University of Leuven (KU Leuven), Leuven, Belgium
| | - Heeseung Yang
- Department of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Tuan Anh Le
- Animal Physiology & Neurobiology, Department of Biology, University of Leuven (KU Leuven), Leuven, Belgium
| | - Johnathan J Dalzell
- School of Biological Sciences, Queen's University Belfast, Northern Ireland, United Kingdom
| | - Isabel Beets
- Animal Physiology & Neurobiology, Department of Biology, University of Leuven (KU Leuven), Leuven, Belgium
| | - Christina Ludwig
- Bavarian Center for Biomolecular Mass Spectrometry (BayBioMS), Technical University of Munich (TUM), Freising, Germany
| | - Junho Lee
- Department of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Liesbet Temmerman
- Animal Physiology & Neurobiology, Department of Biology, University of Leuven (KU Leuven), Leuven, Belgium.
| |
Collapse
|
20
|
Kim DI, Park S, Ye M, Chen JY, Jhang J, Hunker AC, Zweifel LS, Palmiter RD, Han S. Novel genetically encoded tools for imaging or silencing neuropeptide release from presynaptic terminals in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.19.524797. [PMID: 36712060 PMCID: PMC9882317 DOI: 10.1101/2023.01.19.524797] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Neurons produce and release neuropeptides to communicate with one another. Despite their profound impact on critical brain functions, circuit-based mechanisms of peptidergic transmission are poorly understood, primarily due to the lack of tools for monitoring and manipulating neuropeptide release in vivo. Here, we report the development of two genetically encoded tools for investigating peptidergic transmission in behaving mice: a genetically encoded large dense core vesicle (LDCV) sensor that detects the neuropeptides release presynaptically, and a genetically encoded silencer that specifically degrades neuropeptides inside the LDCV. Monitoring and silencing peptidergic and glutamatergic transmissions from presynaptic terminals using our newly developed tools and existing genetic tools, respectively, reveal that neuropeptides, not glutamate, are the primary transmitter in encoding unconditioned stimulus during Pavlovian threat learning. These results show that our sensor and silencer for peptidergic transmission are reliable tools to investigate neuropeptidergic systems in awake behaving animals.
Collapse
Affiliation(s)
- Dong-Il Kim
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Sekun Park
- Howard Hughes Medical Institute, Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Mao Ye
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Jane Y. Chen
- Howard Hughes Medical Institute, Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Jinho Jhang
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Avery C. Hunker
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Larry S. Zweifel
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Richard D. Palmiter
- Howard Hughes Medical Institute, Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Sung Han
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| |
Collapse
|
21
|
Aminopeptidase Activities Interact Asymmetrically between Brain, Plasma and Systolic Blood Pressure in Hypertensive Rats Unilaterally Depleted of Dopamine. Biomedicines 2022; 10:biomedicines10102457. [PMID: 36289718 PMCID: PMC9598709 DOI: 10.3390/biomedicines10102457] [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: 08/21/2022] [Revised: 09/19/2022] [Accepted: 09/22/2022] [Indexed: 11/17/2022] Open
Abstract
Brain dopamine, in relation to the limbic system, is involved in cognition and emotion. These functions are asymmetrically processed. Hypertension not only alters such functions but also their asymmetric brain pattern as well as their bilateral pattern of neurovisceral integration. The central and peripheral renin-angiotensin systems, particularly the aminopeptidases involved in its enzymatic cascade, play an important role in blood pressure control. In the present study, we report how these aminopeptidases from left and right cortico-limbic locations, plasma and systolic blood pressure interact among them in spontaneously hypertensive rats (SHR) unilaterally depleted of dopamine. The study comprises left and right sham and left and right lesioned (dopamine-depleted) rats as research groups. Results revealed important differences in the bilateral behavior comparing sham left versus sham right, lesioned left versus lesioned right, and sham versus lesioned animals. Results also suggest an important role for the asymmetrical functioning of the amygdala in cardiovascular control and an asymmetrical behavior in the interaction between the medial prefrontal cortex, hippocampus and amygdala with plasma, depending on the left or right depletion of dopamine. Compared with previous results of a similar study in Wistar-Kyoto (WKY) normotensive rats, the asymmetrical behaviors differ significantly between both WKY and SHR strains.
Collapse
|
22
|
Wu Y, Berisha A, Borniger JC. Neuropeptides in Cancer: Friend and Foe? Adv Biol (Weinh) 2022; 6:e2200111. [PMID: 35775608 DOI: 10.1002/adbi.202200111] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 05/31/2022] [Indexed: 01/28/2023]
Abstract
Neuropeptides are small regulatory molecules found throughout the body, most notably in the nervous, cardiovascular, and gastrointestinal systems. They serve as neurotransmitters or hormones in the regulation of diverse physiological processes. Cancer cells escape normal growth control mechanisms by altering their expression of growth factors, receptors, or intracellular signals, and neuropeptides have recently been recognized as mitogens in cancer growth and development. Many neuropeptides and their receptors exist in multiple subtypes, coupling with different downstream signaling pathways and playing distinct roles in cancer progression. The consideration of neuropeptide/receptor systems as anticancer targets is already leading to new biological and diagnostic knowledge that has the potential to enhance the understanding and treatment of cancer. In this review, recent discoveries regarding neuropeptides in a wide range of cancers, emphasizing their mechanisms of action, signaling cascades, regulation, and therapeutic potential, are discussed. Current technologies used to manipulate and analyze neuropeptides/receptors are described. Applications of neuropeptide analogs and their receptor inhibitors in translational studies and radio-oncology are rapidly increasing, and the possibility for their integration into therapeutic trials and clinical treatment appears promising.
Collapse
Affiliation(s)
- Yue Wu
- Cold Spring Harbor Laboratory, One Bungtown Rd, Cold Spring Harbor, NY, 11724, USA
| | - Adrian Berisha
- Cold Spring Harbor Laboratory, One Bungtown Rd, Cold Spring Harbor, NY, 11724, USA
| | - Jeremy C Borniger
- Cold Spring Harbor Laboratory, One Bungtown Rd, Cold Spring Harbor, NY, 11724, USA
| |
Collapse
|
23
|
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: 17] [Impact Index Per Article: 5.7] [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
|
24
|
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: 29] [Impact Index Per Article: 9.7] [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
|
25
|
Casello SM, Flores RJ, Yarur HE, Wang H, Awanyai M, Arenivar MA, Jaime-Lara RB, Bravo-Rivera H, Tejeda HA. Neuropeptide System Regulation of Prefrontal Cortex Circuitry: Implications for Neuropsychiatric Disorders. Front Neural Circuits 2022; 16:796443. [PMID: 35800635 PMCID: PMC9255232 DOI: 10.3389/fncir.2022.796443] [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: 10/16/2021] [Accepted: 04/27/2022] [Indexed: 01/08/2023] Open
Abstract
Neuropeptides, a diverse class of signaling molecules in the nervous system, modulate various biological effects including membrane excitability, synaptic transmission and synaptogenesis, gene expression, and glial cell architecture and function. To date, most of what is known about neuropeptide action is limited to subcortical brain structures and tissue outside of the central nervous system. Thus, there is a knowledge gap in our understanding of neuropeptide function within cortical circuits. In this review, we provide a comprehensive overview of various families of neuropeptides and their cognate receptors that are expressed in the prefrontal cortex (PFC). Specifically, we highlight dynorphin, enkephalin, corticotropin-releasing factor, cholecystokinin, somatostatin, neuropeptide Y, and vasoactive intestinal peptide. Further, we review the implication of neuropeptide signaling in prefrontal cortical circuit function and use as potential therapeutic targets. Together, this review summarizes established knowledge and highlights unknowns of neuropeptide modulation of neural function underlying various biological effects while offering insights for future research. An increased emphasis in this area of study is necessary to elucidate basic principles of the diverse signaling molecules used in cortical circuits beyond fast excitatory and inhibitory transmitters as well as consider components of neuropeptide action in the PFC as a potential therapeutic target for neurological disorders. Therefore, this review not only sheds light on the importance of cortical neuropeptide studies, but also provides a comprehensive overview of neuropeptide action in the PFC to serve as a roadmap for future studies in this field.
Collapse
Affiliation(s)
- Sanne M. Casello
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Rodolfo J. Flores
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Hector E. Yarur
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Huikun Wang
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Monique Awanyai
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Miguel A. Arenivar
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Rosario B. Jaime-Lara
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, United States
| | - Hector Bravo-Rivera
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Hugo A. Tejeda
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Hugo A. Tejeda,
| |
Collapse
|
26
|
Roles of Neuropeptides in Sleep-Wake Regulation. Int J Mol Sci 2022; 23:ijms23094599. [PMID: 35562990 PMCID: PMC9103574 DOI: 10.3390/ijms23094599] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/31/2022] [Accepted: 04/19/2022] [Indexed: 12/04/2022] Open
Abstract
Sleep and wakefulness are basic behavioral states that require coordination between several brain regions, and they involve multiple neurochemical systems, including neuropeptides. Neuropeptides are a group of peptides produced by neurons and neuroendocrine cells of the central nervous system. Like traditional neurotransmitters, neuropeptides can bind to specific surface receptors and subsequently regulate neuronal activities. For example, orexin is a crucial component for the maintenance of wakefulness and the suppression of rapid eye movement (REM) sleep. In addition to orexin, melanin-concentrating hormone, and galanin may promote REM sleep. These results suggest that neuropeptides play an important role in sleep–wake regulation. These neuropeptides can be divided into three categories according to their effects on sleep–wake behaviors in rodents and humans. (i) Galanin, melanin-concentrating hormone, and vasoactive intestinal polypeptide are sleep-promoting peptides. It is also noticeable that vasoactive intestinal polypeptide particularly increases REM sleep. (ii) Orexin and neuropeptide S have been shown to induce wakefulness. (iii) Neuropeptide Y and substance P may have a bidirectional function as they can produce both arousal and sleep-inducing effects. This review will introduce the distribution of various neuropeptides in the brain and summarize the roles of different neuropeptides in sleep–wake regulation. We aim to lay the foundation for future studies to uncover the mechanisms that underlie the initiation, maintenance, and end of sleep–wake states.
Collapse
|
27
|
Xia X, Wang Y, Qin Y, Zhao S, Zheng JC. Exosome: A novel neurotransmission modulator or non-canonical neurotransmitter? Ageing Res Rev 2022; 74:101558. [PMID: 34990846 DOI: 10.1016/j.arr.2021.101558] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 08/13/2021] [Accepted: 12/30/2021] [Indexed: 02/08/2023]
Abstract
Neurotransmission is the electrical impulse-triggered propagation of signals between neurons or between neurons and other cell types such as skeletal muscle cells. Recent studies point out the involvement of exosomes, a type of small bilipid layer-enclosed extracellular vesicles, in regulating neurotransmission. Through horizontally transferring proteins, lipids, and nucleic acids, exosomes can modulate synaptic activities rapidly by controlling neurotransmitter release or progressively by regulating neural plasticity including synapse formation, neurite growth & removal, and axon guidance & elongation. In this review, we summarize the similarities and differences between exosomes and synaptic vesicles in their biogenesis, contents, and release. We also highlight the recent progress made in demonstrating the biological roles of exosome in regulating neurotransmission, and propose a modified model of neurotransmission, in which exosomes act as novel neurotransmitters. Lastly, we provide a comprehensive discussion of the enlightenment of the current knowledge on neurotransmission to the future directions of exosome research.
Collapse
|
28
|
Sharma A, Ren X, Zhang H, Pandey GN. Effect of depression and suicidal behavior on neuropeptide Y (NPY) and its receptors in the adult human brain: A postmortem study. Prog Neuropsychopharmacol Biol Psychiatry 2022; 112:110428. [PMID: 34411658 PMCID: PMC8489679 DOI: 10.1016/j.pnpbp.2021.110428] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 08/08/2021] [Accepted: 08/10/2021] [Indexed: 01/12/2023]
Abstract
Neuropeptides are small proteinaceous molecules (3-100 amino acids) that are secreted by neurons and act on both neuronal and non-neuronal cells. Neuropeptide Y (NPY), a highly conserved and expressed neuropeptide in the central nervous system of mammals, plays a major role in stress response and resilience. Increasing evidence suggests that NPY and its receptors are altered in depression and suicide, pointing to their antidepressant-like nature. The objective of this study was to examine the role of NPY system in depression and suicidal behavior. Expression of NPY and its four receptors, NPY1R, NPY2R, NPY4R, and NPY5R was studied at the transcriptional and translational levels in the prefrontal cortex (PFC) and hippocampus regions of the postmortem brain of normal control (NC) (n = 24) and depressed suicide (DS) (n = 24) subjects. We observed a significant decrease in NPY mRNA and upregulation in NPY1R and NPY2R mRNA in both brain regions of DS subjects compared with NC subjects. We also observed a significant decrease in NPY protein expression in the PFC of subjects with DS. This study provides the first detailed evidence of alterations in the NPY system and the associated stress response in depression and suicidal behavior in humans. The outcomes of this study could be applied in the development of novel NPY system-targeted approaches for the treatment of depression.
Collapse
Affiliation(s)
| | | | | | - Ghanshyam N. Pandey
- Corresponding Author: Ghanshyam N. Pandey, Ph.D., University of Illinois at Chicago, 1601 West Taylor Street, Chicago, IL 60612, USA, Phone (312) 413-4540, Fax: (312) 413-4547,
| |
Collapse
|
29
|
Saklani P, Khan H, Gupta S, Kaur A, Singh TG. Neuropeptides: Potential neuroprotective agents in ischemic injury. Life Sci 2022; 288:120186. [PMID: 34852271 DOI: 10.1016/j.lfs.2021.120186] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/22/2021] [Accepted: 11/24/2021] [Indexed: 12/24/2022]
Abstract
AIM Ischemic damage to the brain is linked to an increased rate of morbidity and mortality worldwide. In certain parts of the world, it remains a leading cause of mortality and the primary cause of long-term impairment. Ischemic injury is exacerbated when particular neuropeptides are removed, or their function in the brain is blocked, whereas supplying such neuropeptides lowers ischemic harm. Here, we have discussed the role of neuropeptides in ischemic injury. MATERIALS & METHODS Numerous neuropeptides had their overexpression following cerebral ischemia. Neuropeptides such as NPY, CGRP, CART, SP, BK, PACAP, oxytocin, nociception, neurotensin and opioid peptides act as transmitters, documented in several "in vivo" and "in vitro" studies. Neuropeptides provide neuroprotection by activating the survival pathways or inhibiting the death pathways, i.e., MAPK, BDNF, Nitric Oxide, PI3k/Akt and NF-κB. KEY FINDINGS Neuropeptides have numerous beneficial effects in ischemic models, including antiapoptotic, anti-inflammatory, and antioxidant actions that provide a powerful protective impact in neurons when combined. These innovative therapeutic substances have the potential to treat ischemia injury due to their pleiotropic modes of action. SIGNIFICANCE This review emphasizes the neuroprotective role of neuropeptides in ischemic injury via modulation of various signalling pathways i.e., MAPK, BDNF, Nitric Oxide, PI3k/Akt and NF-κB.
Collapse
Affiliation(s)
- Priyanka Saklani
- Chitkara College of Pharmacy, Chitkara University, 140401, Punjab, India
| | - Heena Khan
- Chitkara College of Pharmacy, Chitkara University, 140401, Punjab, India
| | - Saurabh Gupta
- Chitkara College of Pharmacy, Chitkara University, 140401, Punjab, India
| | - Amarjot Kaur
- Chitkara College of Pharmacy, Chitkara University, 140401, Punjab, India
| | | |
Collapse
|
30
|
Britz S, Markert SM, Witvliet D, Steyer AM, Tröger S, Mulcahy B, Kollmannsberger P, Schwab Y, Zhen M, Stigloher C. Structural Analysis of the Caenorhabditis elegans Dauer Larval Anterior Sensilla by Focused Ion Beam-Scanning Electron Microscopy. Front Neuroanat 2021; 15:732520. [PMID: 34819841 PMCID: PMC8607169 DOI: 10.3389/fnana.2021.732520] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 09/24/2021] [Indexed: 11/13/2022] Open
Abstract
At the end of the first larval stage, the nematode Caenorhabditis elegans developing in harsh environmental conditions is able to choose an alternative developmental path called the dauer diapause. Dauer larvae exhibit different physiology and behaviors from non-dauer larvae. Using focused ion beam-scanning electron microscopy (FIB-SEM), we volumetrically reconstructed the anterior sensory apparatus of C. elegans dauer larvae with unprecedented precision. We provide a detailed description of some neurons, focusing on structural details that were unknown or unresolved by previously published studies. They include the following: (1) dauer-specific branches of the IL2 sensory neurons project into the periphery of anterior sensilla and motor or putative sensory neurons at the sub-lateral cords; (2) ciliated endings of URX sensory neurons are supported by both ILso and AMso socket cells near the amphid openings; (3) variability in amphid sensory dendrites among dauers; and (4) somatic RIP interneurons maintain their projection into the pharyngeal nervous system. Our results support the notion that dauer larvae structurally expand their sensory system to facilitate searching for more favorable environments.
Collapse
Affiliation(s)
- Sebastian Britz
- Imaging Core Facility of the Biocenter, Theodor-Boveri-Institute, Julius-Maximilians-University, Würzburg, Germany
| | - Sebastian Matthias Markert
- Imaging Core Facility of the Biocenter, Theodor-Boveri-Institute, Julius-Maximilians-University, Würzburg, Germany
| | - Daniel Witvliet
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Molecular Genetics, Physiology and Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Anna Maria Steyer
- European Molecular Biology Laboratory, Cell Biology and Biophysics Unit, Heidelberg, Germany
| | - Sarah Tröger
- Imaging Core Facility of the Biocenter, Theodor-Boveri-Institute, Julius-Maximilians-University, Würzburg, Germany
| | - Ben Mulcahy
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Molecular Genetics, Physiology and Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Philip Kollmannsberger
- Center for Computational and Theoretical Biology, Julius-Maximilians-University, Würzburg, Germany
| | - Yannick Schwab
- European Molecular Biology Laboratory, Cell Biology and Biophysics Unit, Heidelberg, Germany
| | - Mei Zhen
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Molecular Genetics, Physiology and Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Christian Stigloher
- Imaging Core Facility of the Biocenter, Theodor-Boveri-Institute, Julius-Maximilians-University, Würzburg, Germany
| |
Collapse
|
31
|
Cifuentes F, Morales MA. Functional Implications of Neurotransmitter Segregation. Front Neural Circuits 2021; 15:738516. [PMID: 34720888 PMCID: PMC8548464 DOI: 10.3389/fncir.2021.738516] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/21/2021] [Indexed: 11/13/2022] Open
Abstract
Here, we present and discuss the characteristics and properties of neurotransmitter segregation, a subtype of neurotransmitter cotransmission. We review early evidence of segregation and discuss its properties, such as plasticity, while placing special emphasis on its probable functional implications, either in the central nervous system (CNS) or the autonomic nervous system. Neurotransmitter segregation is a process by which neurons separately route transmitters to independent and distant or to neighboring neuronal processes; it is a plastic phenomenon that changes according to synaptic transmission requirements and is regulated by target-derived signals. Distant neurotransmitter segregation in the CNS has been shown to be related to an autocrine/paracrine function of some neurotransmitters. In retinal amacrine cells, segregation of acetylcholine (ACh) and GABA, and glycine and glutamate to neighboring terminals has been related to the regulation of the firing rate of direction-selective ganglion cells. In the rat superior cervical ganglion, segregation of ACh and GABA to neighboring varicosities shows a heterogeneous regional distribution, which is correlated to a similar regional distribution in transmission strength. We propose that greater segregation of ACh and GABA produces less GABAergic inhibition, strengthening ganglionic transmission. Segregation of ACh and GABA varies in different physiopathological conditions; specifically, segregation increases in acute sympathetic hyperactivity that occurs in cold stress, does not vary in chronic hyperactivity that occurs in hypertension, and rises in early ages of normotensive and hypertensive rats. Given this, we propose that variations in the extent of transmitter segregation may contribute to the alteration of neural activity that occurs in some physiopathological conditions and with age.
Collapse
Affiliation(s)
- Fredy Cifuentes
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Miguel Angel Morales
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
32
|
Carozzi VA, Salio C, Rodriguez-Menendez V, Ciglieri E, Ferrini F. 2D <em>vs</em> 3D morphological analysis of dorsal root ganglia in health and painful neuropathy. Eur J Histochem 2021; 65. [PMID: 34664808 PMCID: PMC8547168 DOI: 10.4081/ejh.2021.3276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/16/2021] [Indexed: 11/23/2022] Open
Abstract
Dorsal root ganglia (DRGs) are clusters of sensory neurons that transmit the sensory information from the periphery to the central nervous system, and satellite glial cells (SGCs), their supporting trophic cells. Sensory neurons are pseudounipolar neurons with a heterogeneous neurochemistry reflecting their functional features. DRGs, not protected by the blood brain barrier, are vulnerable to stress and damage of different origin (i.e., toxic, mechanical, metabolic, genetic) that can involve sensory neurons, SGCs or, considering their intimate intercommunication, both cell populations. DRG damage, primary or secondary to nerve damage, produces a sensory peripheral neuropathy, characterized by neurophysiological abnormalities, numbness, paraesthesia and dysesthesia, tingling and burning sensations and neuropathic pain. DRG stress can be morphologically detected by light and electron microscope analysis with alterations in cell size (swelling/atrophy) and in different subcellular compartments (i.e., mitochondria, endoplasmic reticulum, and nucleus) of neurons and/or SGCs. In addition, neurochemical changes can be used to portray abnormalities of neurons and SGC. Conventional immunostaining, i.e., immunohistochemical detection of specific molecules in tissue slices, can be employed to detect, localize and quantify particular markers of damage in neurons (i.e., nuclear expression of ATF3) or SGCs (i.e., increased expression of GFAP), markers of apoptosis (i.e., caspases), markers of mitochondrial suffering and oxidative stress (i.e., 8-OHdG), markers of tissue inflammation (i.e., CD68 for macrophage infiltration) etc. However classical (2D) methods of immunostaining disrupt the overall organization of the DRG, thus resulting in the loss of some crucial information. Whole-mount (3D) methods have been recently developed to investigate DRG morphology and neurochemistry without tissue slicing, giving the opportunity to study the intimate relationship between SGCs and sensory neurons in health and disease. Here, we aim to compare classical (2D) vs whole-mount (3D) approaches to highlight “pros” and “cons” of the two methodologies when analysing neuropathy-induced alterations in DRGs.
Collapse
Affiliation(s)
- Valentina Alda Carozzi
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza (MB).
| | - Chiara Salio
- Department of Veterinary Sciences, University of Turin, Grugliasco (TO).
| | | | | | - Francesco Ferrini
- Department of Veterinary Sciences, University of Turin, Grugliasco (TO).
| |
Collapse
|
33
|
Guillaumin MCC, Burdakov D. Neuropeptides as Primary Mediators of Brain Circuit Connectivity. Front Neurosci 2021; 15:644313. [PMID: 33776641 PMCID: PMC7991401 DOI: 10.3389/fnins.2021.644313] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 02/18/2021] [Indexed: 11/21/2022] Open
Abstract
Across sleep and wakefulness, brain function requires inter-neuronal interactions lasting beyond seconds. Yet, most studies of neural circuit connectivity focus on millisecond-scale interactions mediated by the classic fast transmitters, GABA and glutamate. In contrast, neural circuit roles of the largest transmitter family in the brain–the slow-acting peptide transmitters–remain relatively overlooked, or described as “modulatory.” Neuropeptides may efficiently implement sustained neural circuit connectivity, since they are not rapidly removed from the extracellular space, and their prolonged action does not require continuous presynaptic firing. From this perspective, we review actions of evolutionarily-conserved neuropeptides made by brain-wide-projecting hypothalamic neurons, focusing on lateral hypothalamus (LH) neuropeptides essential for stable consciousness: the orexins/hypocretins. Action potential-dependent orexin release inside and outside the hypothalamus evokes slow postsynaptic excitation. This excitation does not arise from modulation of classic neurotransmission, but involves direct action of orexins on their specific G-protein coupled receptors (GPCRs) coupled to ion channels. While millisecond-scale, GABA/glutamate connectivity within the LH may not be strong, re-assessing LH microcircuits from the peptidergic viewpoint is consistent with slow local microcircuits. The sustained actions of neuropeptides on neuronal membrane potential may enable core brain functions, such as temporal integration and the creation of lasting permissive signals that act as “eligibility traces” for context-dependent information routing and plasticity. The slowness of neuropeptides has unique advantages for efficient neuronal processing and feedback control of consciousness.
Collapse
Affiliation(s)
| | - Denis Burdakov
- Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland
| |
Collapse
|
34
|
Higuchi-Sanabria R, Durieux J, Kelet N, Homentcovschi S, de Los Rios Rogers M, Monshietehadi S, Garcia G, Dallarda S, Daniele JR, Ramachandran V, Sahay A, Tronnes SU, Joe L, Dillin A. Divergent Nodes of Non-autonomous UPR ER Signaling through Serotonergic and Dopaminergic Neurons. Cell Rep 2020; 33:108489. [PMID: 33296657 DOI: 10.1016/j.celrep.2020.108489] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/20/2020] [Accepted: 11/12/2020] [Indexed: 01/01/2023] Open
Abstract
In multicellular organisms, neurons integrate a diverse array of external cues to affect downstream changes in organismal health. Specifically, activation of the endoplasmic reticulum (ER) unfolded protein response (UPRER) in neurons increases lifespan by preventing age-onset loss of ER proteostasis and driving lipid depletion in a cell non-autonomous manner. The mechanism of this communication is dependent on the release of small clear vesicles from neurons. We find dopaminergic neurons are necessary and sufficient for activation of cell non-autonomous UPRER to drive lipid depletion in peripheral tissues, whereas serotonergic neurons are sufficient to drive protein homeostasis in peripheral tissues. These signaling modalities are unique and independent and together coordinate the beneficial effects of neuronal cell non-autonomous ER stress signaling upon health and longevity.
Collapse
Affiliation(s)
- Ryo Higuchi-Sanabria
- Department of Molecular & Cellular Biology, Howard Hughes Medical Institute, The University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jenni Durieux
- Department of Molecular & Cellular Biology, Howard Hughes Medical Institute, The University of California, Berkeley, Berkeley, CA 94720, USA
| | - Naame Kelet
- Department of Molecular & Cellular Biology, Howard Hughes Medical Institute, The University of California, Berkeley, Berkeley, CA 94720, USA
| | - Stefan Homentcovschi
- Department of Molecular & Cellular Biology, Howard Hughes Medical Institute, The University of California, Berkeley, Berkeley, CA 94720, USA
| | - Mattias de Los Rios Rogers
- Department of Molecular & Cellular Biology, Howard Hughes Medical Institute, The University of California, Berkeley, Berkeley, CA 94720, USA
| | - Samira Monshietehadi
- Department of Molecular & Cellular Biology, Howard Hughes Medical Institute, The University of California, Berkeley, Berkeley, CA 94720, USA
| | - Gilberto Garcia
- Department of Molecular & Cellular Biology, Howard Hughes Medical Institute, The University of California, Berkeley, Berkeley, CA 94720, USA
| | - Sofia Dallarda
- Department of Molecular & Cellular Biology, Howard Hughes Medical Institute, The University of California, Berkeley, Berkeley, CA 94720, USA
| | - Joseph R Daniele
- TRACTION, The University of Texas MD Anderson Cancer Center, South Campus Research, Houston, TX 77054, USA
| | - Vidhya Ramachandran
- Department of Molecular & Cellular Biology, Howard Hughes Medical Institute, The University of California, Berkeley, Berkeley, CA 94720, USA
| | - Arushi Sahay
- Department of Molecular & Cellular Biology, Howard Hughes Medical Institute, The University of California, Berkeley, Berkeley, CA 94720, USA
| | - Sarah U Tronnes
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Boulder, CO 80309, USA
| | - Larry Joe
- Department of Molecular & Cellular Biology, Howard Hughes Medical Institute, The University of California, Berkeley, Berkeley, CA 94720, USA
| | - Andrew Dillin
- Department of Molecular & Cellular Biology, Howard Hughes Medical Institute, The University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
35
|
Kotsyuba E, Dyachuk V. Localization of neurons expressing choline acetyltransferase, serotonin and/or FMRFamide in the central nervous system of the decapod shore crab Hemigrapsus sanguineus. Cell Tissue Res 2020; 383:959-977. [PMID: 33237479 DOI: 10.1007/s00441-020-03309-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 09/22/2020] [Indexed: 12/22/2022]
Abstract
Although it is now established that neurons in crustacea contain multiple transmitter substances, little is know about patterns of expression and co-expression or about the functional effects of such co-transmission. The present study was designed to characterize the distributions and potential colocalization of choline acetyltransferase (ChAT), serotonin (5-HT) and neuropeptide H-Phe-Met-Arg-Phe-NH2 (FMRFamide) in the central nervous system (CNS) of the Asian shore crab, Hemigrapsus sanguineus using immunohistochemical analyses in combination with laser scanning confocal microscopy. ChAT was found to be expressed by small, medium-sized, and large neurons in all regions of the brain and ventral nerve cord (VNC). For the most part, ChAT, FMRFamide, and 5-HT are expressed in different neurons, although some colocalization of ChAT- with FMRFamide- or 5-HT-LIR is observed in small and medium-sized cells, mostly neurons that immunostain only weakly. In the brain, such double immunolabeling is observed primarily in neurons of the protocerebrum and, to a particularly great extent, in local olfactory interneurons of the deutocerebrum. The clusters of neurons in the VNC that stain most intensely for ChAT, FMRFamide, and 5-HT, with colocalization in some cases, are located in the subesophageal ganglia. This colocalization appears to be related to function, since it is present in regions of the CNS characterized by multiple afferent projections and outputs to a variety of functionally related centers involved in various physiological and behavioral processes. Further elucidation of the functional significance of these neurons and of the widespread process of co-transmission in the crustaceans should provide fascinating new insights.
Collapse
Affiliation(s)
- Elena Kotsyuba
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok, 690041, Russia
| | - Vyacheslav Dyachuk
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok, 690041, Russia.
| |
Collapse
|
36
|
Beck J, Kressel M. FERM domain-containing protein 6 identifies a subpopulation of varicose nerve fibers in different vertebrate species. Cell Tissue Res 2020; 381:13-24. [PMID: 32200438 PMCID: PMC7306050 DOI: 10.1007/s00441-020-03189-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 02/16/2020] [Indexed: 12/29/2022]
Abstract
FERM domain-containing protein 6 (FRMD6) is a member of the FERM protein superfamily, which is evolutionary highly conserved and has recently been identified as an upstream regulator of the conserved growth-promoting Hippo signaling pathway. In clinical studies, the FRMD6 gene is correlated with high significance to Alzheimer's disease and cognitive impairment implicating a wider role of this protein in the nervous system. Scare data are available on the localization of endogenous FRMD6 in neural tissues. Using a FRMD6-directed antiserum, we detected specific immunoreactivity in varicose nerve fibers in the rat central and peripheral nervous system. FRMD6-immunoreactive (-ir) neurons were found in the sensory ganglia of cranial nerves, which were marked by a pool of labeled cytoplasmic granules. Cross-species comparative studies detected a morphologically identical fiber population and a comparable fiber distribution in tissues from xenopus and human cranial nerves and ganglia. In the spinal cord, FRMD6-ir was detectable in the terminal endings of primary afferent neurons containing substance P (SP). In the rat diencephalon, FRMD6-ir was co-localized with either SP- or arginine vasopressin-positive fibers in Broca's diagonal band and the lateral septum. Dense fiber terminals containing both FRMD6-ir and growth hormone-releasing hormone were found in the median eminence. The intimate association of FRMD6 with secretory vesicles was investigated in vitro. Induction of exocytotic vesicles in cultured cells by ectopic expression of the SP precursor molecule preprotachykinin A led to a redistribution and co-localization of endogenous FRMD6 with secretory granules closely mimicking the observations in tissues.
Collapse
Affiliation(s)
- Josefa Beck
- Institute of Anatomy and Cell Biology, University of Erlangen, Krankenhausstr. 9, 91054, Erlangen, Germany
| | - Michael Kressel
- Institute of Anatomy and Cell Biology, University of Erlangen, Krankenhausstr. 9, 91054, Erlangen, Germany.
| |
Collapse
|
37
|
Senior EE, Poulin HE, Dobecki MG, Anair BM, Fabian-Fine R. Co-expression of the neuropeptide proctolin and glutamate in the central nervous system, along mechanosensory neurons and leg muscle in Cupiennius salei. Cell Tissue Res 2020; 382:281-292. [PMID: 32556729 DOI: 10.1007/s00441-020-03217-6] [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: 01/17/2020] [Accepted: 04/07/2020] [Indexed: 11/24/2022]
Abstract
Similar to hair cells in the mammalian cochlear system, mechanosensory neurons in the Central American wandering spider Cupiennius salei are strongly innervated by efferent fibers that originate from neurons whose somata are located in the central nervous system (CNS). In both the mammalian and arachnid systems, efferent fibers have been shown to co-express two or more transmitters; however, our understanding regarding co-transmission and how it affects sensory signal transduction and processing in these systems is only fragmentary. The spider model system is exceptionally suitable for this type of investigation due to the large size and easy accessibility of the sensory and efferent neurons in this system. Thus far, GABA and glutamate have been identified as the main fast-acting transmitters in efferent axons that form synaptic contacts onto sensory neurons in slit sense organs. Ultrastructural investigations suggest an abundance of neuropeptides within these peripheral synapses. In an effort to identify these peptides and conduct functional studies, we have employed immunohistochemistry to investigate whether the neuropeptide proctolin is present in neurons of the leg ganglia and in peripheral leg structures. Here, we demonstrate that ~ 73% of all neurons in the CNS of C. salei show proctolin-like immunoreactivity (proc-LIR) including the leg ganglia. We demonstrate that both strongly and weakly labeled neurons can be distinguished. The majority of proc-LIR neurons show weak labeling intensity and ~ 86.2% co-localize with glutamate. In future experiments, we plan to undertake functional studies to investigate the significance of this co-expression, which has yet to be investigated.
Collapse
Affiliation(s)
- Elizabeth E Senior
- Department of Biology, Saint Michael's College, One Winooski Park, Colchester, VT, 05349, USA
| | - Hailee E Poulin
- Department of Biology, Saint Michael's College, One Winooski Park, Colchester, VT, 05349, USA
| | - Madison G Dobecki
- Department of Biology, Saint Michael's College, One Winooski Park, Colchester, VT, 05349, USA
| | - Bradley M Anair
- Department of Biology, Saint Michael's College, One Winooski Park, Colchester, VT, 05349, USA
| | - Ruth Fabian-Fine
- Department of Biology, Saint Michael's College, One Winooski Park, Colchester, VT, 05349, USA.
| |
Collapse
|
38
|
Dereli AS, Yaseen Z, Carrive P, Kumar NN. Adaptation of Respiratory-Related Brain Regions to Long-Term Hypercapnia: Focus on Neuropeptides in the RTN. Front Neurosci 2019; 13:1343. [PMID: 31920508 PMCID: PMC6923677 DOI: 10.3389/fnins.2019.01343] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 11/28/2019] [Indexed: 12/21/2022] Open
Abstract
Long-term hypercapnia is associated with respiratory conditions including obstructive sleep apnea, chronic obstructive pulmonary disease and obesity hypoventilation syndrome. Animal studies have demonstrated an initial (within hours) increase in ventilatory drive followed by a decrease in this response over the long-term (days–weeks) in response hypercapnia. Little is known about whether changes in the central respiratory chemoreflex are involved. Here we investigated whether central respiratory chemoreceptor neurons of the retrotrapezoid nucleus (RTN), which project to the respiratory pattern generator within the ventral respiratory column (VRC) have a role in the mechanism of neuroplasticity associated with long-term hypercapnia. Adult male C57BL/6 mice (n = 5/group) were used. Our aims were (1) to determine if galanin, neuromedin B and gastrin-releasing peptide gene expression is altered in the RTN after long-term hypercapnia. This was achieved using qPCR to measure mRNA expression changes of neuropeptides in the RTN after short-term hypercapnia (6 or 8 h, 5 or 8% CO2) or long-term hypercapnia exposure (10 day, 5 or 8% CO2), (2) in the mouse brainstem, to determine the distribution of preprogalanin in chemoreceptors, and the co-occurrence of the galanin receptor 1 (GalR1:Gi-coupled receptor) with inhibitory GlyT2 ventral respiratory column neurons using in situ hybridization (ISH) to better characterize galaninergic RTN-VRC circuitry, (3) to investigate whether long-term hypercapnia causes changes to recruitment (detected by cFos immunohistochemistry) of respiratory related neural populations including the RTN neurons and their galaninergic subset, in vivo. Collectively, we found that hypercapnia decreases neuropeptide expression in the RTN in the short-term and has the opposite effect over the long-term. Following long term hypercapnia, the number of RTN galanin neurons remains unchanged, and their responsiveness to acute chemoreflex is sustained; in contrast, we identified multiple respiratory related sites that exhibit blunted chemoreflex activation. GalR1 was distributed in 11% of preBötC and 30% of BötC glycinergic neurons. Our working hypothesis is that during long-term hypercapnia, galanin co-release from RTN neurons may counterbalance glutamatergic inputs to respiratory centers to downscale energetically wasteful hyperventilation, thereby having a role in neuroplasticity by contributing to a decrease in ventilation, through the inhibitory effects of galanin.
Collapse
Affiliation(s)
- Ayse Sumeyra Dereli
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Zarwa Yaseen
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Pascal Carrive
- Department of Anatomy, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Natasha N Kumar
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
39
|
Rodríguez B, Nani JV, Almeida PGC, Brietzke E, Lee RS, Hayashi MAF. Neuropeptides and oligopeptidases in schizophrenia. Neurosci Biobehav Rev 2019; 108:679-693. [PMID: 31794779 DOI: 10.1016/j.neubiorev.2019.11.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 11/14/2019] [Accepted: 11/27/2019] [Indexed: 12/30/2022]
Abstract
Schizophrenia (SCZ) is a complex psychiatric disorder with severe impact on patient's livelihood. In the last years, the importance of neuropeptides in SCZ and other CNS disorders has been recognized, mainly due to their ability to modulate the signaling of classical monoaminergic neurotransmitters as dopamine. In addition, a class of enzymes coined as oligopeptidases are able to cleave several of these neuropeptides, and their potential implication in SCZ was also demonstrated. Interestingly, these enzymes are able to play roles as modulators of neuropeptidergic systems, and they were also implicated in neurogenesis, neurite outgrowth, neuron migration, and therefore, in neurodevelopment and brain formation. Altered activity of oligopeptidases in SCZ was described only more recently, suggesting their possible utility as biomarkers for mental disorders diagnosis or treatment response. We provide here an updated and comprehensive review on neuropeptides and oligopeptidases involved in mental disorders, aiming to attract the attention of physicians to the potential of targeting this system for improving the therapy and for understanding the neurobiology underlying mental disorders as SCZ.
Collapse
Affiliation(s)
- Benjamín Rodríguez
- Departamento de Farmacologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - João Victor Nani
- Departamento de Farmacologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil; National Institute for Translational Medicine (INCT-TM, CNPq/FAPESP/CAPES), Ribeirão Preto, Brazil
| | - Priscila G C Almeida
- Departamento de Farmacologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Elisa Brietzke
- Department of Psychiatry, Queen's University School of Medicine, Kingston, ON, Canada
| | - Richard S Lee
- Department of Psychiatry, Johns Hopkins University, Baltimore, MD, USA
| | - Mirian A F Hayashi
- Departamento de Farmacologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil; National Institute for Translational Medicine (INCT-TM, CNPq/FAPESP/CAPES), Ribeirão Preto, Brazil.
| |
Collapse
|
40
|
Nässel DR, Zandawala M. Recent advances in neuropeptide signaling in Drosophila, from genes to physiology and behavior. Prog Neurobiol 2019; 179:101607. [PMID: 30905728 DOI: 10.1016/j.pneurobio.2019.02.003] [Citation(s) in RCA: 214] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/18/2019] [Accepted: 02/28/2019] [Indexed: 12/11/2022]
Abstract
This review focuses on neuropeptides and peptide hormones, the largest and most diverse class of neuroactive substances, known in Drosophila and other animals to play roles in almost all aspects of daily life, as w;1;ell as in developmental processes. We provide an update on novel neuropeptides and receptors identified in the last decade, and highlight progress in analysis of neuropeptide signaling in Drosophila. Especially exciting is the huge amount of work published on novel functions of neuropeptides and peptide hormones in Drosophila, largely due to the rapid developments of powerful genetic methods, imaging techniques and innovative assays. We critically discuss the roles of peptides in olfaction, taste, foraging, feeding, clock function/sleep, aggression, mating/reproduction, learning and other behaviors, as well as in regulation of development, growth, metabolic and water homeostasis, stress responses, fecundity, and lifespan. We furthermore provide novel information on neuropeptide distribution and organization of peptidergic systems, as well as the phylogenetic relations between Drosophila neuropeptides and those of other phyla, including mammals. As will be shown, neuropeptide signaling is phylogenetically ancient, and not only are the structures of the peptides, precursors and receptors conserved over evolution, but also many functions of neuropeptide signaling in physiology and behavior.
Collapse
Affiliation(s)
- Dick R Nässel
- Department of Zoology, Stockholm University, Stockholm, Sweden.
| | - Meet Zandawala
- Department of Zoology, Stockholm University, Stockholm, Sweden; Department of Neuroscience, Brown University, Providence, RI, USA.
| |
Collapse
|
41
|
Hökfelt T, Barde S, Xu ZQD, Kuteeva E, Rüegg J, Le Maitre E, Risling M, Kehr J, Ihnatko R, Theodorsson E, Palkovits M, Deakin W, Bagdy G, Juhasz G, Prud’homme HJ, Mechawar N, Diaz-Heijtz R, Ögren SO. Neuropeptide and Small Transmitter Coexistence: Fundamental Studies and Relevance to Mental Illness. Front Neural Circuits 2018; 12:106. [PMID: 30627087 PMCID: PMC6309708 DOI: 10.3389/fncir.2018.00106] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 11/05/2018] [Indexed: 12/31/2022] Open
Abstract
Neuropeptides are auxiliary messenger molecules that always co-exist in nerve cells with one or more small molecule (classic) neurotransmitters. Neuropeptides act both as transmitters and trophic factors, and play a role particularly when the nervous system is challenged, as by injury, pain or stress. Here neuropeptides and coexistence in mammals are reviewed, but with special focus on the 29/30 amino acid galanin and its three receptors GalR1, -R2 and -R3. In particular, galanin's role as a co-transmitter in both rodent and human noradrenergic locus coeruleus (LC) neurons is addressed. Extensive experimental animal data strongly suggest a role for the galanin system in depression-like behavior. The translational potential of these results was tested by studying the galanin system in postmortem human brains, first in normal brains, and then in a comparison of five regions of brains obtained from depressed people who committed suicide, and from matched controls. The distribution of galanin and the four galanin system transcripts in the normal human brain was determined, and selective and parallel changes in levels of transcripts and DNA methylation for galanin and its three receptors were assessed in depressed patients who committed suicide: upregulation of transcripts, e.g., for galanin and GalR3 in LC, paralleled by a decrease in DNA methylation, suggesting involvement of epigenetic mechanisms. It is hypothesized that, when exposed to severe stress, the noradrenergic LC neurons fire in bursts and release galanin from their soma/dendrites. Galanin then acts on somato-dendritic, inhibitory galanin autoreceptors, opening potassium channels and inhibiting firing. The purpose of these autoreceptors is to act as a 'brake' to prevent overexcitation, a brake that is also part of resilience to stress that protects against depression. Depression then arises when the inhibition is too strong and long lasting - a maladaption, allostatic load, leading to depletion of NA levels in the forebrain. It is suggested that disinhibition by a galanin antagonist may have antidepressant activity by restoring forebrain NA levels. A role of galanin in depression is also supported by a recent candidate gene study, showing that variants in genes for galanin and its three receptors confer increased risk of depression and anxiety in people who experienced childhood adversity or recent negative life events. In summary, galanin, a neuropeptide coexisting in LC neurons, may participate in the mechanism underlying resilience against a serious and common disorder, MDD. Existing and further results may lead to an increased understanding of how this illness develops, which in turn could provide a basis for its treatment.
Collapse
Affiliation(s)
- Tomas Hökfelt
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Swapnali Barde
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Zhi-Qing David Xu
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurobiology, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Laboratory of Brain Disorders (Ministry of Science and Technology), Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Eugenia Kuteeva
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Joelle Rüegg
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- The Center for Molecular Medicine, Stockholm, Sweden
- Swedish Toxicology Sciences Research Center, Swetox, Södertälje, Sweden
| | - Erwan Le Maitre
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Mårten Risling
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Jan Kehr
- Pronexus Analytical AB, Solna, Sweden
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Robert Ihnatko
- Department of Clinical Chemistry, Linköping University, Linköping, Sweden
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Elvar Theodorsson
- Department of Clinical Chemistry, Linköping University, Linköping, Sweden
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Miklos Palkovits
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - William Deakin
- Neuroscience and Psychiatry Unit, University of Manchester, Manchester, United Kingdom
| | - Gyorgy Bagdy
- Department of Pharmacodynamics, Semmelweis University, Budapest, Hungary
- MTA-SE Neuropsychopharmacology and Neurochemistry Research Group, Hungarian Academy of Sciences, Semmelweis University, Budapest, Hungary
- NAP 2-SE New Antidepressant Target Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary
| | - Gabriella Juhasz
- Neuroscience and Psychiatry Unit, University of Manchester, Manchester, United Kingdom
- Department of Pharmacodynamics, Semmelweis University, Budapest, Hungary
- SE-NAP2 Genetic Brain Imaging Migraine Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary
| | | | - Naguib Mechawar
- Douglas Hospital Research Centre, Verdun, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | | | - Sven Ove Ögren
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
42
|
Distribution of FMRFamide-related peptides and co-localization with glutamate in Cupiennius salei, an invertebrate model system. Cell Tissue Res 2018; 376:83-96. [PMID: 30406824 DOI: 10.1007/s00441-018-2949-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 10/09/2018] [Indexed: 01/18/2023]
Abstract
FMRFamide-related proteins have been described in both vertebrate and invertebrate nervous systems and have been suggested to play important roles in a variety of physiological processes. One proposed function is the modulation of signal transduction in mechanosensory neurons and their associated behavioral pathways in the Central American wandering spider Cupiennius salei; however, little is known about the distribution and abundance of FMRFamide-related proteins (FaRPs) within this invertebrate system. We employ immunohistochemistry, Hoechst nuclear stain and confocal microscopy of serial sections to detect, characterize and quantify FMRFamide-like immunoreactive neurons throughout all ganglia of the spider brain and along leg muscle. Within the different ganglia, between 3.4 and 12.6% of neurons showed immunolabeling. Among the immunoreactive cells, weakly and strongly labeled neurons could be distinguished. Between 71.4 and 81.7% of labeled neurons showed weak labeling, with 18.3 to 28.6% displaying strong labeling intensity. Among the weakly labeled neurons were characteristic motor neurons that have previously been shown to express ɣ-aminobutyric acid or glutamate. Ultrastructural investigations of neuromuscular junctions revealed mixed presynaptic vesicle populations including large electron-dense vesicles characteristic of neuropeptides. Double labeling for glutamate and FaRPs indicated that a subpopulation of neurons may co-express both neuroactive compounds. Our findings suggest that FaRPs are expressed throughout all ganglia and that different neurons have different expression levels. We conclude that FaRPs are likely utilized as neuromodulators in roughly 8% of neurons in the spider nervous system and that the main transmitter in a subpopulation of these neurons is likely glutamate.
Collapse
|
43
|
Systematic Analysis of Transmitter Coexpression Reveals Organizing Principles of Local Interneuron Heterogeneity. eNeuro 2018; 5:eN-NWR-0212-18. [PMID: 30294668 PMCID: PMC6171738 DOI: 10.1523/eneuro.0212-18.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 09/07/2018] [Accepted: 09/13/2018] [Indexed: 01/02/2023] Open
Abstract
Broad neuronal classes are surprisingly heterogeneous across many parameters, and subclasses often exhibit partially overlapping traits including transmitter coexpression. However, the extent to which transmitter coexpression occurs in predictable, consistent patterns is unknown. Here, we demonstrate that pairwise coexpression of GABA and multiple neuropeptide families by olfactory local interneurons (LNs) of the moth Manduca sexta is highly heterogeneous, with a single LN capable of expressing neuropeptides from at least four peptide families and few instances in which neuropeptides are consistently coexpressed. Using computational modeling, we demonstrate that observed coexpression patterns cannot be explained by independent probabilities of expression of each neuropeptide. Our analyses point to three organizing principles that, once taken into consideration, allow replication of overall coexpression structure: (1) peptidergic neurons are highly likely to coexpress GABA; (2) expression probability of allatotropin depends on myoinhibitory peptide expression; and (3) the all-or-none coexpression patterns of tachykinin neurons with several other neuropeptides. For other peptide pairs, the presence of one peptide was not predictive of the presence of the other, and coexpression probability could be replicated by independent probabilities. The stochastic nature of these coexpression patterns highlights the heterogeneity of transmitter content among LNs and argues against clear-cut definition of subpopulation types based on the presence of single neuropeptides. Furthermore, the receptors for all neuropeptides and GABA were expressed within each population of principal neuron type in the antennal lobe (AL). Thus, activation of any given LN results in a dynamic cocktail of modulators that have the potential to influence every level of olfactory processing within the AL.
Collapse
|
44
|
Kaltdorf KV, Theiss M, Markert SM, Zhen M, Dandekar T, Stigloher C, Kollmannsberger P. Automated classification of synaptic vesicles in electron tomograms of C. elegans using machine learning. PLoS One 2018; 13:e0205348. [PMID: 30296290 PMCID: PMC6175533 DOI: 10.1371/journal.pone.0205348] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 09/24/2018] [Indexed: 11/18/2022] Open
Abstract
Synaptic vesicles (SVs) are a key component of neuronal signaling and fulfil different roles depending on their composition. In electron micrograms of neurites, two types of vesicles can be distinguished by morphological criteria, the classical “clear core” vesicles (CCV) and the typically larger “dense core” vesicles (DCV), with differences in electron density due to their diverse cargos. Compared to CCVs, the precise function of DCVs is less defined. DCVs are known to store neuropeptides, which function as neuronal messengers and modulators [1]. In C. elegans, they play a role in locomotion, dauer formation, egg-laying, and mechano- and chemosensation [2]. Another type of DCVs, also referred to as granulated vesicles, are known to transport Bassoon, Piccolo and further constituents of the presynaptic density in the center of the active zone (AZ), and therefore are important for synaptogenesis [3]. To better understand the role of different types of SVs, we present here a new automated approach to classify vesicles. We combine machine learning with an extension of our previously developed vesicle segmentation workflow, the ImageJ macro 3D ART VeSElecT. With that we reliably distinguish CCVs and DCVs in electron tomograms of C. elegans NMJs using image-based features. Analysis of the underlying ground truth data shows an increased fraction of DCVs as well as a higher mean distance between DCVs and AZs in dauer larvae compared to young adult hermaphrodites. Our machine learning based tools are adaptable and can be applied to study properties of different synaptic vesicle pools in electron tomograms of diverse model organisms.
Collapse
Affiliation(s)
- Kristin Verena Kaltdorf
- Imaging Core Facility, Biocenter, University of Würzburg, Würzburg, Germany
- Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany
- Center for Computational and Theoretical Biology, University of Würzburg, Würzburg, Germany
| | - Maria Theiss
- Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany
- Center for Computational and Theoretical Biology, University of Würzburg, Würzburg, Germany
| | | | - Mei Zhen
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Thomas Dandekar
- Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany
- * E-mail: (PK); (CS); (TD)
| | - Christian Stigloher
- Imaging Core Facility, Biocenter, University of Würzburg, Würzburg, Germany
- * E-mail: (PK); (CS); (TD)
| | - Philip Kollmannsberger
- Center for Computational and Theoretical Biology, University of Würzburg, Würzburg, Germany
- * E-mail: (PK); (CS); (TD)
| |
Collapse
|
45
|
Clark T, Hapiak V, Oakes M, Mills H, Komuniecki R. Monoamines differentially modulate neuropeptide release from distinct sites within a single neuron pair. PLoS One 2018; 13:e0196954. [PMID: 29723289 PMCID: PMC5933757 DOI: 10.1371/journal.pone.0196954] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 04/23/2018] [Indexed: 12/14/2022] Open
Abstract
Monoamines and neuropeptides often modulate the same behavior, but monoaminergic-peptidergic crosstalk remains poorly understood. In Caenorhabditis elegans, the adrenergic-like ligands, tyramine (TA) and octopamine (OA) require distinct subsets of neuropeptides in the two ASI sensory neurons to inhibit nociception. TA selectively increases the release of ASI neuropeptides encoded by nlp-14 or nlp-18 from either synaptic/perisynaptic regions of ASI axons or the ASI soma, respectively, and OA selectively increases the release of ASI neuropeptides encoded by nlp-9 asymmetrically, from only the synaptic/perisynaptic region of the right ASI axon. The predicted amino acid preprosequences of genes encoding either TA- or OA-dependent neuropeptides differed markedly. However, these distinct preprosequences were not sufficient to confer monoamine-specificity and additional N-terminal peptide-encoding sequence was required. Collectively, our results demonstrate that TA and OA specifically and differentially modulate the release of distinct subsets of neuropeptides from different subcellular sites within the ASIs, highlighting the complexity of monoaminergic/peptidergic modulation, even in animals with a relatively simple nervous system.
Collapse
Affiliation(s)
- Tobias Clark
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, United States of America
| | - Vera Hapiak
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, United States of America
| | - Mitchell Oakes
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, United States of America
| | - Holly Mills
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, United States of America
| | - Richard Komuniecki
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, United States of America
- * E-mail:
| |
Collapse
|
46
|
Ross JA, Reyes BAS, Van Bockstaele EJ. Amyloid beta peptides, locus coeruleus-norepinephrine system and dense core vesicles. Brain Res 2018; 1702:46-53. [PMID: 29577889 DOI: 10.1016/j.brainres.2018.03.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 03/06/2018] [Accepted: 03/07/2018] [Indexed: 10/17/2022]
Abstract
The evolution of peptidergic signaling systems in the central nervous system serves a distinct and crucial role in brain processes and function. The diversity of physiological peptides and the complexity of their regulation and secretion from the dense core vesicles (DCV) throughout the brain is a topic greatly in need of investigation, though recent years have shed light on cellular and molecular mechanisms that are summarized in this review. Here, we focus on the convergence of peptidergic systems onto the Locus Coeruleus (LC), the sole provider of norepinephrine (NE) to the cortex and hippocampus, via large DCV. As the LC-NE system is one of the first regions of the brain to undergo degeneration in Alzheimer's Disease (AD), and markers of DCV have consistently been demonstrated to have biomarker potential for AD progression, here we summarize the current literature linking the LC-NE system with DCV dysregulation and Aβ peptides. We also include neuroanatomical data suggesting that the building blocks of senile plaques, Aβ monomers, may be localized to DCV of the LC and noradrenergic axon terminals of the prefrontal cortex. Finally, we explore the putative consequences of chronic stress on Aβ production and the role that DCV may play in LC degeneration. Clinical data of immunological markers of DCV in AD patients are discussed.
Collapse
Affiliation(s)
- Jennifer A Ross
- Department of Pharmacology and Physiology, College of Medicine, Drexel University, Philadelphia, PA 19102, United States.
| | - Beverly A S Reyes
- Department of Pharmacology and Physiology, College of Medicine, Drexel University, Philadelphia, PA 19102, United States
| | - Elisabeth J Van Bockstaele
- Department of Pharmacology and Physiology, College of Medicine, Drexel University, Philadelphia, PA 19102, United States
| |
Collapse
|
47
|
Bohlender JM, Nussberger J, Tevaearai H, Imboden H. Angiotensinergic Innervation of the Human Right Atrium: Implications for Cardiac Reflexes. Am J Hypertens 2018; 31:188-196. [PMID: 28985343 PMCID: PMC5861579 DOI: 10.1093/ajh/hpx163] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Accepted: 09/03/2017] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The right atrium is densely innervated and provides sensory input to important cardiocirculatory reflexes controlling cardiac output and blood pressure. Its angiotensin (Ang) II-expressing innervation may release Ang II as a neuropeptide cotransmitter to modulate reflexes but has not yet been characterized. METHODS Intraoperative surgical biopsies from human right atria (n = 7) were immunocytologically stained for Ang II, tyrosine hydroxylase (TH), and synaptophysin (SYN). Tissue angiotensins were extracted and quantified by radioimmunoassay. RESULTS Angiotensinergic fibers were frequent in epicardial nerves and around vessels with variable TH co-localization (none to >50%/bundle). Fibers were also widely distributed between cardiomyocytes and in the endocardium where they were typically nonvaricose, TH/SYN-negative and usually accompanied by varicose catecholaminergic fibers. In the endocardium, some showed large varicosities and were partially TH or SYN-positive. A few endocardial regions showed scattered nonvaricose Ang fibers ending directly between endothelial cells. Occasional clusters of thin varicose terminals co-localizing SYN or TH were located underneath, or protruded into, the endothelium. Endocardial density of Ang and TH-positive fibers was 30-300 vs. 200-450/mm2. Atrial Ang II, III, and I concentrations were 67, 16, and 5 fmol/g (median) while Ang IV and V were mostly undetectable. CONCLUSIONS The human right atrium harbors an abundant angiotensinergic innervation and a novel potential source of atrial Ang II. Most peripheral fibers were noncatecholaminergic afferents or preterminal vagal efferents and a minority was presumably sympathetic. Neuronal Ang II release from these fibers may modulate cardiac and circulatory reflexes independently from plasma and tissue Ang II sources.
Collapse
Affiliation(s)
- Jürgen M Bohlender
- Division of Clinical Pharmacology, Department of General Internal Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Jürg Nussberger
- Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Hendrik Tevaearai
- Department of Cardiovascular Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Hans Imboden
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| |
Collapse
|
48
|
Temporal-Spatial Profiling of Pedunculopontine Galanin-Cholinergic Neurons in the Lactacystin Rat Model of Parkinson's Disease. Neurotox Res 2017; 34:16-31. [PMID: 29218504 DOI: 10.1007/s12640-017-9846-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 11/22/2017] [Accepted: 11/22/2017] [Indexed: 12/17/2022]
Abstract
Parkinson's disease (PD) is conventionally seen as resulting from single-system neurodegeneration affecting nigrostriatal dopaminergic neurons. However, accumulating evidence indicates multi-system degeneration and neurotransmitter deficiencies, including cholinergic neurons which degenerate in a brainstem nucleus, the pedunculopontine nucleus (PPN), resulting in motor and cognitive impairments. The neuropeptide galanin can inhibit cholinergic transmission, while being upregulated in degenerating brain regions associated with cognitive decline. Here we determined the temporal-spatial profile of progressive expression of endogenous galanin within degenerating cholinergic neurons, across the rostro-caudal axis of the PPN, by utilizing the lactacystin-induced rat model of PD. First, we show progressive neuronal death affecting nigral dopaminergic and PPN cholinergic neurons, reflecting that seen in PD patients, to facilitate use of this model for assessing the therapeutic potential of bioactive peptides. Next, stereological analyses of the lesioned brain hemisphere found that the number of PPN cholinergic neurons expressing galanin increased by 11%, compared to sham-lesioned controls, and increasing by a further 5% as the neurodegenerative process evolved. Galanin upregulation within cholinergic PPN neurons was most prevalent closest to the intra-nigral lesion site, suggesting that galanin upregulation in such neurons adapt intrinsically to neurodegeneration, to possibly neuroprotect. This is the first report on the extent and pattern of galanin expression in cholinergic neurons across distinct PPN subregions in both the intact rat CNS and lactacystin-lesioned rats. The findings pave the way for future work to target galanin signaling in the PPN, to determine the extent to which upregulated galanin expression could offer a viable treatment strategy for ameliorating PD symptoms associated with cholinergic degeneration.
Collapse
|
49
|
FMRFamide-like peptides expand the behavioral repertoire of a densely connected nervous system. Proc Natl Acad Sci U S A 2017; 114:E10726-E10735. [PMID: 29167374 DOI: 10.1073/pnas.1710374114] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Animals, including humans, can adapt to environmental stress through phenotypic plasticity. The free-living nematode Caenorhabditis elegans can adapt to harsh environments by undergoing a whole-animal change, involving exiting reproductive development and entering the stress-resistant dauer larval stage. The dauer is a dispersal stage with dauer-specific behaviors for finding and stowing onto carrier animals, but how dauers acquire these behaviors, despite having a physically limited nervous system of 302 neurons, is poorly understood. We compared dauer and reproductive development using whole-animal RNA sequencing at fine time points and at sufficient depth to measure transcriptional changes within single cells. We detected 8,042 genes differentially expressed during dauer and reproductive development and observed striking up-regulation of neuropeptide genes during dauer entry. We knocked down neuropeptide processing using sbt-1 mutants and demonstrate that neuropeptide signaling promotes the decision to enter dauer rather than reproductive development. We also demonstrate that during dauer neuropeptides modulate the dauer-specific nictation behavior (carrier animal-hitchhiking) and are necessary for switching from repulsion to CO2 (a carrier animal cue) in nondauers to CO2 attraction in dauers. We tested individual neuropeptides using CRISPR knockouts and existing strains and demonstrate that the combined effects of flp-10 and flp-17 mimic the effects of sbt-1 on nictation and CO2 attraction. Through meta-analysis, we discovered similar up-regulation of neuropeptides in the dauer-like infective juveniles of diverse parasitic nematodes, suggesting the antiparasitic target potential of SBT-1. Our findings reveal that, under stress, increased neuropeptide signaling in C. elegans enhances their decision-making accuracy and expands their behavioral repertoire.
Collapse
|
50
|
Fabian-Fine R, Anderson CM, Roush MA, Johnson JAG, Liu H, French AS, Torkkeli PH. The distribution of cholinergic neurons and their co-localization with FMRFamide, in central and peripheral neurons of the spider Cupiennius salei. Cell Tissue Res 2017; 370:71-88. [PMID: 28687927 DOI: 10.1007/s00441-017-2652-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 05/23/2017] [Indexed: 12/21/2022]
Abstract
The spider Cupiennius salei is a well-established model for investigating information processing in arthropod sensory systems. Immunohistochemistry has shown that several neurotransmitters exist in the C. salei nervous system, including GABA, glutamate, histamine, octopamine and FMRFamide, while electrophysiology has found functional roles for some of these transmitters. There is also evidence that acetylcholine (ACh) is present in some C. salei neurons but information about the distribution of cholinergic neurons in spider nervous systems is limited. Here, we identify C. salei genes that encode enzymes essential for cholinergic transmission: choline ACh transferase (ChAT) and vesicular ACh transporter (VAChT). We used in-situ hybridization with an mRNA probe for C. salei ChAT gene to locate somata of cholinergic neurons in the central nervous system and immunohistochemistry with antisera against ChAT and VAChT to locate these proteins in cholinergic neurons. All three markers labeled similar, mostly small neurons, plus a few mid-sized neurons, in most ganglia. In the subesophageal ganglia, labeled neurons are putative efferent, motor or interneurons but the largest motor and interneurons were unlabeled. Groups of anti-ChAT labeled small neurons also connect the optic neuropils in the spider protocerebrum. Differences in individual cell labeling intensities were common, suggesting a range of ACh expression levels. Double-labeling found a subpopulation of anti-VAChT-labeled central and mechanosensory neurons that were also immunoreactive to antiserum against FMRFamide-like peptides. Our findings suggest that ACh is an important neurotransmitter in the C. salei central and peripheral nervous systems.
Collapse
Affiliation(s)
- Ruth Fabian-Fine
- Department of Biology, Saint Michael's College, One Winooski Park, Box 283, Colchester, VT, 05439, USA.
| | - Carly M Anderson
- Department of Biology, Saint Michael's College, One Winooski Park, Box 283, Colchester, VT, 05439, USA
| | - Molly A Roush
- Department of Biology, Saint Michael's College, One Winooski Park, Box 283, Colchester, VT, 05439, USA
| | - Jessica A G Johnson
- Department of Physiology and Biophysics, Dalhousie University, PO Box 15000, Halifax, NS, B3H 4R2, Canada
| | - Hongxia Liu
- Department of Physiology and Biophysics, Dalhousie University, PO Box 15000, Halifax, NS, B3H 4R2, Canada
| | - Andrew S French
- Department of Physiology and Biophysics, Dalhousie University, PO Box 15000, Halifax, NS, B3H 4R2, Canada
| | - Päivi H Torkkeli
- Department of Physiology and Biophysics, Dalhousie University, PO Box 15000, Halifax, NS, B3H 4R2, Canada
| |
Collapse
|