1
|
Ghanbari A. Beneficial Effects of Exercise in Neuropathic Pain: An Overview of the Mechanisms Involved. Pain Res Manag 2025; 2025:3432659. [PMID: 40040749 PMCID: PMC11879594 DOI: 10.1155/prm/3432659] [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: 11/02/2024] [Accepted: 02/08/2025] [Indexed: 03/06/2025]
Abstract
Neuropathic pain is a prevalent issue that often arises following injuries to the peripheral or central nervous system. Unfortunately, there is currently no definitive and flawless treatment available to alleviate this type of pain. However, exercise has emerged as a promising nonpharmacological and adjunctive approach, demonstrating a significant impact in reducing pain intensity. This is why physical therapy is considered a beneficial approach for diminishing pain and promoting functional recovery following nerve injuries. Regular physical activity exerts its hypoalgesic effects through a diverse array of mechanisms. These include inhibiting oxidative stress, suppressing inflammation, and modulating neurotransmitter levels, among others. It is possible that multiple activated mechanisms may coexist within an individual. However, the priming mechanism does not need to be the same across all subjects. Each person's response to physical activity and pain modulation may vary depending on their unique physiological and genetic factors. In this review, we aimed to provide a concise overview of the mechanisms underlying the beneficial effects of regular exercise on neuropathic pain. We have discussed several key mechanisms that contribute to the improvement of neuropathic pain through exercise. However, it is important to note that this is not an exhaustive analysis, and there may be other mechanisms at play. Our goal was to provide a brief yet informative exploration of the topic.
Collapse
Affiliation(s)
- Ali Ghanbari
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
- Department of Physiology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
2
|
Takasaki I, Nagashima R, Ueda T, Ashihara Y, Nakamachi T, Okada T, Toyooka N, Miyata A, Kurihara T. Spinal pituitary adenylate cyclase-activating polypeptide and PAC1 receptor signaling system is involved in the oxaliplatin-induced acute cold allodynia in mice. THE JOURNAL OF PAIN 2025; 27:104751. [PMID: 39615811 DOI: 10.1016/j.jpain.2024.104751] [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: 06/28/2024] [Revised: 11/04/2024] [Accepted: 11/25/2024] [Indexed: 02/10/2025]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a type of peripheral neuropathy that develops in patients treated with certain anticancer drugs. Oxaliplatin (OXA) causes CIPN in approximately 80-90 % of patients; thus, it is necessary to elucidate its underlying mechanism and develop effective treatments and prevention methods. The purpose of this study was to determine whether the pituitary adenylate cyclase-activating polypeptide (PACAP)/PAC1 receptor system in the spinal dorsal horn is involved in OXA-induced acute cold allodynia and examine the effect of a PAC1 receptor antagonist. Administration of OXA induced acute cold allodynia in wild-type mice, but not in PACAP-/- mice. In the dorsal root ganglia, OXA upregulated PACAP expression, particularly in small-sized neurons. OXA-induced cold allodynia was ameliorated by intrathecal (i.t.) injection of PACAP6-38 (peptide antagonist for PACAP receptor) and PA-8 (small-molecule antagonist specific for PAC1 receptor). I.t. PACAP, but not vasoactive intestinal polypeptide, resulted in cold allodynia, which was blocked by PA-8. OXA induced the activation of spinal astrocytes in a PAC1 receptor-dependent manner. The results suggest that spinal PACAP/PAC1 receptor systems are involved in OXA-induced acute cold allodynia through astrocyte activation. Furthermore, we demonstrated that the systemic administration of PA-8 resulted in therapeutic and preventative effects on OXA-induced acute cold allodynia. Because PA-8 did not affect the anticancer effects of OXA, we propose PAC1 receptor inhibition as a new strategy for the treatment and prevention of CIPN. PERSPECTIVE: Cold allodynia is a hallmark of OXA-induced peripheral neuropathy. This study demonstrated the involvement of spinal PACAP/PAC1 receptors in OXA-induced acute cold allodynia. We propose PAC1 receptor inhibition as a new strategy for the treatment and prevention of OXA-induced acute cold allodynia.
Collapse
Affiliation(s)
- Ichiro Takasaki
- Laboratory of Pharmacology, Faculty of Engineering, University of Toyama, Toyama 930-8555, Japan; Graduate School of Pharma-Medical Sciences, University of Toyama, Toyama 930-8555, Japan; Graduate School of Innovative Life Science, University of Toyama, Toyama 930-8555, Japan.
| | - Ryota Nagashima
- Laboratory of Pharmacology, Faculty of Engineering, University of Toyama, Toyama 930-8555, Japan
| | - Takahiro Ueda
- Laboratory of Pharmacology, Faculty of Engineering, University of Toyama, Toyama 930-8555, Japan
| | - Yuya Ashihara
- Laboratory of Pharmacology, Faculty of Engineering, University of Toyama, Toyama 930-8555, Japan; Graduate School of Pharma-Medical Sciences, University of Toyama, Toyama 930-8555, Japan
| | - Tomoya Nakamachi
- Laboratory of Regulatory Biology, Faculty of Science, University of Toyama, Toyama 930-8555, Japan
| | - Takuya Okada
- Graduate School of Pharma-Medical Sciences, University of Toyama, Toyama 930-8555, Japan; Graduate School of Innovative Life Science, University of Toyama, Toyama 930-8555, Japan; Laboratory of Bio-functional Molecular Engineering, Faculty of Engineering, University of Toyama, Toyama 930-8555, Japan
| | - Naoki Toyooka
- Graduate School of Pharma-Medical Sciences, University of Toyama, Toyama 930-8555, Japan; Graduate School of Innovative Life Science, University of Toyama, Toyama 930-8555, Japan; Laboratory of Bio-functional Molecular Engineering, Faculty of Engineering, University of Toyama, Toyama 930-8555, Japan
| | - Atsuro Miyata
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Takashi Kurihara
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| |
Collapse
|
3
|
Crosson T, Bhat S, Wang JC, Salaun C, Fontaine E, Roversi K, Herzog H, Rafei M, Blunck R, Talbot S. Cytokines reprogram airway sensory neurons in asthma. Cell Rep 2024; 43:115045. [PMID: 39661516 DOI: 10.1016/j.celrep.2024.115045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/01/2024] [Accepted: 11/18/2024] [Indexed: 12/13/2024] Open
Abstract
Nociceptor neurons play a crucial role in maintaining the body's homeostasis by detecting and responding to potential environmental dangers. However, this function can be detrimental during allergic reactions, as vagal nociceptors contribute to immune cell infiltration, bronchial hypersensitivity, and mucus imbalance in addition to causing pain and coughing. Despite this, the specific mechanisms by which nociceptors acquire pro-inflammatory characteristics during allergic reactions are not yet fully understood. In this study, we investigate the changes in the molecular profile of airway nociceptor neurons during allergic airway inflammation and identify the signals driving such reprogramming. Using retrograde tracing and lineage reporting, we identify a specific class of inflammatory vagal nociceptor neurons that exclusively innervate the airways. In the ovalbumin mouse model of allergic airway inflammation, these neurons undergo significant reprogramming characterized by the upregulation of the neuropeptide Y (NPY) receptor Npy1r. A screening of cytokines and neurotrophins reveals that interleukin 1β (IL-1β), IL-13, and brain-derived neurotrophic factor (BDNF) drive part of this reprogramming. IL-13 triggers Npy1r overexpression in nociceptors via the JAK/STAT6 pathway. In parallel, NPY is released into the bronchoalveolar fluid of asthmatic mice, which limits the excitability of nociceptor neurons. Single-cell RNA sequencing of lung immune cells reveals that a cell-specific knockout of NPY1R in nociceptor neurons in asthmatic mice altered T cell infiltration. Opposite findings are observed in asthmatic mice in which nociceptor neurons are chemically ablated. In summary, allergic airway inflammation reprograms airway nociceptor neurons to acquire a pro-inflammatory phenotype, while a compensatory mechanism involving NPY1R limits the activity of nociceptor neurons.
Collapse
Affiliation(s)
- Theo Crosson
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | - Shreyas Bhat
- Centre Interdisciplinaire sur le Cerveau et l'Apprentissage, Université de Montréal, Montreal, QC, Canada; Département de Physique, Université de Montréal, Montreal, QC, Canada
| | - Jo-Chiao Wang
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | - Clara Salaun
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | - Eleanne Fontaine
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | - Katiane Roversi
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | - Herbert Herzog
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Moutih Rafei
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | - Rikard Blunck
- Centre Interdisciplinaire sur le Cerveau et l'Apprentissage, Université de Montréal, Montreal, QC, Canada; Département de Physique, Université de Montréal, Montreal, QC, Canada
| | - Sebastien Talbot
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Sweden; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
4
|
Hayward R, Moore S, Artun D, Madhavan A, Harte E, Torres-Pérez JV, Nagy I. Transcriptional reprogramming post-peripheral nerve injury: A systematic review. Neurobiol Dis 2024; 200:106624. [PMID: 39097036 DOI: 10.1016/j.nbd.2024.106624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/27/2024] [Accepted: 07/30/2024] [Indexed: 08/05/2024] Open
Abstract
Neuropathic pain is characterised by periodic or continuous hyperalgesia, numbness, or allodynia, and results from insults to the somatosensory nervous system. Peripheral nerve injury induces transcriptional reprogramming in peripheral sensory neurons, contributing to increased spinal nociceptive input and the development of neuropathic pain. Effective treatment for neuropathic pain remains an unmet medical need as current therapeutics offer limited effectiveness and have undesirable effects. Understanding transcriptional changes in peripheral nerve injury-induced neuropathy might offer a path for novel analgesics. Our literature search identified 65 papers exploring transcriptomic changes post-peripheral nerve injury, many of which were conducted in animal models. We scrutinize their transcriptional changes data and conduct gene ontology enrichment analysis to reveal their common functional profile. Focusing on genes involved in 'sensory perception of pain' (GO:0019233), we identified transcriptional changes for different ion channels, receptors, and neurotransmitters, shedding light on its role in nociception. Examining peripheral sensory neurons subtype-specific transcriptional reprograming and regeneration-associated genes, we delved into downstream regulation of hypersensitivity. Identifying the temporal program of transcription regulatory mechanisms might help develop better therapeutics to target them effectively and selectively, thus preventing the development of neuropathic pain without affecting other physiological functions.
Collapse
Affiliation(s)
- R Hayward
- Nociception Group, Department of Surgery and Cancer, Division of Anaesthetics, Pain Medicine and Intensive Care, Chelsea and Westminster Hospital Campus, Imperial College London, 369 Fulham Road, London SW10 9FJ, UK
| | - S Moore
- Nociception Group, Department of Surgery and Cancer, Division of Anaesthetics, Pain Medicine and Intensive Care, Chelsea and Westminster Hospital Campus, Imperial College London, 369 Fulham Road, London SW10 9FJ, UK
| | - D Artun
- Nociception Group, Department of Surgery and Cancer, Division of Anaesthetics, Pain Medicine and Intensive Care, Chelsea and Westminster Hospital Campus, Imperial College London, 369 Fulham Road, London SW10 9FJ, UK
| | - A Madhavan
- Nociception Group, Department of Surgery and Cancer, Division of Anaesthetics, Pain Medicine and Intensive Care, Chelsea and Westminster Hospital Campus, Imperial College London, 369 Fulham Road, London SW10 9FJ, UK
| | - E Harte
- Nociception Group, Department of Surgery and Cancer, Division of Anaesthetics, Pain Medicine and Intensive Care, Chelsea and Westminster Hospital Campus, Imperial College London, 369 Fulham Road, London SW10 9FJ, UK
| | - J V Torres-Pérez
- Departament de Biologia Cel·lular, Biologia Funcional i Antropologia Física, Facultat de Ciències Biològiques, Universitat de València, C/Dr. Moliner 50, 46100 Burjassot, Spain.
| | - I Nagy
- Nociception Group, Department of Surgery and Cancer, Division of Anaesthetics, Pain Medicine and Intensive Care, Chelsea and Westminster Hospital Campus, Imperial College London, 369 Fulham Road, London SW10 9FJ, UK.
| |
Collapse
|
5
|
Longwill O. Exploring the Role of Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) and Kynurenine Pathway Dysregulation in Migraine Pathophysiology Among Women With Polycystic Ovary Syndrome (PCOS). Cureus 2024; 16:e71199. [PMID: 39525239 PMCID: PMC11549845 DOI: 10.7759/cureus.71199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
A narrative review was undertaken to explore the current understanding of the relationship between polycystic ovary syndrome and migraine headaches, with a focus on the potential roles of pituitary adenylate cyclase-activating polypeptide and the kynurenine pathway in the shared pathophysiology of these conditions. Emerging evidence suggests that pituitary adenylate cyclase-activating polypeptide may be a key player in the development of migraine headaches, with potential implications for the higher incidence of migraine observed in women with polycystic ovary syndrome. Additionally, dysregulation of the kynurenine pathway and altered levels of kynurenine metabolites have been linked to both migraine and polycystic ovary syndrome, indicating a complex interplay between hormonal, metabolic, and neurological factors in the comorbid presentation of these disorders. Further research is needed to elucidate the specific mechanisms underlying these associations and to develop targeted therapeutic approaches for managing migraine in the context of polycystic ovary syndrome.
Collapse
|
6
|
Crosson T, Bhat S, Wang JC, Salaun C, Fontaine E, Roversi K, Herzog H, Rafei M, Blunck R, Talbot S. Cytokines reprogram airway sensory neurons in asthma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.01.26.525731. [PMID: 39345572 PMCID: PMC11429693 DOI: 10.1101/2023.01.26.525731] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Nociceptor neurons play a crucial role in maintaining the body's homeostasis by detecting and responding to potential dangers in the environment. However, this function can be detrimental during allergic reactions, since vagal nociceptors can contribute to immune cell infiltration, bronchial hypersensitivity, and mucus imbalance, in addition to causing pain and coughing. Despite this, the specific mechanisms by which nociceptors acquire pro-inflammatory characteristics during allergic reactions are not yet fully understood. In this study, we aimed to investigate the molecular profile of airway nociceptor neurons during allergic airway inflammation and identify the signals driving such reprogramming. Using retrograde tracing and lineage reporting, we identified a unique class of inflammatory vagal nociceptor neurons that exclusively innervate the airways. In the ovalbumin mouse model of airway inflammation, these neurons undergo significant reprogramming characterized by the upregulation of the NPY receptor Npy1r. A screening of cytokines and neurotrophins revealed that IL-1β, IL-13 and BDNF drive part of this reprogramming. IL-13 triggered Npy1r overexpression in nociceptors via the JAK/STAT6 pathway. In parallel, sympathetic neurons and macrophages release NPY in the bronchoalveolar fluid of asthmatic mice, which limits the excitability of nociceptor neurons. Single-cell RNA sequencing of lung immune cells has revealed that a cell-specific knockout of Npy1r in nociceptor neurons in asthmatic mice leads to an increase in airway inflammation mediated by T cells. Opposite findings were observed in asthmatic mice in which nociceptor neurons were chemically ablated. In summary, allergic airway inflammation reprograms airway nociceptor neurons to acquire a pro-inflammatory phenotype, while a compensatory mechanism involving NPY1R limits nociceptor neurons' activity.
Collapse
Affiliation(s)
- Théo Crosson
- Département de Pharmacologie et Physiologie, Université de Montréal, Canada
| | - Shreyas Bhat
- Centre Interdisciplinaire sur le Cerveau et l’Apprentissage, Université de Montréal, Canada
- Département de Physique, Université de Montréal, Canada
| | - Jo-Chiao Wang
- Département de Pharmacologie et Physiologie, Université de Montréal, Canada
| | - Clara Salaun
- Département de Pharmacologie et Physiologie, Université de Montréal, Canada
| | - Eleanne Fontaine
- Département de Pharmacologie et Physiologie, Université de Montréal, Canada
| | - Katiane Roversi
- Département de Pharmacologie et Physiologie, Université de Montréal, Canada
| | | | - Moutih Rafei
- Département de Pharmacologie et Physiologie, Université de Montréal, Canada
| | - Rikard Blunck
- Centre Interdisciplinaire sur le Cerveau et l’Apprentissage, Université de Montréal, Canada
- Département de Physique, Université de Montréal, Canada
| | - Sebastien Talbot
- Department of Physiology and Pharmacology, Karolinska Institutet. Sweden
- Department of Biomedical and Molecular Sciences, Queen’s University. Canada
| |
Collapse
|
7
|
Yamaguchi M, Noda-Asano S, Inoue R, Himeno T, Motegi M, Hayami T, Nakai-Shimoda H, Kono A, Sasajima S, Miura-Yura E, Morishita Y, Kondo M, Tsunekawa S, Kato Y, Kato K, Naruse K, Nakamura J, Kamiya H. Dipeptidyl Peptidase (DPP)-4 Inhibitors and Pituitary Adenylate Cyclase-Activating Polypeptide, a DPP-4 Substrate, Extend Neurite Outgrowth of Mouse Dorsal Root Ganglia Neurons: A Promising Approach in Diabetic Polyneuropathy Treatment. Int J Mol Sci 2024; 25:8881. [PMID: 39201570 PMCID: PMC11354620 DOI: 10.3390/ijms25168881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 08/08/2024] [Accepted: 08/14/2024] [Indexed: 09/02/2024] Open
Abstract
Individuals suffering from diabetic polyneuropathy (DPN) experience debilitating symptoms such as pain, paranesthesia, and sensory disturbances, prompting a quest for effective treatments. Dipeptidyl-peptidase (DPP)-4 inhibitors, recognized for their potential in ameliorating DPN, have sparked interest, yet the precise mechanism underlying their neurotrophic impact on the peripheral nerve system (PNS) remains elusive. Our study delves into the neurotrophic effects of DPP-4 inhibitors, including Diprotin A, linagliptin, and sitagliptin, alongside pituitary adenylate cyclase-activating polypeptide (PACAP), Neuropeptide Y (NPY), and Stromal cell-derived factor (SDF)-1a-known DPP-4 substrates with neurotrophic properties. Utilizing primary culture dorsal root ganglia (DRG) neurons, we meticulously evaluated neurite outgrowth in response to these agents. Remarkably, all DPP-4 inhibitors and PACAP demonstrated a significant elongation of neurite length in DRG neurons (PACAP 0.1 μM: 2221 ± 466 μm, control: 1379 ± 420, p < 0.0001), underscoring their potential in nerve regeneration. Conversely, NPY and SDF-1a failed to induce neurite elongation, accentuating the unique neurotrophic properties of DPP-4 inhibition and PACAP. Our findings suggest that the upregulation of PACAP, facilitated by DPP-4 inhibition, plays a pivotal role in promoting neurite elongation within the PNS, presenting a promising avenue for the development of novel DPN therapies with enhanced neurodegenerative capabilities.
Collapse
Affiliation(s)
- Masahiro Yamaguchi
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute 480-1185, Japan
| | - Saeko Noda-Asano
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute 480-1185, Japan
| | - Rieko Inoue
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute 480-1185, Japan
| | - Tatsuhito Himeno
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute 480-1185, Japan
- Department of Innovative Diabetes Therapy, Aichi Medical University School of Medicine, Nagakute 480-1185, Japan
| | - Mikio Motegi
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute 480-1185, Japan
| | - Tomohide Hayami
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute 480-1185, Japan
| | - Hiromi Nakai-Shimoda
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute 480-1185, Japan
| | - Ayumi Kono
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute 480-1185, Japan
| | - Sachiko Sasajima
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute 480-1185, Japan
| | - Emiri Miura-Yura
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute 480-1185, Japan
| | - Yoshiaki Morishita
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute 480-1185, Japan
| | - Masaki Kondo
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute 480-1185, Japan
| | - Shin Tsunekawa
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute 480-1185, Japan
| | - Yoshiro Kato
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute 480-1185, Japan
| | - Koichi Kato
- Department of Medicine, Aichi Gakuin University School of Pharmacy, Nagoya 464-8650, Japan
| | - Keiko Naruse
- Department of Internal Medicine, Aichi Gakuin University School of Dentistry, Nagoya 464-0821, Japan
| | - Jiro Nakamura
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute 480-1185, Japan
- Department of Innovative Diabetes Therapy, Aichi Medical University School of Medicine, Nagakute 480-1185, Japan
| | - Hideki Kamiya
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute 480-1185, Japan
| |
Collapse
|
8
|
Tasma Z, Rees TA, Guo S, Tan S, O'Carroll SJ, Faull RLM, Curtis MA, Christensen SL, Hay DL, Walker CS. Pharmacology of PACAP and VIP receptors in the spinal cord highlights the importance of the PAC 1 receptor. Br J Pharmacol 2024; 181:2655-2675. [PMID: 38616050 DOI: 10.1111/bph.16376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/18/2023] [Accepted: 01/20/2024] [Indexed: 04/16/2024] Open
Abstract
BACKGROUND AND PURPOSE The spinal cord is a key structure involved in the transmission and modulation of pain. Pituitary adenylate cyclase-activating peptide (PACAP) and vasoactive intestinal peptide (VIP), are expressed in the spinal cord. These peptides activate G protein-coupled receptors (PAC1, VPAC1 and VPAC2) that could provide targets for the development of novel pain treatments. However, it is not clear which of these receptors are expressed within the spinal cord and how these receptors signal. EXPERIMENTAL APPROACH Dissociated rat spinal cord cultures were used to examine agonist and antagonist receptor pharmacology. Signalling profiles were determined for five signalling pathways. The expression of different PACAP and VIP receptors was then investigated in mouse, rat and human spinal cords using immunoblotting and immunofluorescence. KEY RESULTS PACAP, but not VIP, potently stimulated cAMP, IP1 accumulation and ERK and cAMP response element-binding protein (CREB) but not Akt phosphorylation in spinal cord cultures. Signalling was antagonised by M65 and PACAP6-38. PACAP-27 was more effectively antagonised than either PACAP-38 or VIP. The patterns of PAC1 and VPAC2 receptor-like immunoreactivity appeared to be distinct in the spinal cord. CONCLUSIONS AND IMPLICATIONS The pharmacological profile in the spinal cord suggested that a PAC1 receptor is the major functional receptor subtype present and thus likely mediates the nociceptive effects of the PACAP family of peptides in the spinal cord. However, the potential expression of both PAC1 and VPAC2 receptors in the spinal cord highlights that these receptors may play differential roles and are both possible therapeutic targets.
Collapse
MESH Headings
- Animals
- Spinal Cord/metabolism
- Spinal Cord/drug effects
- Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide, Type I/metabolism
- Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide, Type I/agonists
- Humans
- Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology
- Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism
- Vasoactive Intestinal Peptide/metabolism
- Vasoactive Intestinal Peptide/pharmacology
- Mice
- Rats
- Signal Transduction/drug effects
- Receptors, Vasoactive Intestinal Peptide/metabolism
- Receptors, Vasoactive Intestinal Peptide/antagonists & inhibitors
- Cells, Cultured
- Rats, Sprague-Dawley
- Male
- Mice, Inbred C57BL
- Cyclic AMP/metabolism
- Receptors, Vasoactive Intestinal Peptide, Type II/metabolism
- Receptors, Vasoactive Intestinal Peptide, Type II/agonists
Collapse
Affiliation(s)
- Zoe Tasma
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | - Tayla A Rees
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Song Guo
- Department of Neurology, Danish Headache Center, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Odontology, Panum Institute, Faculty of Health, University of Copenhagen, Copenhagen, Denmark
| | - Sheryl Tan
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, The University of Auckland, Auckland, New Zealand
| | - Simon J O'Carroll
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, The University of Auckland, Auckland, New Zealand
| | - Richard L M Faull
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, The University of Auckland, Auckland, New Zealand
| | - Maurice A Curtis
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, The University of Auckland, Auckland, New Zealand
| | - Sarah L Christensen
- Department of Neurology, Danish Headache Center, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Debbie L Hay
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
- Department of Pharmacology and Toxicology, The University of Otago, Dunedin, New Zealand
| | - Christopher S Walker
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
9
|
Kato N, Kambe T, Chiba T, Taguchi K, Abe K. Analgesic effect of a cholinergic agonist (carbachol) in a sural nerve ligation-induced hypersensitivity mouse model. Neurol Res 2024; 46:505-515. [PMID: 38569563 DOI: 10.1080/01616412.2024.2337512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 03/26/2024] [Indexed: 04/05/2024]
Abstract
OBJECTIVES Neuropathic pain is characterized by long-lasting, intractable pain. Sciatic nerve ligation is often used as an animal model of neuropathic pain, and the spared nerve injury (SNI) model, in which the common peroneal nerve (CPN) and tibial nerve (TN) are ligated, is widely used. In the present study, we evaluated the analgesic effect of a cholinergic agonist, carbachol, on a neuropathic pain model prepared by sural nerve (SN) ligation in mice. METHODS The SN was tightly ligated as a branch of the sciatic nerve. Mechanical and thermal allodynia, and hyperalgesia were assessed using von Frey filaments and heat from a hot plate. The analgesic effects of intracerebroventricularly-administered morphine and carbachol were compared. RESULTS SN ligation resulted in a significant decrease in pain threshold for mechanical stimulation 1 day after ligation. In response to thermal stimulation, allodynia was observed at 50°C and hyperalgesia at 53 and 56°C 3 days after ligation. Content of thiobarbituric acid reactive substances (TBARS) in the spinal cord increased significantly at 6 and 12 h after ligation. Acetylcholine content of the spinal cord also increased at 5 and 7 days after ligation. Intracerebroventricular administration of carbachol at 7 days after ligation produced a marked analgesic effect against mechanical and thermal stimuli, which was stronger and longer-lasting than morphine at all experimental time points. CONCLUSION These findings suggest that cholinergic nerves are involved in allodynia and hyperalgesia of the SN ligation neuropathic pain model.
Collapse
Affiliation(s)
- Nodoka Kato
- Department of Pharmacology, School of Pharmaceutical Sciences, Ohu University, Koriyama, Fukushima, Japan
| | - Toshie Kambe
- Department of Medicinal Pharmacology, Showa Pharmaceutical University, Machida, Tokyo, Japan
| | - Terumasa Chiba
- Faculty of Pharmaceutical Sciences, Nihon Pharmaceutical University, Saitama, Japan
| | - Kyoji Taguchi
- Department of Pharmacology, ShowaPharmaceutical University, Machida, Tokyo, Japan
| | - Kenji Abe
- Department of Pharmacology, School of Pharmaceutical Sciences, Ohu University, Koriyama, Fukushima, Japan
- Faculty of Pharmaceutical Sciences, Nihon Pharmaceutical University, Saitama, Japan
| |
Collapse
|
10
|
Dong LG, An MQ, Gu HY, Zhang LG, Zhang JB, Li CJ, Mao CJ, Wang F, Liu CF. PACAP/PAC1-R activation contributes to hyperalgesia in 6-OHDA-induced Parkinson's disease model rats via promoting excitatory synaptic transmission of spinal dorsal horn neurons. Acta Pharmacol Sin 2023; 44:2418-2431. [PMID: 37563446 PMCID: PMC10692161 DOI: 10.1038/s41401-023-01141-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 07/12/2023] [Indexed: 08/12/2023]
Abstract
Pain is a common annoying non-motor symptom in Parkinson's disease (PD) that causes distress to patients. Treatment for PD pain remains a big challenge, as its underlying mechanisms are elusive. Pituitary adenylate cyclase-activating polypeptide (PACAP) and its receptor PAC1-R play important roles in regulating a variety of pathophysiological processes. In this study, we investigated whether PACAP/PAC1-R signaling was involved in the mechanisms of PD pain. 6-hydroxydopamine (6-OHDA)-induced PD model was established in rats. Behavioral tests, electrophysiological and Western blotting analysis were conducted 3 weeks later. We found that 6-OHDA rats had significantly lower mechanical paw withdrawal 50% threshold in von Frey filament test and shorter tail flick latency, while mRNA levels of Pacap and Adcyap1r1 (gene encoding PAC1-R) in the spinal dorsal horn were significantly upregulated. Whole-cell recordings from coronal spinal cord slices at L4-L6 revealed that the frequency of spontaneous excitatory postsynaptic currents (sEPSCs) in dorsal horn neurons was significantly increased, which was reversed by application of a PAC1-R antagonist PACAP 6-38 (250 nM). Furthermore, we demonstrated that intrathecal microinjection of PACAP 6-38 (0.125, 0.5, 2 μg) dose-dependently ameliorated the mechanical and thermal hyperalgesia in 6-OHDA rats. Inhibition of PACAP/PAC1-R signaling significantly suppressed the activation of Ca2+/calmodulin-dependent protein kinase II and extracellular signal-regulated kinase (ERK) in spinal dorsal horn of 6-OHDA rats. Microinjection of pAAV-Adcyap1r1 into L4-L6 spinal dorsal horn alleviated hyperalgesia in 6-OHDA rats. Intrathecal microinjection of ERK antagonist PD98059 (10 μg) significantly alleviated hyperalgesia in 6-OHDA rats associated with the inhibition of sEPSCs in dorsal horn neurons. In addition, we found that serum PACAP-38 concentration was significantly increased in PD patients with pain, and positively correlated with numerical rating scale score. In conclusion, activation of PACAP/PAC1-R induces the development of PD pain and targeting PACAP/PAC1-R is an alternative strategy for treating PD pain.
Collapse
Affiliation(s)
- Li-Guo Dong
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Meng-Qi An
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Han-Ying Gu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Li-Ge Zhang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Jin-Bao Zhang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Cheng-Jie Li
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Cheng-Jie Mao
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Fen Wang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Chun-Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China.
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China.
- Department of Neurology, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, 830063, China.
| |
Collapse
|
11
|
Wang H, Zhang C, Hong SH, Maye P, Rowe D, Shin DG. CGCom: a framework for inferring Cell-cell Communication based on Graph Neural Network. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.10.566642. [PMID: 38014057 PMCID: PMC10680670 DOI: 10.1101/2023.11.10.566642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Cell-cell communication is crucial in maintaining cellular homeostasis, cell survival and various regulatory relationships among interacting cells. Thanks to recent advances of spatial transcriptomics technologies, we can now explore if and how cells' proximal information available from spatial transcriptomics datasets can be used to infer cell-cell communication. Here we present a cell-cell communication inference framework, called CGCom, which uses a graph neural network (GNN) to learn communication patterns among interacting cells by combining single-cell spatial transcriptomic datasets with publicly available ligand-receptor information and the molecular regulatory information down-stream of the ligand-receptor signaling. To evaluate the performance of CGCom, we applied it to mouse embryo seqFISH datasets. Our results demonstrate that CGCom can not only accurately infer cell communication between individual cell pairs but also generalize its learning to predict communication between different cell types. We compared the performance of CGCom with two existing methods, CellChat and CellPhoneDB, and our comparative study revealed both common and unique communication patterns from the three approaches. Commonly found communication patterns include three sets of ligand-receptor communication relationships, one between surface ectoderm cells and spinal cord cells, one between gut tube cells and endothelium, and one between neural crest and endothelium, all of which have already been reported in the literature thus offering credibility of all three methods. However, we hypothesize that CGCom is superior in reducing false positives thanks to its use of cell proximal information and its learning between specific cell pairs rather than between cell types. CGCom is a GNN-based solution that can take advantage of spatially resolved single-cell transcriptomic data in predicting cell-cell communication with a higher accuracy.
Collapse
|
12
|
Seiglie MP, Lepeak L, Miracle S, Cottone P, Sabino V. Stimulation of lateral parabrachial (LPB) to central amygdala (CeA) pituitary adenylate cyclase-activating polypeptide (PACAP) neurons induces anxiety-like behavior and mechanical allodynia. Pharmacol Biochem Behav 2023; 230:173605. [PMID: 37499765 DOI: 10.1016/j.pbb.2023.173605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
BACKGROUND Anxiety disorders are the most prevalent psychiatric disorders, and they are highly comorbid with chronic pain conditions. The central nucleus of the amygdala (CeA) is known not only for its role in the regulation of anxiety but also as an important site for the negative affective dimension of pain. Pituitary adenylate cyclase activating polypeptide (PACAP), a neuropeptide whose terminals are abundant in the CeA, is strongly implicated in the stress response as well as in pain processing. Here, using Cre-dependent viral vectors, we explored in greater detail the role of the PACAP projection to the CeA that originates in the lateral parabrachial nucleus (LPB). METHODS We first performed a circuit mapping experiment by injecting an anterograde Cre-dependent virus expressing a fluorescent reporter in the LPB of PACAP-Cre mice and observing their projections. Then, we used a chemogenetic approach (a Cre-dependent Designer Receptors Activated by Designer Drugs, DREADDs) to assess the effects of the direct stimulation of the PACAP LPB to CeA projection on general locomotor activity, anxiety-like behavior (using a defensive withdrawal test), and mechanical pain sensitivity (using the von Frey test). RESULTS We found that the CeA, together with other areas, is one of the major downstream projection targets of PACAP neurons originating in the lateral parabrachial nucleus (LPB). In the DREADD experiment, we then found that the selective activation of this neuronal pathway is sufficient to increase both anxiety-like behavior and mechanical pain sensitivity in mice, without affecting general locomotor activity. CONCLUSION In conclusion, our data suggest that the dysregulation of this circuit may contribute to a variety of anxiety disorders and chronic pain states, and that PACAP may represent an important therapeutic target for the treatment of these conditions.
Collapse
Affiliation(s)
- Mariel P Seiglie
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Lauren Lepeak
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Sophia Miracle
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Pietro Cottone
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Valentina Sabino
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
13
|
Zheng JH, Yuan N, Zhang P, Liu DF, Lin W, Miao J. Acupuncture combined with moxibustion mitigates spinal cord injury-induced motor dysfunction in mice by NLRP3-IL-18 signaling pathway inhibition. J Orthop Surg Res 2023; 18:419. [PMID: 37296436 DOI: 10.1186/s13018-023-03902-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023] Open
Abstract
BACKGROUND Spinal cord injury (SCI), which reportedly induces severe motor dysfunction, imposes a significant social and financial burden on affected individuals, families, communities, and nations. Acupuncture combined with moxibustion (AM) therapy has been widely used for motor dysfunction treatment, but the underlying mechanisms remain unknown. In this work, we aimed to determine whether AM therapy could alleviate motor impairment post-SCI and, if so, the potential mechanism. METHODS A SCI model was established in mice through impact methods. AM treatment was performed in SCI model mice at Dazhui (GV14) and Jiaji points (T7-T12), Mingmen (GV4), Zusanli (ST36), and Ciliao (BL32) on both sides for 30 min once per day for 28 days. The Basso-Beattie-Bresnahan score was used to assess motor function in mice. A series of experiments including astrocytes activation detected by immunofluorescence, the roles of NOD-like receptor pyrin domain-containing-3 (NLRP3)-IL-18 signaling pathway with the application of astrocyte-specific NLRP3 knockout mice, and western blot were performed to explore the specific mechanism of AM treatment in SCI. RESULTS Our data indicated that mice with SCI exposure exhibited motor dysfunction, a significant decrease of neuronal cells, a remarkable activation of astrocytes and microglia, an increase of IL-6, TNF-α, IL-18 expression, and an elevation of IL-18 colocalized with astrocytes, while astrocytes-specific NLRP3 knockout heavily reversed these changes. Besides, AM treatment simulated the neuroprotective effects of astrocyte-specific NLRP3 knockout, whereas an activator of NLRP3 nigericin partially reversed the AM neuroprotective effects. CONCLUSION AM treatment mitigates SCI-induced motor dysfunction in mice; this protective mechanism may be related to the NLRP3-IL18 signaling pathway inhibition in astrocytes.
Collapse
Affiliation(s)
- Ji-Hui Zheng
- Department of OrthopaedicsThe Graduate School, Tianjin Medical University, Tianjin, China
- Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research (Preparing), Cangzhou, China
| | - Na Yuan
- Department of Orthopaedics, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China
| | - Peng Zhang
- Department of Orthopaedics, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China
| | - De-Feng Liu
- Department of Orthopaedics, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China
| | - Wei Lin
- Department of Orthopaedics, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China
| | - Jun Miao
- Department of OrthopaedicsTianjin Hospital, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
14
|
High plasma calcitonin gene-related peptide and serum pituitary adenylate cyclase-activating polypeptide levels in patients with neuropathic pain. Rev Neurol (Paris) 2023; 179:289-296. [PMID: 36754669 DOI: 10.1016/j.neurol.2022.10.005] [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: 05/20/2022] [Revised: 10/06/2022] [Accepted: 10/13/2022] [Indexed: 02/09/2023]
Abstract
INTRODUCTION Based on animal studies, calcitonin gene-related peptide (CGRP) and pituitary adenylate cyclase-activating polypeptide (PACAP) are thought to play a role in neurobiological events such as neuropathic pain, neuroprotection, neurotransmission, neural plasticity, and neurotrophic effects. The aim of the study is to investigate whether there is a change in the blood level of CGRP and PACAP in patients with neuropathic pain and to look for clues about the utility of these peptides as pharmacological targets in the treatment of neuropathic pain in humans. METHODS The study included 60 polyneuropathy patients with neuropathic pain, 30 polyneuropathy patients without neuropathic pain (NNP) and 29 healthy subjects as control group. Polyneuropathy patients with neuropathic pain were divided into two groups as diabetic (D-PNP) and non-diabetic polyneuropathy (ND-PNP) patients. Plasma CGRP and serum PACAP levels were measured from venous blood samples of the patients and healthy controls. RESULTS The CGRP level was significantly higher in the D-PNP and ND-PNP groups compared to the control and NNP groups (P<0.05). PACAP levels were significantly higher in the D-PNP and ND-PNP groups compared to the control and NNP groups (P<0.05). There was no significant correlation between CGRP and PACAP levels and neuropathic pain scale (NPS). CONCLUSIONS This study is the first to demonstrate elevated plasma CGRP and serum PACAP levels in polyneuropathy patients with neuropathic pain. The results of this study are important in terms of showing that both CGRP and PACAP can be new pharmacological targets in the treatment of neuropathic pain and polyneuropathy in humans.
Collapse
|
15
|
Kambe Y, Youkai M, Hashiguchi K, Sameshima Y, Takasaki I, Miyata A, Kurihara T. Spinal Astrocyte-Neuron Lactate Shuttle Contributes to the Pituitary Adenylate Cyclase-Activating Polypeptide/PAC1 Receptor-Induced Nociceptive Behaviors in Mice. Biomolecules 2022; 12:biom12121859. [PMID: 36551287 PMCID: PMC9775268 DOI: 10.3390/biom12121859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
We have previously shown that spinal pituitary adenylate cyclase-activating polypeptide (PACAP)/PACAP type 1 (PAC1) receptor signaling triggered long-lasting nociceptive behaviors through astroglial activation in mice. Since astrocyte-neuron lactate shuttle (ANLS) could be essential for long-term synaptic facilitation, we aimed to elucidate a possible involvement of spinal ANLS in the development of the PACAP/PAC1 receptor-induced nociceptive behaviors. A single intrathecal administration of PACAP induced short-term spontaneous aversive behaviors, followed by long-lasting mechanical allodynia in mice. These nociceptive behaviors were inhibited by 1,4-dideoxy-1,4-imino-d-arabinitol (DAB), an inhibitor of glycogenolysis, and this inhibition was reversed by simultaneous L-lactate application. In the cultured spinal astrocytes, the PACAP-evoked glycogenolysis and L-lactate secretion were inhibited by DAB. In addition, a protein kinase C (PKC) inhibitor attenuated the PACAP-induced nociceptive behaviors as well as the PACAP-evoked glycogenolysis and L-lactate secretion. Finally, an inhibitor for the monocarboxylate transporters blocked the L-lactate secretion from the spinal astrocytes and inhibited the PACAP- and spinal nerve ligation-induced nociceptive behaviors. These results suggested that spinal PAC1 receptor-PKC-ANLS signaling contributed to the PACAP-induced nociceptive behaviors. This signaling system could be involved in the peripheral nerve injury-induced pain-like behaviors.
Collapse
Affiliation(s)
- Yuki Kambe
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Masafumi Youkai
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Kohei Hashiguchi
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Yoshimune Sameshima
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Ichiro Takasaki
- Department of Pharmacology, Graduate School of Science and Engineering, University of Toyama, Toyama 930-8555, Japan
| | - Atsuro Miyata
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Takashi Kurihara
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
- Correspondence: ; Tel.: +81-99-275-5256
| |
Collapse
|
16
|
Southey BR, Rodriguez-Zas SL. Alternative Splicing of Neuropeptide Prohormone and Receptor Genes Associated with Pain Sensitivity Was Detected with Zero-Inflated Models. Biomedicines 2022; 10:biomedicines10040877. [PMID: 35453627 PMCID: PMC9031102 DOI: 10.3390/biomedicines10040877] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/01/2022] [Accepted: 04/07/2022] [Indexed: 02/01/2023] Open
Abstract
Migraine is often accompanied by exacerbated sensitivity to stimuli and pain associated with alternative splicing of genes in signaling pathways. Complementary analyses of alternative splicing of neuropeptide prohormone and receptor genes involved in cell–cell communication in the trigeminal ganglia and nucleus accumbens regions of mice presenting nitroglycerin-elicited hypersensitivity and control mice were conducted. De novo sequence assembly detected 540 isoforms from 168 neuropeptide prohormone and receptor genes. A zero-inflated negative binomial model that accommodates for potential excess of zero isoform counts enabled the detection of 27, 202, and 12 differentially expressed isoforms associated with hypersensitivity, regions, and the interaction between hypersensitivity and regions, respectively. Skipped exons and alternative 3′ splice sites were the most frequent splicing events detected in the genes studied. Significant differential splicing associated with hypersensitivity was identified in CALCA and VGF neuropeptide prohormone genes and ADCYAP1R1, CRHR2, and IGF1R neuropeptide receptor genes. The prevalent region effect on differential isoform levels (202 isoforms) and alternative splicing (82 events) were consistent with the distinct splicing known to differentiate central nervous structures. Our findings highlight the changes in alternative splicing in neuropeptide prohormone and receptor genes associated with hypersensitivity to pain and the necessity to target isoform profiles for enhanced understanding and treatment of associated disorders such as migraine.
Collapse
Affiliation(s)
- Bruce R. Southey
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA;
- Correspondence:
| | - Sandra L. Rodriguez-Zas
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA;
- Department of Statistics, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
17
|
PACAP-38 Induces Transcriptomic Changes in Rat Trigeminal Ganglion Cells Related to Neuroinflammation and Altered Mitochondrial Function Presumably via PAC1/VPAC2 Receptor-Independent Mechanism. Int J Mol Sci 2022; 23:ijms23042120. [PMID: 35216232 PMCID: PMC8874739 DOI: 10.3390/ijms23042120] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 11/17/2022] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a broadly expressed neuropeptide which has diverse effects in both the peripheral and central nervous systems. While its neuroprotective effects have been shown in a variety of disease models, both animal and human data support the role of PACAP in migraine generation. Both PACAP and its truncated derivative PACAP(6-38) increased calcium influx in rat trigeminal ganglia (TG) primary sensory neurons in most experimental settings. PACAP(6-38), however, has been described as an antagonist for PACAP type I (known as PAC1), and Vasoactive Intestinal Polypeptide Receptor 2 (also known as VPAC2) receptors. Here, we aimed to compare the signaling pathways induced by the two peptides using transcriptomic analysis. Rat trigeminal ganglion cell cultures were incubated with 1 µM PACAP-38 or PACAP(6-38). Six hours later RNA was isolated, next-generation RNA sequencing was performed and transcriptomic changes were analyzed to identify differentially expressed genes. Functional analysis was performed for gene annotation using the Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Reactome databases. We found 200 common differentially expressed (DE) genes for these two neuropeptides. Both PACAP-38 and PACAP(6-38) treatments caused significant downregulation of NADH: ubiquinone oxidoreductase subunit B6 and upregulation of transient receptor potential cation channel, subfamily M, member 8. The common signaling pathways induced by both peptides indicate that they act on the same target, suggesting that PACAP activates trigeminal primary sensory neurons via a mechanism independent of the identified and cloned PAC1/VPAC2 receptor, either via another target structure or a different splice variant of PAC1/VPAC2 receptors. Identification of the target could help to understand key mechanisms of migraine.
Collapse
|
18
|
Takasaki I, Watanabe A, Okada T, Kanayama D, Nagashima R, Shudo M, Shimodaira A, Nunomura K, Lin B, Watanabe Y, Gouda H, Miyata A, Kurihara T, Toyooka N. Design and synthesis of pyrido[2,3-d]pyrimidine derivatives for a novel PAC1 receptor antagonist. Eur J Med Chem 2022; 231:114160. [DOI: 10.1016/j.ejmech.2022.114160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 11/04/2022]
|
19
|
Mandwie M, Karunia J, Niaz A, Keay KA, Musumeci G, Rennie C, McGrath K, Al-Badri G, Castorina A. Metformin Treatment Attenuates Brain Inflammation and Rescues PACAP/VIP Neuropeptide Alterations in Mice Fed a High-Fat Diet. Int J Mol Sci 2021; 22:ijms222413660. [PMID: 34948457 PMCID: PMC8706124 DOI: 10.3390/ijms222413660] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 12/28/2022] Open
Abstract
High-fat diet (HFD)-induced comorbid cognitive and behavioural impairments are thought to be the result of persistent low-grade neuroinflammation. Metformin, a first-line medication for the treatment of type-2 diabetes, seems to ameliorate these comorbidities, but the underlying mechanism(s) are not clear. Pituitary adenylate cyclase-activating peptide (PACAP) and vasoactive intestinal peptide (VIP) are neuroprotective peptides endowed with anti-inflammatory properties. Alterations to the PACAP/VIP system could be pivotal during the development of HFD-induced neuroinflammation. To unveil the pathogenic mechanisms underlying HFD-induced neuroinflammation and assess metformin’s therapeutic activities, (1) we determined if HFD-induced proinflammatory activity was present in vulnerable brain regions associated with the development of comorbid behaviors, (2) investigated if the PACAP/VIP system is altered by HFD, and (3) assessed if metformin rescues such diet-induced neurochemical alterations. C57BL/6J male mice were divided into two groups to receive either standard chow (SC) or HFD for 16 weeks. A further HFD group received metformin (HFD + M) (300 mg/kg BW daily for 5 weeks) via oral gavage. Body weight, fasting glucose, and insulin levels were measured. After 16 weeks, the proinflammatory profile, glial activation markers, and changes within the PI3K/AKT intracellular pathway and the PACAP/VIP system were evaluated by real-time qPCR and/or Western blot in the hypothalamus, hippocampus, prefrontal cortex, and amygdala. Our data showed that HFD causes widespread low-grade neuroinflammation and gliosis, with regional-specific differences across brain regions. HFD also diminished phospho-AKT(Ser473) expression and caused significant disruptions to the PACAP/VIP system. Treatment with metformin attenuated these neuroinflammatory signatures and reversed PI3K/AKT and PACAP/VIP alterations caused by HFD. Altogether, our findings demonstrate that metformin treatment rescues HFD-induced neuroinflammation in vulnerable brain regions, most likely by a mechanism involving the reinstatement of PACAP/VIP system homeostasis. Data also suggests that the PI3K/AKT pathway, at least in part, mediates some of metformin’s beneficial effects.
Collapse
Affiliation(s)
- Mawj Mandwie
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (M.M.); (J.K.); (A.N.); (G.A.-B.)
| | - Jocelyn Karunia
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (M.M.); (J.K.); (A.N.); (G.A.-B.)
| | - Aram Niaz
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (M.M.); (J.K.); (A.N.); (G.A.-B.)
| | - Kevin A. Keay
- Laboratory of Neural Structure and Function, School of Medical Science (Neuroscience), University of Sydney, Sydney, NSW 2006, Australia;
| | - Giuseppe Musumeci
- Department of Biomedical and Biotechnological Sciences, Anatomy, Histology and Movement Sciences Section, School of Medicine, University of Catania, 95125 Catania, Italy;
| | - Claire Rennie
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (C.R.); (K.M.)
| | - Kristine McGrath
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (C.R.); (K.M.)
| | - Ghaith Al-Badri
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (M.M.); (J.K.); (A.N.); (G.A.-B.)
| | - Alessandro Castorina
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (M.M.); (J.K.); (A.N.); (G.A.-B.)
- Laboratory of Neural Structure and Function, School of Medical Science (Neuroscience), University of Sydney, Sydney, NSW 2006, Australia;
- Correspondence:
| |
Collapse
|
20
|
Liang Y, Ma Y, Wang J, Nie L, Hou X, Wu W, Zhang X, Tian Y. Leptin Contributes to Neuropathic Pain via Extrasynaptic NMDAR-nNOS Activation. Mol Neurobiol 2021; 58:1185-1195. [PMID: 33099751 PMCID: PMC7878206 DOI: 10.1007/s12035-020-02180-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 10/14/2020] [Indexed: 11/28/2022]
Abstract
Leptin is an adipocytokine that is primarily secreted by white adipose tissue, and it contributes to the pathogenesis of neuropathic pain in collaboration with N-methyl-D-aspartate receptors (NMDARs). Functional NMDARs are a heteromeric complex that primarily comprise two NR1 subunits and two NR2 subunits. NR2A is preferentially located at synaptic sites, and NR2B is enriched at extrasynaptic sites. The roles of synaptic and extrasynaptic NMDARs in the contribution of leptin to neuropathic pain are not clear. The present study examined whether the important role of leptin in neuropathic pain was related to synaptic or extrasynaptic NMDARs. We used a rat model of spared nerve injury (SNI) and demonstrated that the intrathecal administration of the NR2A-selective antagonist NVP-AAM077 and the NR2B-selective antagonist Ro25-6981 prevented and reversed mechanical allodynia following SNI. Administration of exogenous leptin mimicked SNI-induced behavioral allodynia, which was also prevented by NVP-AAM077 and Ro25-6981. Mechanistic studies showed that leptin enhanced NR2B- but not NR2A-mediated currents in spinal lamina II neurons of naïve rats. Leptin also upregulated the expression of NR2B, which was blocked by the NR2B-selective antagonist Ro25-6981, in cultured dorsal root ganglion (DRG) neurons. Leptin enhanced neuronal nitric oxide synthase (nNOS) expression, which was also blocked by Ro25-6981, in cultured DRG cells. However, leptin did not change NR2A expression, and the NR2A-selective antagonist NVP-AAM077 had no effect on leptin-enhanced nNOS expression. Our data suggest an important cellular link between the spinal effects of leptin and the extrasynaptic NMDAR-nNOS-mediated cellular mechanism of neuropathic pain.
Collapse
Affiliation(s)
- Yanling Liang
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, No. 1838 Guangzhou Avenue, Guangzhou, 510515, China
| | - Yuxin Ma
- Department of Anatomy, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Jieqin Wang
- Department of Pancreatobiliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510515, China
| | - Lei Nie
- Department of Anesthesiology, The Third Xiangya Hospital of Central South University, Changsha, 410000, China
| | - Xusheng Hou
- Department of Functional Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Wenyu Wu
- Target and Interventional Therapy Department of Oncology, First People's Hospital of Foshan, Foshan, 528000, China
| | - Xingmei Zhang
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, No. 1838 Guangzhou Avenue, Guangzhou, 510515, China.
| | - Yinghong Tian
- Experiment Teaching & Administration Center, School of Basic Medical Sciences, Southern Medical University, No. 1838 Guangzhou Avenue, Guangzhou, 510515, China.
| |
Collapse
|
21
|
Hayata-Takano A, Shintani Y, Moriguchi K, Encho N, Kitagawa K, Nakazawa T, Hashimoto H. PACAP-PAC1 Signaling Regulates Serotonin 2A Receptor Internalization. Front Endocrinol (Lausanne) 2021; 12:732456. [PMID: 34759890 PMCID: PMC8574227 DOI: 10.3389/fendo.2021.732456] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 09/27/2021] [Indexed: 11/26/2022] Open
Abstract
Mice lacking pituitary adenylate cyclase-activating polypeptide (PACAP) display psychomotor abnormalities, most of which are ameliorated by atypical antipsychotics with serotonin (5-HT) 2A receptor (5-HT2A) antagonism. Heterozygous Pacap mutant mice show a significantly higher hallucinogenic response than wild-type mice to a 5-HT2A agonist. Endogenous PACAP may, therefore, affect 5-HT2A signaling; however, the underlying neurobiological mechanism for this remains unclear. Here, we examined whether PACAP modulates 5-HT2A signaling by addressing cellular protein localization. PACAP induced an increase in internalization of 5-HT2A but not 5-HT1A, 5-HT2C, dopamine D2 receptors or metabotropic glutamate receptor 2 in HEK293T cells. This PACAP action was inhibited by protein kinase C inhibitors, β-arrestin2 silencing, the PACAP receptor PAC1 antagonist PACAP6-38, and PAC1 silencing. In addition, the levels of endogenous 5-HT2A were decreased on the cell surface of primary cultured cortical neurons after PACAP stimulation and were increased in frontal cortex cell membranes of Pacap-/- mice. Finally, intracerebroventricular PACAP administration suppressed 5-HT2A agonist-induced head twitch responses in mice. These results suggest that PACAP-PAC1 signaling increases 5-HT2A internalization resulting in attenuation of 5-HT2A-mediated signaling, although further study is necessary to determine the relationship between behavioral abnormalities in Pacap-/- mice and PACAP-induced 5-HT2A internalization.
Collapse
Affiliation(s)
- Atsuko Hayata-Takano
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
- Molecular Research Center for Children’s Mental Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Japan
- *Correspondence: Hitoshi Hashimoto, ; Atsuko Hayata-Takano,
| | - Yusuke Shintani
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Keita Moriguchi
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Naoki Encho
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Kohei Kitagawa
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Takanobu Nakazawa
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
- Department of Bioscience, Tokyo University of Agriculture, Setagaya-ku, Japan
| | - Hitoshi Hashimoto
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
- Molecular Research Center for Children’s Mental Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Japan
- Division of Bioscience, Institute for Datability Science, Osaka University, Suita, Japan
- Transdimensional Life Imaging Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
- Department of Molecular Pharmaceutical Science, Graduate School of Medicine, Osaka University, Suita, Japan
- *Correspondence: Hitoshi Hashimoto, ; Atsuko Hayata-Takano,
| |
Collapse
|
22
|
Ke R, Lok SIS, Singh K, Chow BKC, Lee LTO. GIP receptor suppresses PAC1receptor-mediated neuronal differentiation via formation of a receptor heterocomplex. J Neurochem 2020; 157:1850-1860. [PMID: 33078390 DOI: 10.1111/jnc.15220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 10/07/2020] [Accepted: 10/13/2020] [Indexed: 01/22/2023]
Abstract
Pituitary adenylate cyclase-activating peptide (PACAP) receptor (PAC1R) is a class B Gprotein-coupled receptor (GPCR) that is widely expressed in the human body and is involved in neuronal differentiation. As class B GPCRs are known to form heterocomplexes with family members, we hypothesized that PAC1R mediates neuronal differentiation through interaction with a class B GPCR. We used the BRET assay to identify potential interactions between PAC1R and 11 class B GPCRs. Gastric inhibitory polypeptide receptor (GIPR) and secretin receptor were identified as putative binding partners of PAC1R. The effect of heterocomplex formation by PAC1R on receptor activation was evaluated with the cyclic (c)AMP, luciferase reporter, and calcium signaling assays; and the effects on receptor internalization and subcellular localization were examined by confocal microscopy. The results suggested he PAC1R/GIPR heterocomplex suppressed signaling events downstream of PAC1R, including cAMP production, serum response element and calcium signaling, and β-arrestin recruitment. Protein-protein interaction was analyzed in silico, and induction of neuronal differentiation by the PAC1R heterocomplex was assessed in SH-SY5Y neuronal cells by measure the morphological changes and marker genes expression by real-time quantitative PCR and western blot. Over-expression of GIPR suppressed PACAP/PAC1R-mediated neuronal differentiation and the differentiation markers expression in SH-SY5Y cells. GIPR regulates neuronal differentiation through heterocomplex formation with PAC1R.
Collapse
Affiliation(s)
- Ran Ke
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Samson I S Lok
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Kailash Singh
- School of Biological Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Billy K C Chow
- School of Biological Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Leo T O Lee
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau, China.,Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| |
Collapse
|
23
|
Vollesen ALH, Snoer A, Chaudhry B, Petersen AS, Hagedorn A, Hoffmann J, Jensen RH, Ashina M. The effect of pituitary adenylate cyclase-activating peptide-38 and vasoactive intestinal peptide in cluster headache. Cephalalgia 2020; 40:1474-1488. [PMID: 32962406 DOI: 10.1177/0333102420940689] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Previously reported increases in serum levels of vasodilating neuropeptides pituitary adenylate cyclase-activating peptide-38 (PACAP38) and vasoactive intestinal peptide (VIP) during attacks of cluster headache could indicate their involvement in cluster headache attack initiation. We investigated the attack-inducing effects of PACAP38 and vasoactive intestinal peptide in cluster headache, hypothesising that PACAP38, but not vasoactive intestinal peptide, would induce cluster-like attacks in episodic active phase and chronic cluster headache patients. METHODS In a double-blind crossover study, 14 episodic cluster headache patients in active phase, 15 episodic cluster headache patients in remission phase and 15 chronic cluster headache patients were randomly allocated to receive intravenous infusion of PACAP38 (10 pmol/kg/min) or vasoactive intestinal peptide (8 pmol/kg/min) over 20 min on two study days separated by at least 7 days. We recorded headache intensity, incidence of cluster-like attacks, cranial autonomic symptoms and vital signs using a questionnaire (0-90 min). RESULTS In episodic cluster headache active phase, PACAP38 induced cluster-like attacks in 6/14 patients and vasoactive intestinal peptide induced cluster-like attacks in 5/14 patients (p = 1.000). In chronic cluster headache, PACAP38 and vasoactive intestinal peptide both induced cluster-like attacks in 7/15 patients (p = 0.765). In episodic cluster headache remission phase, neither PACAP38 nor vasoactive intestinal peptide induced cluster-like attacks. CONCLUSIONS Contrary to our hypothesis, attack induction was lower than expected and roughly equal by PACAP38 and vasoactive intestinal peptide in episodic active phase and chronic cluster headache patients, which contradicts the PAC1-receptor as being solely responsible for attack induction.Trial registration: clinicaltrials.gov (identifier NCT03814226).
Collapse
Affiliation(s)
- Anne Luise H Vollesen
- Danish Headache Center and Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Agneta Snoer
- Danish Headache Center and Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Basit Chaudhry
- Danish Headache Center and Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anja Sofie Petersen
- Danish Headache Center and Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Andreas Hagedorn
- Danish Headache Center and Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jan Hoffmann
- Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Rigmor H Jensen
- Danish Headache Center and Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Messoud Ashina
- Danish Headache Center and Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
24
|
Takasaki I, Ogashi H, Okada T, Shimodaira A, Hayakawa D, Watanabe A, Miyata A, Kurihara T, Gouda H, Toyooka N. Synthesis of a novel and potent small-molecule antagonist of PAC1 receptor for the treatment of neuropathic pain. Eur J Med Chem 2020; 186:111902. [DOI: 10.1016/j.ejmech.2019.111902] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/30/2019] [Accepted: 11/18/2019] [Indexed: 11/25/2022]
|
25
|
Ueda H. LPA receptor signaling as a therapeutic target for radical treatment of neuropathic pain and fibromyalgia. Pain Manag 2019; 10:43-53. [PMID: 31852400 DOI: 10.2217/pmt-2019-0036] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Since the first discovery that the bioactive lipid, lysophosphatidic acid (LPA) and LPA1 receptor signaling play a role in the initiation of neuropathic pain (NeuP), accumulated reports have supported the original findings and extended the study toward possible therapeutic applications. The present review describes beneficial roles of LPA receptor signaling in a variety of chronic pain, such as peripheral NeuP induced by nerve injury, chemotherapy and diabetes, central NeuP induced by cerebral ischemia with hemorrhage and spinal cord injury, and fibromyalgia-like wide spread pain induced by repeated cold, psychological and muscular acidic stress. Emerging mechanistic findings are the feed-forward amplification of LPA production through LPA1, LPA3 and microglia and the evidence for maintenance of chronic pain by LPA receptor signaling.
Collapse
Affiliation(s)
- Hiroshi Ueda
- Department of Molecular Pharmacology, Kyoto University Graduate School of Pharmaceutical Sciences, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
26
|
Pituitary Adenylate Cyclase-Activating Polypeptide Modulates Dendritic Spine Maturation and Morphogenesis via MicroRNA-132 Upregulation. J Neurosci 2019; 39:4208-4220. [PMID: 30886013 DOI: 10.1523/jneurosci.2468-18.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 02/18/2019] [Accepted: 03/13/2019] [Indexed: 12/11/2022] Open
Abstract
Alterations in pituitary adenylate cyclase-activating polypeptide (PACAP), a multifunctional neuropeptide, and its receptors have been identified as risk factors for certain psychiatric disorders, including schizophrenia. Increasing evidence from human genetic and animal model studies suggest an association between various psychiatric disorders and altered dendritic spine morphology. In the present study, we investigated the role of exogenous and endogenous PACAP in spine formation and maturation. PACAP modified the density and morphology of PSD-95-positive spines in primary cultured hippocampal neurons. Notably, PACAP increased the levels of microRNA (miR)-132 and decreased expression of corresponding miR-132 target genes and protein expression of p250GAP, a miR-132 effector known to be involved in spine morphology regulation. In corroboration, PSD-95-positive spines were reduced in PACAP-deficient (PACAP -/-) mice versus WT mice. Golgi staining of hippocampal CA1 neurons revealed a reduced spine densities and atypical morphologies in the male PACAP -/- mice. Furthermore, viral miR-132 overexpression reversed the reduction in hippocampal spinal density in the male PACAP -/- mice. These results indicate that PACAP signaling plays a critical role in spine morphogenesis possibly via miR-132. We suggest that dysfunction of PACAP signaling may contribute to the pathogenesis of neuropsychiatric disorders, at least partly through its effects on spine formation.SIGNIFICANCE STATEMENT Pituitary adenylate cyclase-activating polypeptide (PACAP) signaling dysfunction and dendritic spine morphology alterations have recently been suggested as important pathophysiological mechanisms underlying several psychiatric and neurological disorders. In this study, we investigated whether PACAP regulates dendritic spine morphogenesis. In a combination of pharmacological and viral gain- and loss-of-function approaches in vitro and in vivo experiments, we found PACAP to increase the size and density of dendritic spines via miR-132 upregulation. Together, our data suggest that a dysfunction of PACAP signaling may contribute to the pathogenesis of neuropsychiatric disorders, at least partly through abnormal spine formation.
Collapse
|
27
|
Shintani Y, Hayata-Takano A, Moriguchi K, Nakazawa T, Ago Y, Kasai A, Seiriki K, Shintani N, Hashimoto H. β-Arrestin1 and 2 differentially regulate PACAP-induced PAC1 receptor signaling and trafficking. PLoS One 2018; 13:e0196946. [PMID: 29734363 PMCID: PMC5937772 DOI: 10.1371/journal.pone.0196946] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 04/23/2018] [Indexed: 01/14/2023] Open
Abstract
A pituitary adenylate cyclase-activating polypeptide (PACAP)-specific receptor, PAC1R, is coupled with multiple signal transduction pathways including stimulation of adenylate cyclase, phospholipase C and extracellular-signal regulated kinase (ERK)1/2. PAC1R has been shown to exert its long-lasting and potent signals via β-arrestin1 and β-arrestin2. However, the precise roles of the two β-arrestin isoforms in PACAP-PAC1R signaling remain unclear. Here we examined the interaction between the two β-arrestin isoforms and PAC1R, β-arrestin-dependent PAC1R subcellular localization and ERK1/2 activation. Upon PACAP stimulation, although PAC1R similarly interacted with β-arrestin1 and β-arrestin2 in HEK293T cells, the complex of PAC1R and β-arrestin2 was translocated from the cell surface into cytosol, but that of β-arrestin1 remained in the cell surface regions in HeLa cells and mouse primary cultured neurons. Silencing of β-arrestin2 blocked PACAP-induced PAC1R internalization and ERK1/2 phosphorylation, but silencing of β-arrestin1 increased ERK1/2 phosphorylation. These results show that β-arrestin1 and β-arrestin2 exert differential actions on PAC1R internalization and PAC1R-dependent ERK1/2 activation, and suggest that the two β-arrestin isoforms may be involved in fine and precise tuning of the PAC1R signaling pathways.
Collapse
Affiliation(s)
- Yusuke Shintani
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Atsuko Hayata-Takano
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
- Center for Child Mental Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Osaka, Japan
- * E-mail: (HS); (AH-T)
| | - Keita Moriguchi
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Takanobu Nakazawa
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
- Department of Pharmacology, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Yukio Ago
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Atsushi Kasai
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Kaoru Seiriki
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
- Interdisciplinary Program for Biomedical Sciences, Institute for Academic Initiatives, Osaka University, Suita, Osaka, Japan
| | - Norihito Shintani
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Hitoshi Hashimoto
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
- Center for Child Mental Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Osaka, Japan
- Division of Bioscience, Institute for Datability Science, Osaka University, Suita, Osaka, Japan
- * E-mail: (HS); (AH-T)
| |
Collapse
|
28
|
Takasaki I, Watanabe A, Yokai M, Watanabe Y, Hayakawa D, Nagashima R, Fukuchi M, Okada T, Toyooka N, Miyata A, Gouda H, Kurihara T. In Silico Screening Identified Novel Small-molecule Antagonists of PAC1 Receptor. J Pharmacol Exp Ther 2018; 365:1-8. [PMID: 29363578 DOI: 10.1124/jpet.117.245415] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 01/18/2018] [Indexed: 01/07/2023] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) and its receptors are present in the spinal dorsal horn and dorsal root ganglia, suggesting an important role of PACAP signaling systems in the modulation of spinal nociceptive transmission. Previously, we found that intrathecal injection of PACAP or maxadilan, a selective PACAP type I (PAC1) receptor agonist, induced transient aversive responses followed by a long-lasting mechanical allodynia in mice, suggesting that PACAP-PAC1 receptor systems are involved in chronic pain and that selective PAC1 antagonists may become a new class of analgesics. Although several PAC1 antagonists, such as PACAP 6-38, have been reported, all of them are peptide compounds. In the present study, we identified new small-molecule antagonists of the PAC1 receptor using in silico screening and in vitro/vivo pharmacological assays. The identified small-molecule compounds, named PA-8 and PA-9, dose dependently inhibited the phosphorylation of CREB induced by PACAP in PAC1-, but not VPAC1- or VPAC2-receptor-expressing CHO cells. PA-8 and PA-9 also dose dependently inhibited PACAP-induced cAMP elevation with an IC50 of 2.0 and 5.6 nM, respectively. In vivo pharmacological assays showed that intrathecal injection of these compounds blocked the induction of PACAP-induced aversive responses and mechanical allodynia in mice. In contrast, the compounds when administered alone exerted neither agonistic nor algesic actions in the in vitro/vivo assays. The compounds identified in the present study are new and the first small-molecule antagonists of the PAC1 receptor; they may become seed compounds for developing novel analgesics.
Collapse
Affiliation(s)
- Ichiro Takasaki
- Department of Pharmacology, Graduate School of Science and Engineering (I.T., A.W., R.N.), Graduate School of Innovative Life Sciences (I.T., T.O., N.T.), Department of Molecular Neurobiology, Graduate School of Medical and Pharmaceutical Sciences (M.F.), and Department of Bio-functional Molecular Engineering, Graduate School of Science and Engineering (T.O., N.T.), University of Toyama, Toyama, Japan; Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan (M.Y., A.M., T.K.); and Department of Analytical and Physical Chemistry, School of Pharmacy, Showa University, Tokyo, Japan (Y.W., D.H., H.G.)
| | - Ai Watanabe
- Department of Pharmacology, Graduate School of Science and Engineering (I.T., A.W., R.N.), Graduate School of Innovative Life Sciences (I.T., T.O., N.T.), Department of Molecular Neurobiology, Graduate School of Medical and Pharmaceutical Sciences (M.F.), and Department of Bio-functional Molecular Engineering, Graduate School of Science and Engineering (T.O., N.T.), University of Toyama, Toyama, Japan; Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan (M.Y., A.M., T.K.); and Department of Analytical and Physical Chemistry, School of Pharmacy, Showa University, Tokyo, Japan (Y.W., D.H., H.G.)
| | - Masafumi Yokai
- Department of Pharmacology, Graduate School of Science and Engineering (I.T., A.W., R.N.), Graduate School of Innovative Life Sciences (I.T., T.O., N.T.), Department of Molecular Neurobiology, Graduate School of Medical and Pharmaceutical Sciences (M.F.), and Department of Bio-functional Molecular Engineering, Graduate School of Science and Engineering (T.O., N.T.), University of Toyama, Toyama, Japan; Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan (M.Y., A.M., T.K.); and Department of Analytical and Physical Chemistry, School of Pharmacy, Showa University, Tokyo, Japan (Y.W., D.H., H.G.)
| | - Yurie Watanabe
- Department of Pharmacology, Graduate School of Science and Engineering (I.T., A.W., R.N.), Graduate School of Innovative Life Sciences (I.T., T.O., N.T.), Department of Molecular Neurobiology, Graduate School of Medical and Pharmaceutical Sciences (M.F.), and Department of Bio-functional Molecular Engineering, Graduate School of Science and Engineering (T.O., N.T.), University of Toyama, Toyama, Japan; Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan (M.Y., A.M., T.K.); and Department of Analytical and Physical Chemistry, School of Pharmacy, Showa University, Tokyo, Japan (Y.W., D.H., H.G.)
| | - Daichi Hayakawa
- Department of Pharmacology, Graduate School of Science and Engineering (I.T., A.W., R.N.), Graduate School of Innovative Life Sciences (I.T., T.O., N.T.), Department of Molecular Neurobiology, Graduate School of Medical and Pharmaceutical Sciences (M.F.), and Department of Bio-functional Molecular Engineering, Graduate School of Science and Engineering (T.O., N.T.), University of Toyama, Toyama, Japan; Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan (M.Y., A.M., T.K.); and Department of Analytical and Physical Chemistry, School of Pharmacy, Showa University, Tokyo, Japan (Y.W., D.H., H.G.)
| | - Ryota Nagashima
- Department of Pharmacology, Graduate School of Science and Engineering (I.T., A.W., R.N.), Graduate School of Innovative Life Sciences (I.T., T.O., N.T.), Department of Molecular Neurobiology, Graduate School of Medical and Pharmaceutical Sciences (M.F.), and Department of Bio-functional Molecular Engineering, Graduate School of Science and Engineering (T.O., N.T.), University of Toyama, Toyama, Japan; Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan (M.Y., A.M., T.K.); and Department of Analytical and Physical Chemistry, School of Pharmacy, Showa University, Tokyo, Japan (Y.W., D.H., H.G.)
| | - Mamoru Fukuchi
- Department of Pharmacology, Graduate School of Science and Engineering (I.T., A.W., R.N.), Graduate School of Innovative Life Sciences (I.T., T.O., N.T.), Department of Molecular Neurobiology, Graduate School of Medical and Pharmaceutical Sciences (M.F.), and Department of Bio-functional Molecular Engineering, Graduate School of Science and Engineering (T.O., N.T.), University of Toyama, Toyama, Japan; Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan (M.Y., A.M., T.K.); and Department of Analytical and Physical Chemistry, School of Pharmacy, Showa University, Tokyo, Japan (Y.W., D.H., H.G.)
| | - Takuya Okada
- Department of Pharmacology, Graduate School of Science and Engineering (I.T., A.W., R.N.), Graduate School of Innovative Life Sciences (I.T., T.O., N.T.), Department of Molecular Neurobiology, Graduate School of Medical and Pharmaceutical Sciences (M.F.), and Department of Bio-functional Molecular Engineering, Graduate School of Science and Engineering (T.O., N.T.), University of Toyama, Toyama, Japan; Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan (M.Y., A.M., T.K.); and Department of Analytical and Physical Chemistry, School of Pharmacy, Showa University, Tokyo, Japan (Y.W., D.H., H.G.)
| | - Naoki Toyooka
- Department of Pharmacology, Graduate School of Science and Engineering (I.T., A.W., R.N.), Graduate School of Innovative Life Sciences (I.T., T.O., N.T.), Department of Molecular Neurobiology, Graduate School of Medical and Pharmaceutical Sciences (M.F.), and Department of Bio-functional Molecular Engineering, Graduate School of Science and Engineering (T.O., N.T.), University of Toyama, Toyama, Japan; Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan (M.Y., A.M., T.K.); and Department of Analytical and Physical Chemistry, School of Pharmacy, Showa University, Tokyo, Japan (Y.W., D.H., H.G.)
| | - Atsuro Miyata
- Department of Pharmacology, Graduate School of Science and Engineering (I.T., A.W., R.N.), Graduate School of Innovative Life Sciences (I.T., T.O., N.T.), Department of Molecular Neurobiology, Graduate School of Medical and Pharmaceutical Sciences (M.F.), and Department of Bio-functional Molecular Engineering, Graduate School of Science and Engineering (T.O., N.T.), University of Toyama, Toyama, Japan; Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan (M.Y., A.M., T.K.); and Department of Analytical and Physical Chemistry, School of Pharmacy, Showa University, Tokyo, Japan (Y.W., D.H., H.G.)
| | - Hiroaki Gouda
- Department of Pharmacology, Graduate School of Science and Engineering (I.T., A.W., R.N.), Graduate School of Innovative Life Sciences (I.T., T.O., N.T.), Department of Molecular Neurobiology, Graduate School of Medical and Pharmaceutical Sciences (M.F.), and Department of Bio-functional Molecular Engineering, Graduate School of Science and Engineering (T.O., N.T.), University of Toyama, Toyama, Japan; Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan (M.Y., A.M., T.K.); and Department of Analytical and Physical Chemistry, School of Pharmacy, Showa University, Tokyo, Japan (Y.W., D.H., H.G.)
| | - Takashi Kurihara
- Department of Pharmacology, Graduate School of Science and Engineering (I.T., A.W., R.N.), Graduate School of Innovative Life Sciences (I.T., T.O., N.T.), Department of Molecular Neurobiology, Graduate School of Medical and Pharmaceutical Sciences (M.F.), and Department of Bio-functional Molecular Engineering, Graduate School of Science and Engineering (T.O., N.T.), University of Toyama, Toyama, Japan; Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan (M.Y., A.M., T.K.); and Department of Analytical and Physical Chemistry, School of Pharmacy, Showa University, Tokyo, Japan (Y.W., D.H., H.G.)
| |
Collapse
|
29
|
Abstract
Pituitary adenylate cyclase-activating peptide (PACAP) is a neuropeptide implicated in a wide range of functions, such as nociception and in primary headaches. Regarding its localization, PACAP has been observed in the sensory trigeminal ganglion (TG), in the parasympathetic sphenopalatine (SPG) and otic ganglia (OTG), and in the brainstem trigeminocervical complex. Immunohistochemistry has shown PACAP-38 in numerous cell bodies of SPG/OTG, co-stored with vasoactive intestinal peptide (VIP), nitric oxide synthase (NOS) and, to a minor degree, with choline acetyltransferase. PACAP has in addition been found in a subpopulation of calcitonin gene-related peptide (CGRP)-immunoreactive cells in the trigeminal system. The PACAP/VIP receptors (PAC1, VPAC1, and VPAC2) are present in sensory neurons and in vascular smooth muscle related to the trigeminovascular system. It is postulated that PACAP is involved in nociception. In support, abolishment of PACAP synthesis or reception leads to diminished pain responses, whereas systemic PACAP-38 infusion triggers pain behavior in animals and delayed migraine-like attacks in migraine patients without marked vasodilatory effects. In addition, increased plasma levels have been documented in acute migraine attacks and in cluster headache, in accordance with findings in experimental models of trigeminal activation. This suggest that the activation of the trigeminal system may result in elevated venous levels of PACAP, a change that can be reduced when headache is treated. The data presented in this review indicate that PACAP and its receptors may be promising targets for migraine therapeutics.
Collapse
|
30
|
Körtési T, Tuka B, Tajti J, Bagoly T, Fülöp F, Helyes Z, Vécsei L. Kynurenic Acid Inhibits the Electrical Stimulation Induced Elevated Pituitary Adenylate Cyclase-Activating Polypeptide Expression in the TNC. Front Neurol 2018; 8:745. [PMID: 29387039 PMCID: PMC5775965 DOI: 10.3389/fneur.2017.00745] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 12/26/2017] [Indexed: 12/13/2022] Open
Abstract
Background Migraine is a primary headache of imprecisely known mechanism, but activation of the trigeminovascular system (TS) appears to be essential during the attack. Intensive research has recently focused on pituitary adenylate cyclase-activating polypeptide (PACAP) and the kynurenine systems as potential pathogenic factors. Aim We investigated the link between these important mediators and the effects of kynurenic acid (KYNA) and its synthetic analog (KYNA-a) on PACAP expression in the rat trigeminal nucleus caudalis (TNC) in a TS stimulation model related to migraine mechanisms. Methods Adult male Sprague-Dawley rats were pretreated with KYNA, KYNA-a, the NMDA receptor antagonist MK-801, or saline (vehicle). Next, the trigeminal ganglion (TRG) was electrically stimulated, the animals were transcardially perfused following 180 min, and the TNC was removed. In the TNC samples, 38 amino acid form of PACAP (PACAP1–38)-like radioimmunoactivity was measured by radioimmunoassay, the relative optical density of preproPACAP was assessed by Western blot analysis, and PACAP1–38 mRNA was detected by real-time PCR. Results and conclusion Electrical TRG stimulation resulted in significant increases of PACAP1–38-LI, preproPACAP, and PACAP1–38 mRNA in the TNC. These increases were prevented by the pretreatments with KYNA, KYNA-a, and MK-801. This is the first study to provide evidence for a direct link between PACAP and the kynurenine system during TS activation.
Collapse
Affiliation(s)
- Tamás Körtési
- Faculty of Medicine, Department of Neurology, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - Bernadett Tuka
- MTA-SZTE Neuroscience Research Group, University of Szeged, Szeged, Hungary
| | - János Tajti
- Faculty of Medicine, Department of Neurology, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - Teréz Bagoly
- Faculty of Medicine, Department of Pharmacology and Pharmacotherapy, University of Pécs, Pécs, Hungary
| | - Ferenc Fülöp
- Faculty of Pharmacy, Institute of Pharmaceutical Chemistry, University of Szeged, Szeged, Hungary.,MTA-SZTE Stereochemistry Research Group, Hungarian Academy of Sciences, Szeged, Hungary
| | - Zsuzsanna Helyes
- Faculty of Medicine, Department of Pharmacology and Pharmacotherapy, University of Pécs, Pécs, Hungary.,János Szentágothai Research Center, University of Pécs, Pécs, Hungary.,MTA-PTE NAP B Chronic Pain Research Group, University of Pécs, Pécs, Hungary
| | - László Vécsei
- Faculty of Medicine, Department of Neurology, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary.,MTA-SZTE Neuroscience Research Group, University of Szeged, Szeged, Hungary
| |
Collapse
|
31
|
Kambe Y, Kurihara T, Miyata A. [Astrocyte-neuron lactate shuttle, the major effector of astrocytic PACAP signaling for CNS functions]. Nihon Yakurigaku Zasshi 2018; 151:239-243. [PMID: 29887572 DOI: 10.1254/fpj.151.239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Transfer of lactate from astrocytes to neurons is activated when synaptic activity is increased, and this mechanism is now known as the astrocyte-neuron lactate shuttle (ANLS), that could account for the coupling between synaptic activity and energy delivery. Many lines of evidence suggested that ANLS contributes to neuronal activation or synaptic plasticity at the cellular level as well as learning/memory and cocaine addiction at the behavioral level. However, the candidate neurotransmitters which evoke ANLS activation are still under discussion. Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neurotransmitter which distributed widely in central nervous system. Since PACAP might activate ANLS from very low concentration in cultured forebrain astrocytes, PACAP might be one of the candidates for the endogenous ANLS activator. In the present study, we investigated the potential relevance of PACAP/ANLS signaling in the learning/memory and spinal nociceptive transmission. In this study, we made the following findings: 1) PACAP could be an endogenous inducer for ANLS activation in central nervous system; 2) ANLS activation by PACAP/PAC1 receptor signaling contributed to learning/memory and induced long-lasting nociceptive behaviors; 3) PKC activation played an important role in the PACAP/PAC1 receptor-evoked ANLS.
Collapse
Affiliation(s)
- Yuki Kambe
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University
| | - Takashi Kurihara
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University
| | - Atsuro Miyata
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University
| |
Collapse
|
32
|
Sundrum T, Walker CS. Pituitary adenylate cyclase-activating polypeptide receptors in the trigeminovascular system: implications for migraine. Br J Pharmacol 2017; 175:4109-4120. [PMID: 28977676 DOI: 10.1111/bph.14053] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 08/24/2017] [Accepted: 09/11/2017] [Indexed: 12/13/2022] Open
Abstract
The neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) has been implicated in a wide range of functions including vasodilatation, neuroprotection, nociception and neurogenic inflammation. PACAP activates three distinct receptors, the PAC1 receptor, which responds to PACAP, and the VPAC1 and VPAC2 receptors, which respond to both PACAP and vasoactive intestinal polypeptide. The trigeminovascular system plays a key role in migraine and contains the trigeminal nerve, which is the major conduit of craniofacial pain. PACAP is expressed throughout the trigeminovascular system and in higher brain regions involved in processing pain. Evidence from human clinical studies suggests that PACAP may act outside the blood-brain barrier in the pathogenesis of migraine. However, the precise mechanisms involved remain unclear. PACAP potentially induces migraine attacks by activating different receptors in different cell types and tissues. This complexity prompted this review of PACAP receptor pharmacology, expression and function in the trigeminovascular system. Current evidence suggests that the PAC1 receptor is the likely pathophysiological target of PACAP in migraine. However, multiple PACAP receptors are expressed in key parts of the trigeminovascular system and further work is required to determine their contribution to PACAP physiology and the pathology of migraine. LINKED ARTICLES This article is part of a themed section on Molecular Pharmacology of GPCRs. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.21/issuetoc.
Collapse
Affiliation(s)
- Tahlia Sundrum
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Christopher S Walker
- School of Biological Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre and Centre for Brain Research, University of Auckland, Auckland, New Zealand
| |
Collapse
|
33
|
Altered Gene Expression of RNF34 and PACAP Possibly Involved in Mechanism of Exercise-Induced Analgesia for Neuropathic Pain in Rats. Int J Mol Sci 2017; 18:ijms18091962. [PMID: 28902127 PMCID: PMC5618611 DOI: 10.3390/ijms18091962] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 08/29/2017] [Accepted: 09/04/2017] [Indexed: 01/19/2023] Open
Abstract
Despite the availability of several modalities of treatment, including surgery, pharmacological agents, and nerve blocks, neuropathic pain is often unresponsive and sometimes progresses to intractable chronic pain. Although exercise therapy is a candidate for treatment of neuropathic pain, the mechanism underlying its efficacy has not been elucidated. To clarify the molecular mechanism for pain relief induced by exercise, we measured Rnf34 and Pacap mRNA levels in the spinal cord dorsal horn of SNL rats, a model of neuropathic pain. SNL model rats exhibited stable mechanical hyperalgesia for at least 6 weeks. When the rats were forced to exercise on a treadmill, mechanical and thermal hyperalgesia were significantly ameliorated compared with the non-exercise group. Accordingly, gene expression level of Rnf34 and Pacap were also significantly altered in the time course analysis after surgery. These results suggest that exercise therapy possibly involves pain relief in SNL rats by suppressing Rnf34 and Pacap expression in the spinal cord.
Collapse
|
34
|
Bilateral tactile hypersensitivity and neuroimmune responses after spared nerve injury in mice lacking vasoactive intestinal peptide. Exp Neurol 2017; 293:62-73. [DOI: 10.1016/j.expneurol.2017.03.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 03/21/2017] [Accepted: 03/24/2017] [Indexed: 12/30/2022]
|
35
|
Lukács M, Warfvinge K, Tajti J, Fülöp F, Toldi J, Vécsei L, Edvinsson L. Topical dura mater application of CFA induces enhanced expression of c-fos and glutamate in rat trigeminal nucleus caudalis: attenuated by KYNA derivate (SZR72). J Headache Pain 2017; 18:39. [PMID: 28337634 PMCID: PMC5364126 DOI: 10.1186/s10194-017-0746-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Accepted: 03/14/2017] [Indexed: 11/17/2022] Open
Abstract
Background Migraine is a debilitating neurological disorder where trigeminovascular activation plays a key role. We have previously reported that local application of Complete Freund’s Adjuvant (CFA) onto the dura mater caused activation in rat trigeminal ganglion (TG) which was abolished by a systemic administration of kynurenic acid (KYNA) derivate (SZR72). Here, we hypothesize that this activation may extend to the trigeminal complex in the brainstem and is attenuated by treatment with SZR72. Methods Activation in the trigeminal nucleus caudalis (TNC) and the trigeminal tract (Sp5) was achieved by application of CFA onto the dural parietal surface. SZR72 was given intraperitoneally (i.p.), one dose prior CFA deposition and repeatedly daily for 7 days. Immunohistochemical studies were performed for mapping glutamate, c-fos, PACAP, substance P, IL-6, IL-1β and TNFα in the TNC/Sp5 and other regions of the brainstem and at the C1-C2 regions of the spinal cord. Results We found that CFA increased c-fos and glutamate immunoreactivity in TNC and C1-C2 neurons. This effect was mitigated by SZR72. PACAP positive fibers were detected in the fasciculus cuneatus and gracilis. Substance P, TNFα, IL-6 and IL-1β immunopositivity were detected in fibers of Sp5 and neither of these molecules showed any change in immunoreactivity following CFA administration. Conclusion This is the first study demonstrating that dural application of CFA increases the expression of c-fos and glutamate in TNC neurons. Treatment with the KYNA analogue prevented this expression.
Collapse
Affiliation(s)
- M Lukács
- Department of Clinical Sciences, Division of Experimental Vascular Research, Lund University, Lund, Sweden. .,Department of Neurology, University of Szeged, 6725 Semmelweis street nr. 6, Szeged, Hungary.
| | - K Warfvinge
- Department of Clinical Sciences, Division of Experimental Vascular Research, Lund University, Lund, Sweden.,Department of Clinical Experimental Research, Copenhagen University, Glostrup Hospital, Copenhagen, Denmark
| | - J Tajti
- Department of Neurology, University of Szeged, 6725 Semmelweis street nr. 6, Szeged, Hungary
| | - F Fülöp
- Institute of Pharmaceutical Chemistry and MTA-SZTE Research Group for Stereochemistry, University of Szeged, Szeged, Hungary
| | - J Toldi
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, Hungary.,MTA SZTE Neuroscience Research Group, Szeged, Hungary
| | - L Vécsei
- Department of Neurology, University of Szeged, 6725 Semmelweis street nr. 6, Szeged, Hungary.,MTA SZTE Neuroscience Research Group, Szeged, Hungary
| | - L Edvinsson
- Department of Clinical Sciences, Division of Experimental Vascular Research, Lund University, Lund, Sweden.,Department of Clinical Experimental Research, Copenhagen University, Glostrup Hospital, Copenhagen, Denmark
| |
Collapse
|
36
|
Kozłowska A, Mikołajczyk A, Adamiak Z, Majewski M. Distribution and chemical coding of sensory neurons innervating the skin of the porcine hindlimb. Neuropeptides 2017; 61:1-14. [PMID: 27866657 DOI: 10.1016/j.npep.2016.10.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 10/17/2016] [Accepted: 10/24/2016] [Indexed: 11/18/2022]
Abstract
The aim of the present study was to establish the origin and chemical phenotyping of neurons involved in skin innervation of the porcine hind leg. The dorsal root ganglia (DRGs) of the lumbar (L4-L6) and sacral (S1-S3) spinal nerves were visualized using the fluorescent tracer Fast Blue (FB). The morphometric analysis of FB-positive (FB+)neurons showed that in the L4, L5, S1 and S2 DRGs, the small-sized perikarya constituted the major population, whereas in the L6 and S3 DRGs the medium-sized cells made up the major population. In all these ganglia, large-sized FB+ perikarya constituted only a small percentage of all FB+ neurons. Immunohistochemistry revealed that small- and medium-sized FB+ perikarya contained sensory markers such as: substance P (SP), calcitonin gene related peptide (CGRP) and galanin (GAL); as well as various other factors such as somatostatin (SOM), calbindin-D28k (CB), pituitary adenylate cyclase-activating polypeptide (PACAP) and neuronal nitric oxide synthase (nNOS). Meanwhile large-sized FB+ perikarya usually expressed SP, CGRP or PACAP. In the lumbar DRGs, some large cells also contained SOM and CB. Double-labeling immunohistochemistry showed that SP-positive neurons co-expressed CGRP, GAL or PACAP; while PACAP-positive cells co-expressed GAL or nNOS. Neurons stained for SOM were also immunoreactive for CB or GAL, while neurons stained for nNOS were also immunoreactive for GAL. In conclusion, the present data has indicated that the distribution and chemical phenotyping of the porcine skin-projecting neurons are different within DRGs of the lumbar (forming a femoral nerve) and sacral (forming a sciatic nerve) spinal nerves.
Collapse
Affiliation(s)
- Anna Kozłowska
- Department of Human Physiology, Faculty of Medical Sciences, University of Warmia and Mazury in Olsztyn, Poland.
| | - Anita Mikołajczyk
- Department of Public Health, Epidemiology and Microbiology, Faculty of Medical Sciences, University of Warmia and Mazury in Olsztyn, Poland
| | - Zbigniew Adamiak
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Poland
| | - Mariusz Majewski
- Department of Human Physiology, Faculty of Medical Sciences, University of Warmia and Mazury in Olsztyn, Poland
| |
Collapse
|
37
|
Uchida H, Matsumura S, Okada S, Suzuki T, Minami T, Ito S. RNA editing enzyme ADAR2 is a mediator of neuropathic pain after peripheral nerve injury. FASEB J 2017; 31:1847-1855. [PMID: 28126736 DOI: 10.1096/fj.201600950r] [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: 08/31/2016] [Accepted: 01/03/2017] [Indexed: 12/15/2022]
Abstract
Transcriptional and post-translational regulations are important in peripheral nerve injury-induced neuropathic pain, but little is known about the role of post-transcriptional modification. Our objective was to determine the possible effect of adenosine deaminase acting on RNA (ADAR) enzymes, which catalyze post-transcriptional RNA editing, in tactile allodynia, a hallmark of neuropathic pain. Seven days after L5 spinal nerve transection (SNT) in adult mice, we found an increase in ADAR2 expression and a decrease in ADAR3 expression in the injured, but not in the uninjured, dorsal root ganglions (DRGs). These changes were accompanied by elevated levels of editing at the D site of the serotonin (5-hydroxytryptamine) 2C receptor (5-HT2CR), at the I/V site of coatomer protein complex subunit α (COPA), and at the R/G site of AMPA receptor subunit GluA2 in the injured DRG. Compared to Adar2+/+/Gria2R/R littermate controls, Adar2-/-/Gria2R/R mice completely lacked the increased editing of 5-HT2CR, COPA, and GluA2 transcripts in the injured DRG and showed attenuated tactile allodynia after SNT. Furthermore, the antidepressant fluoxetine inhibited neuropathic allodynia after injury and reduced the COPA I/V site editing in the injured DRG. These findings suggest that ADAR2 is a mediator of injury-induced tactile allodynia and thus a potential therapeutic target for the treatment of neuropathic pain.-Uchida, H., Matsumura, S., Okada, S., Suzuki, T., Minami, T., Ito, S. RNA editing enzyme ADAR2 is a mediator of neuropathic pain after peripheral nerve injury.
Collapse
Affiliation(s)
- Hitoshi Uchida
- Department of Medical Chemistry, Kansai Medical University, Osaka, Japan
| | - Shinji Matsumura
- Department of Medical Chemistry, Kansai Medical University, Osaka, Japan
| | - Shunpei Okada
- Department of Chemistry and Biotechnology, Graduate School of Engineering, University of Tokyo, Tokyo, Japan; and
| | - Tsutomu Suzuki
- Department of Chemistry and Biotechnology, Graduate School of Engineering, University of Tokyo, Tokyo, Japan; and
| | - Toshiaki Minami
- Department of Anesthesiology, Osaka Medical College, Osaka, Japan
| | - Seiji Ito
- Department of Medical Chemistry, Kansai Medical University, Osaka, Japan;
| |
Collapse
|
38
|
Yokai M, Kurihara T, Miyata A. Spinal astrocytic activation contributes to both induction and maintenance of pituitary adenylate cyclase-activating polypeptide type 1 receptor-induced long-lasting mechanical allodynia in mice. Mol Pain 2016; 12:12/0/1744806916646383. [PMID: 27175011 PMCID: PMC4956379 DOI: 10.1177/1744806916646383] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 03/29/2016] [Indexed: 12/05/2022] Open
Abstract
Background Pituitary adenylate cyclase-activating polypeptide (PACAP) and its receptors are present in the spinal dorsal horn and dorsal root ganglia, suggesting an important role of PACAP–PACAP receptors signaling system in the modulation of spinal nociceptive transmission. We have previously reported that a single intrathecal injection of PACAP or a PACAP specific (PAC1) receptor selective agonist, maxadilan, in mice induced dose-dependent aversive behaviors, which lasted more than 30 min, and suggested that the maintenance of the nociceptive behaviors was associated with the spinal astrocytic activation. Results We found that a single intrathecal administration of PACAP or maxadilan also produced long-lasting hind paw mechanical allodynia, which persisted at least 84 days without affecting thermal nociceptive threshold. In contrast, intrathecal application of vasoactive intestinal polypeptide did not change mechanical threshold, and substance P, calcitonin gene-related peptide, or N-methyl-D-aspartate induced only transient mechanical allodynia, which disappeared within 21 days. Western blot and immunohistochemical analyses with an astrocytic marker, glial fibrillary acidic protein, revealed that the spinal PAC1 receptor stimulation caused sustained astrocytic activation, which also lasted more than 84 days. Intrathecal co-administration of L-α-aminoadipate, an astroglial toxin, with PACAP or maxadilan almost completely prevented the induction of the mechanical allodynia. Furthermore, intrathecal treatment of L-α-aminoadipate at 84 days after the PAC1 stimulation transiently reversed the mechanical allodynia accompanied by the reduction of glial fibrillary acidic protein expression level. Conclusion Our data suggest that spinal astrocytic activation triggered by the PAC1 receptor stimulation contributes to both induction and maintenance of the long-term mechanical allodynia.
Collapse
Affiliation(s)
- Masafumi Yokai
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima City, Japan
| | - Takashi Kurihara
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima City, Japan
| | - Atsuro Miyata
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima City, Japan
| |
Collapse
|
39
|
Ohnou T, Yokai M, Kurihara T, Hasegawa-Moriyama M, Shimizu T, Inoue K, Kambe Y, Kanmura Y, Miyata A. Pituitary adenylate cyclase-activating polypeptide type 1 receptor signaling evokes long-lasting nociceptive behaviors through the activation of spinal astrocytes in mice. J Pharmacol Sci 2016; 130:194-203. [PMID: 26948958 DOI: 10.1016/j.jphs.2016.01.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 01/21/2016] [Accepted: 01/25/2016] [Indexed: 12/14/2022] Open
Abstract
Intrathecal (i.t.) administration of pituitary adenylate cyclase-activating polypeptide (PACAP) induces long-lasting nociceptive behaviors for more than 60 min in mice, while the involvement of PACAP type1 receptor (PAC1-R) has not been clarified yet. The present study investigated signaling mechanisms of the PACAP-induced prolonged nociceptive behaviors. Single i.t. injection of a selective PAC1-R agonist, maxadilan (Max), mimicked nociceptive behaviors in a dose-dependent manner similar to PACAP. Pre- or post-treatment of a selective PAC1-R antagonist, max.d.4, significantly inhibited the nociceptive behaviors by PACAP or Max. Coadministration of a protein kinase A inhibitor, Rp-8-Br-cAMPS, a mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) kinase inhibitor, PD98059 or a c-Jun N-terminal kinase (JNK) inhibitor, SP600125, significantly inhibited the nociceptive behaviors by Max. Immunohistochemistry and immunoblotting analysis revealed that spinal administration of Max-induced ERK phosphorylation and JNK phosphorylation, and also augmented an astrocyte marker, glial fibrillary acidic protein in mouse spinal cord. Furthermore, an astroglial toxin, l-α-aminoadipate, significantly attenuated the development of the nociceptive behaviors and ERK phosphorylation by Max. These results suggest that the activation of spinal PAC1-R induces long-lasting nociception through the interaction of neurons and astrocytes.
Collapse
Affiliation(s)
- Tetsuya Ohnou
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima City, Kagoshima 890-8544, Japan; Department of Anesthesiology and Critical Care Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima City, Kagoshima 890-8544, Japan
| | - Masafumi Yokai
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima City, Kagoshima 890-8544, Japan
| | - Takashi Kurihara
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima City, Kagoshima 890-8544, Japan
| | - Maiko Hasegawa-Moriyama
- Department of Anesthesiology and Critical Care Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima City, Kagoshima 890-8544, Japan
| | - Takao Shimizu
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima City, Kagoshima 890-8544, Japan
| | - Kazuhiko Inoue
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima City, Kagoshima 890-8544, Japan
| | - Yuki Kambe
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima City, Kagoshima 890-8544, Japan
| | - Yuichi Kanmura
- Department of Anesthesiology and Critical Care Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima City, Kagoshima 890-8544, Japan
| | - Atsuro Miyata
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima City, Kagoshima 890-8544, Japan.
| |
Collapse
|
40
|
Shudo Y, Shimojo M, Fukunaga M, Ito S. Pituitary adenylate cyclase-activating polypeptide is regulated by alternative splicing of transcriptional repressor REST/NRSF in nerve injury. Life Sci 2015; 143:174-81. [PMID: 26518165 DOI: 10.1016/j.lfs.2015.10.033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 09/28/2015] [Accepted: 10/26/2015] [Indexed: 12/22/2022]
Abstract
AIMS The pathophysiological mechanism for neuropathic pain (NP), one of the most common types of intractable pain, remains largely unknown. We previously reported that pituitary adenylate-cyclase activating polypeptide (PACAP) is required for the development of spinal sensitization and induction of NP. Previous in vitro studies suggest that PACAP transcription unit has two RE1-like elements and that the transcriptional repressor REST controls expression of PACAP gene. However the regulation of PACAP gene through its RE1 sites in vivo has not been studied. We have analyzed the functional role of PACAP gene RE1 element following nerve injury. MAIN METHODS An L5-spinal nerve transection (L5-SNT) in mice was used as a model of spinal injury. DRGs after the L5-SNT were studied. KEY FINDINGS PACAP mRNA increased in the DRG following spinal nerve injury. REST4, an alternatively spliced isoform of REST was shown to be regulated by the splicing activator (nSR100) and nSR100 itself also increased. Overexpression of either REST4 or nSR100 in vitro increased PACAP expression, while overexpression of REST repressed PACAP mRNA production. Reporter gene analysis showed that a novel RE1 previously predicted by in silico analysis was indeed functional. ChIP analysis showed that REST bound significantly to this RE1 in the DRG of naïve mice, while REST binding to this RE1 was decreased following spinal nerve injury. The expression of REST was decreased by nSR100-dependent alternative splicing of the REST gene, leading to derepression of PACAP. SIGNIFICANCE PACAP expression in the DRG following spinal nerve injury is controlled through a novel RE1 by REST.
Collapse
Affiliation(s)
- Yoshie Shudo
- Department of Medical Chemistry, Kansai Medical University, Osaka, Japan; Department of Psychosomatic Medicine, Kansai Medical University, Osaka, Japan
| | - Masahito Shimojo
- Department of Medical Chemistry, Kansai Medical University, Osaka, Japan.
| | - Mikihiko Fukunaga
- Department of Psychosomatic Medicine, Kansai Medical University, Osaka, Japan
| | - Seiji Ito
- Department of Medical Chemistry, Kansai Medical University, Osaka, Japan
| |
Collapse
|
41
|
Gupte RP, Kadunganattil S, Shepherd AJ, Merrill R, Planer W, Bruchas MR, Strack S, Mohapatra DP. Convergent phosphomodulation of the major neuronal dendritic potassium channel Kv4.2 by pituitary adenylate cyclase-activating polypeptide. Neuropharmacology 2015; 101:291-308. [PMID: 26456351 DOI: 10.1016/j.neuropharm.2015.10.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 09/29/2015] [Accepted: 10/03/2015] [Indexed: 12/30/2022]
Abstract
The endogenous neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) is secreted by both neuronal and non-neuronal cells in the brain and spinal cord, in response to pathological conditions such as stroke, seizures, chronic inflammatory and neuropathic pain. PACAP has been shown to exert various neuromodulatory and neuroprotective effects. However, direct influence of PACAP on the function of intrinsically excitable ion channels that are critical to both hyperexcitation as well as cell death, remain largely unexplored. The major dendritic K(+) channel Kv4.2 is a critical regulator of neuronal excitability, back-propagating action potentials in the dendrites, and modulation of synaptic inputs. We identified, cloned and characterized the downstream signaling originating from the activation of three PACAP receptor (PAC1) isoforms that are expressed in rodent hippocampal neurons that also exhibit abundant expression of Kv4.2 protein. Activation of PAC1 by PACAP leads to phosphorylation of Kv4.2 and downregulation of channel currents, which can be attenuated by inhibition of either PKA or ERK1/2 activity. Mechanistically, this dynamic downregulation of Kv4.2 function is a consequence of reduction in the density of surface channels, without any influence on the voltage-dependence of channel activation. Interestingly, PKA-induced effects on Kv4.2 were mediated by ERK1/2 phosphorylation of the channel at two critical residues, but not by direct channel phosphorylation by PKA, suggesting a convergent phosphomodulatory signaling cascade. Altogether, our findings suggest a novel GPCR-channel signaling crosstalk between PACAP/PAC1 and Kv4.2 channel in a manner that could lead to neuronal hyperexcitability.
Collapse
Affiliation(s)
- Raeesa P Gupte
- Department of Pharmacology, The University of Iowa Roy J. and Lucile A. Carver College of Medicine, Iowa City, IA 52242, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Washington University Pain Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Suraj Kadunganattil
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Washington University Pain Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Andrew J Shepherd
- Department of Pharmacology, The University of Iowa Roy J. and Lucile A. Carver College of Medicine, Iowa City, IA 52242, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Washington University Pain Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ronald Merrill
- Department of Pharmacology, The University of Iowa Roy J. and Lucile A. Carver College of Medicine, Iowa City, IA 52242, USA
| | - William Planer
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael R Bruchas
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Stefan Strack
- Department of Pharmacology, The University of Iowa Roy J. and Lucile A. Carver College of Medicine, Iowa City, IA 52242, USA
| | - Durga P Mohapatra
- Department of Pharmacology, The University of Iowa Roy J. and Lucile A. Carver College of Medicine, Iowa City, IA 52242, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Washington University Pain Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
42
|
RGS9-2--controlled adaptations in the striatum determine the onset of action and efficacy of antidepressants in neuropathic pain states. Proc Natl Acad Sci U S A 2015; 112:E5088-97. [PMID: 26305935 DOI: 10.1073/pnas.1504283112] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The striatal protein Regulator of G-protein signaling 9-2 (RGS9-2) plays a key modulatory role in opioid, monoamine, and other G-protein-coupled receptor responses. Here, we use the murine spared-nerve injury model of neuropathic pain to investigate the mechanism by which RGS9-2 in the nucleus accumbens (NAc), a brain region involved in mood, reward, and motivation, modulates the actions of tricyclic antidepressants (TCAs). Prevention of RGS9-2 action in the NAc increases the efficacy of the TCA desipramine and dramatically accelerates its onset of action. By controlling the activation of effector molecules by G protein α and βγ subunits, RGS9-2 affects several protein interactions, phosphoprotein levels, and the function of the epigenetic modifier histone deacetylase 5, which are important for TCA responsiveness. Furthermore, information from RNA-sequencing analysis reveals that RGS9-2 in the NAc affects the expression of many genes known to be involved in nociception, analgesia, and antidepressant drug actions. Our findings provide novel information on NAc-specific cellular mechanisms that mediate the actions of TCAs in neuropathic pain states.
Collapse
|
43
|
Yamasaki A, Kasai A, Toi A, Kurita M, Kimoto S, Hayata-Takano A, Nakazawa T, Nagayasu K, Shintani N, Hashimoto R, Ito A, Meltzer HY, Ago Y, Waschek JA, Onaka Y, Matsuda T, Baba A, Hashimoto H. Identification of the role of bone morphogenetic protein (BMP) and transforming growth factor-β (TGF-β) signaling in the trajectory of serotonergic differentiation in a rapid assay in mouse embryonic stem cells in vitro. J Neurochem 2015; 132:418-28. [PMID: 25421849 DOI: 10.1111/jnc.12999] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 10/18/2014] [Accepted: 11/15/2014] [Indexed: 12/25/2022]
Abstract
The mechanism by which extracellular molecules control serotonergic cell fate remains elusive. Recently, we showed that noggin, which inactivates bone morphogenetic proteins (BMPs), induces serotonergic differentiation of mouse embryonic (ES) and induced pluripotent stem cells with coordinated gene expression along the serotonergic lineage. Here, we created a rapid assay for serotonergic induction by generating knock-in ES cells expressing a naturally secreted Gaussia luciferase driven by the enhancer of Pet-1/Fev, a landmark of serotonergic differentiation. Using these cells, we performed candidate-based screening and identified BMP type I receptor kinase inhibitors LDN-193189 and DMH1 as activators of luciferase. LDN-193189 induced ES cells to express the genes encoding Pet-1, tryptophan hydroxylase 2, and the serotonin transporter, and increased serotonin release without altering dopamine release. In contrast, TGF-β receptor inhibitor SB-431542 selectively inhibited serotonergic differentiation, without changing overall neuronal differentiation. LDN-193189 inhibited expression of the BMP signaling target gene Id, and induced the TGF-β target gene Lefty, whereas the opposite effect was observed with SB-431542. This study thus provides a new tool to investigate serotonergic differentiation and suggests that inhibition of BMP type I receptors and concomitant activation of TGF-β receptor signaling are implicated in serotonergic differentiation. Candidate-based screening for serotonergic induction using a rapid assay in mouse embryonic stem cells revealed that the bone morphogenetic protein (BMP) type I receptor kinase inhibitors selectively induce serotonergic differentiation, whereas the TGF-β receptor inhibitor SB-431542 inhibits the differentiation. These results suggest that inhibition of BMP type I receptors and concomitant activation of transforming growth factor-β (TGF-β) receptor signaling are involved in the early trajectory of serotonergic differentiation.
Collapse
Affiliation(s)
- Atsushi Yamasaki
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Botz B, Bölcskei K, Kereskai L, Kovács M, Németh T, Szigeti K, Horváth I, Máthé D, Kovács N, Hashimoto H, Reglődi D, Szolcsányi J, Pintér E, Mócsai A, Helyes Z. Differential regulatory role of pituitary adenylate cyclase-activating polypeptide in the serum-transfer arthritis model. Arthritis Rheumatol 2014; 66:2739-50. [PMID: 25048575 PMCID: PMC4320777 DOI: 10.1002/art.38772] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 07/01/2014] [Indexed: 01/07/2023]
Abstract
Objective Pituitary adenylate cyclase–activating polypeptide (PACAP) expressed in capsaicin-sensitive sensory neurons and immune cells has divergent functions in inflammatory and pain processes. This study was undertaken to investigate the involvement of PACAP in a mouse model of rheumatoid arthritis. Methods Arthritis was induced in PACAP−/− and wild-type (PACAP+/+) mice by K/BxN serum transfer. General features of the disease were investigated by semiquantitative scoring, plethysmometry, and histopathologic analysis. Mechano- and thermonociceptive thresholds and motor functions were also evaluated. Metabolic activity was assessed by positron emission tomography. Bone morphology was measured by in vivo micro–computed tomography, myeloperoxidase activity and superoxide production by bioluminescence imaging with luminol and lucigenin, respectively, and vascular permeability by fluorescent indocyanine green dye study. Results PACAP+/+ mice developed notable joint swelling, reduced grasping ability, and mechanical (but not thermal) hyperalgesia after K/BxN serum transfer. In PACAP−/− mice clinical scores and edema were significantly reduced, and mechanical hyperalgesia and motor impairment were absent, throughout the 2-week period of observation. Metabolic activity and superoxide production increased in the tibiotarsal joints of wild-type mice but were significantly lower in PACAP−/− animals. Myeloperoxidase activity in the ankle joints of PACAP−/− mice was significantly reduced in the early phase of arthritis, but increased in the late phase. Synovial hyperplasia was also significantly increased, and progressive bone spur formation was observed in PACAP-deficient mice only. Conclusion In PACAP-deficient mice with serum-transfer arthritis, joint swelling, vascular leakage, hyperalgesia, and early inflammatory cell accumulation are reduced; in the later phase of the disease, immune cell function and bone neoformation are increased. Elucidation of the underlying pathways of PACAP activity may open promising new avenues for development of therapy in inflammatory arthritis.
Collapse
Affiliation(s)
- Bálint Botz
- Department of Pharmacology and Pharmacotherapy, University of Pécs Medical School, and Molecular Pharmacology Research Team, János Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Shintani N, Onaka Y, Hashimoto R, Takamura H, Nagata T, Umeda-Yano S, Mouri A, Mamiya T, Haba R, Matsuzaki S, Katayama T, Yamamori H, Nakazawa T, Nagayasu K, Ago Y, Yagasaki Y, Nabeshima T, Takeda M, Hashimoto H. Behavioral characterization of mice overexpressing human dysbindin-1. Mol Brain 2014; 7:74. [PMID: 25298178 PMCID: PMC4201722 DOI: 10.1186/s13041-014-0074-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 09/25/2014] [Indexed: 11/30/2022] Open
Abstract
Background The dysbindin-1 gene (DTNBP1: dystrobrevin binding protein 1) is a promising schizophrenia susceptibility gene, known to localize almost exclusively to neurons in the brain, and participates in the regulation of neurotransmitter release, membrane-surface receptor expression, and synaptic plasticity. Sandy mice, with spontaneous Dtnbp1 deletion, display behavioral abnormalities relevant to symptoms of schizophrenia. However, it remains unknown if dysbindin-1 gain-of-function is beneficial or detrimental. Results To answer this question and gain further insight into the pathophysiology and therapeutic potential of dysbindin-1, we developed transgenic mice expressing human DTNBP1 (Dys1A-Tg) and analyzed their behavioral phenotypes. Dys1A-Tg mice were born viable in the expected Mendelian ratios, apparently normal and fertile. Primary screening of behavior and function showed a marginal change in limb grasping in Dys1A-Tg mice. In addition, Dys1A-Tg mice exhibited increased hyperlocomotion after methamphetamine injection. Transcriptomic analysis identified several up- and down-regulated genes, including the immediate-early genes Arc and Egr2, in the prefrontal cortex of Dys1A-Tg mice. Conclusions The present findings in Dys1A-Tg mice support the role of dysbindin-1 in psychiatric disorders. The fact that either overexpression (Dys1A-Tg) or underexpression (Sandy) of dysbindin-1 leads to behavioral alterations in mice highlights the functional importance of dysbindin-1 in vivo.
Collapse
Affiliation(s)
- Norihito Shintani
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Yusuke Onaka
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Ryota Hashimoto
- Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan. .,Department of Psychiatry, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Hironori Takamura
- Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Tsuyoshi Nagata
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Satomi Umeda-Yano
- Department of Molecular Neuropsychiatry, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Akihiro Mouri
- Department of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya, 468-8503, Japan.
| | - Takayoshi Mamiya
- Department of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya, 468-8503, Japan.
| | - Ryota Haba
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Shinsuke Matsuzaki
- Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan. .,Molecular Brain Science, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan. .,Anatomy and Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Taiichi Katayama
- Molecular Brain Science, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Hidenaga Yamamori
- Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan. .,Department of Molecular Neuropsychiatry, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Takanobu Nakazawa
- iPS Cell-based Research Project on Brain Neuropharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Kazuki Nagayasu
- iPS Cell-based Research Project on Brain Neuropharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Yukio Ago
- Laboratory of Medicinal Pharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Yuki Yagasaki
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashicho, Kodaira, Tokyo, 187-8502, Japan.
| | - Toshitaka Nabeshima
- Department of Regional Pharmaceutical Care & Sciences, Graduate School of Pharmaceutical Sciences, Meijo University, 150 Yagotoyama, Tenpaku-ku, Nagoya, 468-8503, Japan.
| | - Masatoshi Takeda
- Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan. .,Department of Psychiatry, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Hitoshi Hashimoto
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan. .,Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan. .,iPS Cell-based Research Project on Brain Neuropharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
46
|
Parabrachial nucleus (PBn) pituitary adenylate cyclase activating polypeptide (PACAP) signaling in the amygdala: implication for the sensory and behavioral effects of pain. Neuropharmacology 2014; 86:38-48. [PMID: 24998751 DOI: 10.1016/j.neuropharm.2014.06.022] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 06/17/2014] [Accepted: 06/19/2014] [Indexed: 11/22/2022]
Abstract
The intricate relationships that associate pain, stress responses and emotional behavior have been well established. Acute stressful situations can decrease nociceptive sensations and conversely, chronic pain can enhance other pain experiences and heighten the emotional and behavioral consequences of stress. Accordingly, chronic pain is comorbid with a number of behavioral disorders including depression, anxiety abnormalities and associated stress-related disorders including post traumatic stress disorder (PTSD). The central nucleus of the amygdala (CeA) represents a convergence of pathways for pain, stress and emotion, and we have identified pituitary adenylate cyclase activating polypeptide (PACAP) immunoreactivity in fiber elements in the lateral capsular division of the CeA (CeLC). The PACAP staining patterns colocalized in part with those for calcitonin gene related peptide (CGRP); anterograde fiber tracing and excitotoxic lesion studies demonstrated that the CeLC PACAP/CGRP immunoreactivities represented sensory fiber projections from the lateral parabrachial nucleus (LPBn) along the spino-parabrachioamygdaloid tract. The same PBn PACAP/CGRP fiber system also projected to the BNST. As in the BNST, CeA PACAP signaling increased anxiety-like behaviors accompanied by weight loss and decreased feeding. But in addition to heightened anxiety-like responses, CeA PACAP signaling also altered nociception as reflected by decreased latency and threshold responses in thermal and mechanical sensitivity tests, respectively. From PACAP expression in major pain pathways, the current observations are novel and suggest that CeA PACAP nociceptive signaling and resulting neuroplasticity via the spino-parabrachioamygdaloid tract may represent mechanisms that associate chronic pain with sensory hypersensitivity, fear memory consolidation and severe behavioral disorders.
Collapse
|
47
|
Pettersson LME, Geremia NM, Ying Z, Verge VMK. Injury-associated PACAP expression in rat sensory and motor neurons is induced by endogenous BDNF. PLoS One 2014; 9:e100730. [PMID: 24968020 PMCID: PMC4072603 DOI: 10.1371/journal.pone.0100730] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 05/28/2014] [Indexed: 11/19/2022] Open
Abstract
Peripheral nerve injury results in dramatic upregulation in pituitary adenylate cyclase activating polypeptide (PACAP) expression in adult rat dorsal root ganglia and spinal motor neurons mirroring that described for the neurotrophin brain derived neurotrophic factor (BDNF). Thus, we posited that injury-associated alterations in BDNF expression regulate the changes in PACAP expression observed in the injured neurons. The role of endogenous BDNF in induction and/or maintenance of PACAP mRNA expression in injured adult rat motor and sensory neurons was examined by intrathecally infusing or intraperitoneally injecting BDNF-specific antibodies or control IgGs immediately at the time of L4-L6 spinal nerve injury, or in a delayed fashion one week later for 3 days followed by analysis of impact on PACAP expression. PACAP mRNA in injured lumbar sensory and motor neurons was detected using in situ hybridization, allowing quantification of relative changes between experimental groups, with ATF-3 immunofluorescence serving to identify the injured subpopulation of motor neurons. Both the incidence and level of PACAP mRNA expression were dramatically reduced in injured sensory and motor neurons in response to immediate intrathecal anti-BDNF treatment. In contrast, neither intraperitoneal injections nor delayed intrathecal infusions of anti-BDNF had any discernible impact on PACAP expression. This impact on PACAP expression in response to BDNF immunoneutralization in DRG was confirmed using qRT-PCR or by using BDNF selective siRNAs to reduce neuronal BDNF expression. Collectively, our findings support that endogenous injury-associated BDNF expression is critically involved in induction, but not maintenance, of injury-associated PACAP expression in sensory and motor neurons.
Collapse
Affiliation(s)
- Lina M. E. Pettersson
- CMSNRC & Department of Anatomy & Cell Biology, University of Saskatchewan, Saskatoon, SK, Canada
- * E-mail:
| | - Nicole M. Geremia
- CMSNRC & Department of Anatomy & Cell Biology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Zhengxin Ying
- CMSNRC & Department of Anatomy & Cell Biology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Valerie M. K. Verge
- CMSNRC & Department of Anatomy & Cell Biology, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
48
|
Malafoglia V, Colasanti M, Raffaeli W, Balciunas D, Giordano A, Bellipanni G. Extreme thermal noxious stimuli induce pain responses in zebrafish larvae. J Cell Physiol 2014; 229:300-8. [PMID: 23929528 DOI: 10.1002/jcp.24447] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 08/01/2013] [Indexed: 11/06/2022]
Abstract
Exposing tissues to extreme high or low temperature leads to burns. Burned animals sustain several types of damage, from the disruption of the tissue to degeneration of axons projecting through muscle and skin. Such damage causes pain due to both inflammation and axonal degeneration (neuropathic-like pain). Thus, the approach to cure and alleviate the symptoms of burns must be twofold: rebuilding the tissue that has been destroyed and alleviating the pain derived from the burns. While tissue regeneration techniques have been developed, less is known on the treatment of the induced pain. Thus, appropriate animal models are necessary for the development of the best treatment for pain induced in burned tissues. We have developed a methodology in the zebrafish aimed to produce a new animal model for the study of pain induced by burns. Here, we show that two events linked to the onset of burn-induced inflammation and neuropathic-like pain in mammals, degeneration of axons innervating the affected tissues and over-expression of specific genes in sensory tissues, are conserved from zebrafish to mammals.
Collapse
Affiliation(s)
- Valentina Malafoglia
- Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, Pennsylvania; ISAL-Foundation, Institute for Research on Pain, Torre Pedrera (RN), Italy
| | | | | | | | | | | |
Collapse
|
49
|
Botz B, Imreh A, Sándor K, Elekes K, Szolcsányi J, Reglődi D, Quinn JP, Stewart J, Zimmer A, Hashimoto H, Helyes Z. Role of Pituitary Adenylate-Cyclase Activating Polypeptide and Tac1 gene derived tachykinins in sensory, motor and vascular functions under normal and neuropathic conditions. Peptides 2013; 43:105-12. [PMID: 23499760 DOI: 10.1016/j.peptides.2013.03.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 03/04/2013] [Accepted: 03/04/2013] [Indexed: 12/15/2022]
Abstract
Pituitary Adenylate-Cyclase Activating Polypeptide (PACAP) and Tac1 gene-encoded tachykinins (substance P: SP, neurokinin A: NKA) are expressed in capsaicin-sensitive nerves, but their role in nociception, inflammation and vasoregulation is unclear. Therefore, we investigated the function of these neuropeptides and the NK1 tachykinin receptor (from Tacr1 gene) in the partial sciatic nerve ligation-induced traumatic mononeuropathy model using gene deficient (PACAP(-/-), Tac1(-/-), and Tacr1(-/-)) mice. Mechanonociceptive threshold of the paw was measured with dynamic plantar aesthesiometry, motor coordination with Rota-Rod and cutaneous microcirculation with laser Doppler imaging. Neurogenic vasodilation was evoked by mustard oil stimulating sensory nerves. In wildtype mice 30-40% mechanical hyperalgesia developed one week after nerve ligation, which was not altered in Tac1(-/-) and Tacr1(-/-) mice, but was absent in PACAP(-/-) animals. Motor coordination of the PACAP(-/-) and Tac1(-/-) groups was significantly worse both before and after nerve ligation compared to their wildtypes, but it did not change in Tacr1(-/-) mice. Basal postoperative microcirculation on the plantar skin of PACAP(-/-) mice did not differ from the wildtypes, but was significantly lower in Tac1(-/-) and Tacr1(-/-) ones. In contrast, mustard oil-induced neurogenic vasodilation was significantly smaller in PACAP(-/-) mice, but not in Tacr1(-/-) and Tac1(-/-) animals. Both PACAP and SP/NKA, but not NK1 receptors participate in normal motor coordination. Tachykinins maintain basal cutaneous microcirculation. PACAP is a crucial mediator of neuropathic mechanical hyperalgesia and neurogenic vasodilation. Therefore identifying its target and developing selective, potent antagonists, might open promising new perspectives for the treatment of neuropathic pain and vascular complications.
Collapse
Affiliation(s)
- Bálint Botz
- Department of Pharmacology and Pharmacotherapy, University of Pécs, H-7624 Pécs, Szigeti u. 12, Hungary
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Tuka B, Helyes Z, Markovics A, Bagoly T, Szolcsányi J, Szabó N, Tóth E, Kincses ZT, Vécsei L, Tajti J. Alterations in PACAP-38-like immunoreactivity in the plasma during ictal and interictal periods of migraine patients. Cephalalgia 2013; 33:1085-95. [PMID: 23598374 DOI: 10.1177/0333102413483931] [Citation(s) in RCA: 160] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND Recent studies on migraineurs and our own animal experiments have revealed that pituitary adenylate cyclase-activating polypeptide-38 (PACAP-38) has an important role in activation of the trigeminovascular system. The aim of this study was to determine the PACAP-38-like immunoreactivity (LI) in the plasma of healthy subjects, and parallel with the calcitonin gene-related peptide (CGRP)-LI in migraine patients in the ictal and interictal periods. METHODS A total of 87 migraineurs and 40 healthy control volunteers were enrolled in the examination. Blood samples were collected from the cubital veins in both periods in 21 patients, and in either the ictal or the interictal period in the remaining 66 patients, and were analysed by radioimmunoassay. RESULTS A significantly lower PACAP-38-LI was measured in the interictal plasma of the migraineurs as compared with the healthy control group ( P < 0.011). In contrast, elevated peptide levels were detected in the ictal period relative to the attack-free period in the 21 migraineurs ( P PACAP-38 < 0.001; P CGRP < 0.035) and PACAP-38-LI in the overall population of migraineurs ( P < 0.009). A negative correlation was observed between the interictal PACAP-38-LI and the disease duration. CONCLUSION This is the first study that has provided evidence of a clear association between migraine phases (ictal and interictal) and plasma PACAP-38-LI alterations.
Collapse
Affiliation(s)
- Bernadett Tuka
- Department of Neurology, Faculty of Medicine, University of Szeged, Hungary
| | | | | | | | | | | | | | | | | | | |
Collapse
|