1
|
Tasma Z, Garelja ML, Jamaluddin A, Alexander TI, Rees TA. Where are we now? Biased signalling of Class B G protein-coupled receptor-targeted therapeutics. Pharmacol Ther 2025; 270:108846. [PMID: 40216261 DOI: 10.1016/j.pharmthera.2025.108846] [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: 10/31/2024] [Revised: 02/07/2025] [Accepted: 03/24/2025] [Indexed: 04/19/2025]
Abstract
Class B G protein-coupled receptors (GPCRs) are a subfamily of 15 peptide hormone receptors with diverse roles in physiological functions and disease pathogenesis. Over the past decade, several novel therapeutics targeting these receptors have been approved for conditions like migraine, diabetes, and obesity, many of which are ground-breaking and first-in-class. Most of these therapeutics are agonist analogues with modified endogenous peptide sequences to enhance receptor activation or stability. Several small molecule and monoclonal antibody antagonists have also been approved or are in late-stage development. Differences in the sequence and structure of these therapeutic ligands lead to distinct signalling profiles, including biased behaviour or inhibition of specific pathways. Understanding this biased pharmacology offers unique development opportunities for improving therapeutic efficacy and reducing adverse effects. This review summarises current knowledge on the ligand bias of approved class B GPCR drugs, highlights strategies to refine and exploit their pharmacological profiles, and discusses key considerations related to receptor structure, localisation, and regulation for developing new therapies.
Collapse
Affiliation(s)
- Zoe Tasma
- Department of Pharmacology and Toxicology, University of Otago, Dunedin 9016, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland 1010, New Zealand
| | - Michael L Garelja
- Department of Pharmacology and Toxicology, University of Otago, Dunedin 9016, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland 1010, New Zealand
| | - Aqfan Jamaluddin
- Department of Metabolism and Systems Science, College of Medicine and Health, University of Birmingham, Birmingham, UK; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, UK
| | - Tyla I Alexander
- Department of Pharmacology and Toxicology, University of Otago, Dunedin 9016, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland 1010, New Zealand
| | - Tayla A Rees
- Headache Group, Wolfson Sensory Pain and Regeneration Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| |
Collapse
|
2
|
Karsan N, Marzoughi S, Goadsby PJ. Advances in understanding migraine for the development of novel pharmacotherapies: the use of human provocation migraine models. Expert Opin Pharmacother 2025. [PMID: 40353527 DOI: 10.1080/14656566.2025.2505231] [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: 03/12/2025] [Revised: 05/01/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
INTRODUCTION Whilst migraine treatment has advanced significantly over recent times, the mechanisms of attack genesis and heterogeneity in treatment response are two amidst several areas that remain poorly understood and require further development. Experimental migraine provocation is an area that holds promise in advancing this understanding. AREAS COVERED We conducted a literature search using PubMed, of 'human migraine triggering' and 'human migraine provocation' to identify articles of interest. We discuss therapeutic targets that have emerged from such work, including calcitonin family peptides (amylin (AMY) and adrenomedullin (ADM)), pituitary adenylate cyclase-activating peptide (PACAP) and potassium channels. We discuss our views on the clinical translation of the outcomes of such studies, and their previous and potential future impact on migraine therapeutics. EXPERT OPINION Migraine provocation models provide a valuable means to study human migraine phenotypically and biologically, as well as to assess treatment response. Downstream intracellular mechanisms of provocation agents can be targeted during cellular processing to alter cell function and influence migraine mechanisms. It is important to caveat the clinical translation of provocation studies, given that just because a substance triggers migraine experimentally, does not necessarily mean that the substance is involved in the spontaneous human condition.
Collapse
Affiliation(s)
- Nazia Karsan
- Headache Group, Wolfson Sensory, Pain and Regeneration Centre (SPaRC), Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
- NIHR King's Clinical Research Facility, NIHR Maudsley Biochemical Research Centre, King's College Hospital, UK
| | - Sina Marzoughi
- Headache Group, Wolfson Sensory, Pain and Regeneration Centre (SPaRC), Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
- NIHR King's Clinical Research Facility, NIHR Maudsley Biochemical Research Centre, King's College Hospital, UK
| | - Peter J Goadsby
- Headache Group, Wolfson Sensory, Pain and Regeneration Centre (SPaRC), Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
- NIHR King's Clinical Research Facility, NIHR Maudsley Biochemical Research Centre, King's College Hospital, UK
- Department of Neurology, University of California, Los Angeles, USA
| |
Collapse
|
3
|
Salinas-Abarca AB, Gamal-Eltrabily M, Romero-Reyes M, Akerman S. The role and interaction of hypothalamic-related neurotransmitters in migraine. J Headache Pain 2025; 26:110. [PMID: 40350428 PMCID: PMC12067729 DOI: 10.1186/s10194-025-02044-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/22/2025] [Indexed: 05/14/2025] Open
Abstract
Migraine is a complex neurological disorder frequently associated with hypothalamic dysfunction. This brain region is essential for maintaining homeostasis due to its regulation of autonomic, endocrine, and circadian systems. While the pathophysiology of migraine remains incompletely understood, clinical features such as the cyclic nature of attacks and symptoms, including nausea, vomiting, yawning, irritability, and sensitivity to light, indicate a significant role for the hypothalamus. Further, potential triggers of migraine, such as stress and disruption to feeding habits, also impact hypothalamic mechanisms. The higher prevalence of migraine in women compared to men suggests a link to hormonal fluctuations involving estrogen, progesterone, and prolactin. These hormones interact with the hypothalamus, potentially influencing the onset and severity of migraine episodes. Additionally, the hypothalamus synthesizes neuropeptides such as orexins, neuropeptide Y, PACAP, oxytocin, and vasopressin, which are all implicated in migraine mechanisms. Understanding the interplay between the hypothalamus, sex hormones, and neuropeptides offers valuable opportunities for endogenous targeted migraine therapies. In this review we discuss hypothalamic contributions to migraine pathophysiology, highlighting the mechanisms affected by hypothalamic connections, neuropeptides, and hormones, and their role as migraine triggers, particularly focusing on factors like stress, fasting, and changes in sleep patterns.
Collapse
Affiliation(s)
- Ana Belen Salinas-Abarca
- Department of Neural and Pain Sciences, University of Maryland Baltimore, Baltimore, Maryland, MD, 21201, USA
- Center to Advance Chronic Pain Research, University of Maryland Baltimore, Baltimore, Maryland, MD, 21201, USA
| | - Mohammed Gamal-Eltrabily
- Department of Neural and Pain Sciences, University of Maryland Baltimore, Baltimore, Maryland, MD, 21201, USA
- Center to Advance Chronic Pain Research, University of Maryland Baltimore, Baltimore, Maryland, MD, 21201, USA
| | - Marcela Romero-Reyes
- Department of Neural and Pain Sciences, University of Maryland Baltimore, Baltimore, Maryland, MD, 21201, USA
- Center to Advance Chronic Pain Research, University of Maryland Baltimore, Baltimore, Maryland, MD, 21201, USA
| | - Simon Akerman
- Department of Neural and Pain Sciences, University of Maryland Baltimore, Baltimore, Maryland, MD, 21201, USA.
- Center to Advance Chronic Pain Research, University of Maryland Baltimore, Baltimore, Maryland, MD, 21201, USA.
| |
Collapse
|
4
|
Firdos F, Pramanik T, Mittal A. Temporal Effects of a Viral Peptide on Glucose-Stimulated Insulin Secretion. ACS Infect Dis 2025; 11:1167-1179. [PMID: 40272277 DOI: 10.1021/acsinfecdis.4c01025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
Impaired glucose homeostasis, often attributed to the destruction of insulin-secreting pancreatic β-cells, results in Type 1 diabetes (T1D). While both genetic and environmental factors have been implicated in the onset of T1D, there is an increase in clinical observations of T1D resulting from or triggered by viral infections. Despite the increase mentioned above, mechanistic insights into viral-induced T1D onset remain scarce. The key question of how viral infections may impact insulin secretion, thereby affecting glucose-stimulated insulin secretion (GSIS), is still unanswered. In this work, we correlate GSIS and viral peptides for the first time. Using the Mouse Insulinoma 6 (MIN6) cell line as a model system for β cells, we studied pituitary adenylate cyclase-activating polypeptide (PACAP) potentiated GSIS. Specifically, we noted that two forms of PACAP, namely, PACAP38 (P38) and PACAP27 (P27) are important in GSIS. From proteomes of chikungunya and dengue viruses (CHIKV and DV, respectively) implicated in triggering T1D, we identified viral peptides that were similar (but not identical) to PACAP. We report that a peptide from CHIKV, termed as CV1, enhances insulin secretion in MIN6 cells along with P38; however, independent treatments with CV1 yield variable outcomes based on duration of incubation. While short-term treatment shows viral peptides outcompeting P38, leading to increased insulin secretion without cytotoxicity, there is a decrease in cell viability with longer incubations under some conditions. Remarkably, long-term CV1 incubation significantly reduces glucose sensitivity of and GSIS by MIN6 cells. We report the first experimental evidence linking viral peptides to GSIS (specifically PACAP-stimulated).
Collapse
Affiliation(s)
- Firdos Firdos
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi (IIT Delhi), Hauz Khas, New Delhi 110016, India
| | - Tapabrata Pramanik
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi (IIT Delhi), Hauz Khas, New Delhi 110016, India
| | - Aditya Mittal
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi (IIT Delhi), Hauz Khas, New Delhi 110016, India
| |
Collapse
|
5
|
Sammari H, Abidi A, Jedidi S, Dhawefi N, Sebai H. Laxative Effect of Crataegus azarolus Leaves Decoction Extract Against Loperamide-Induced Constipation and Oxidative Stress in Rats. J Med Food 2025; 28:465-477. [PMID: 40107769 DOI: 10.1089/jmf.2023.0255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025] Open
Abstract
Constipation represents a common gastrointestinal disorder that has various adverse effects on the gastrointestinal tract. As a result, various civilizations have developed phytomedicines in order to treat and relieve its symptoms. In the current study, we evaluated the effect of Crataegus azarolus L. leaves decoction extract (CALDE) against loperamide (LOP)-induced constipation in rats. For 3 days, treated rats were administered LOP (3 mg/kg, b.w., p.o.) and CALDE (50, 100, and 200 mg/kg, b.w., p.o.) or yohimbine (2 mg/kg, b.w., p.o.). The gastric emptying test or intestinal transit time was calculated. The oxidative status was studied and evaluated using biochemical colorimetric methods. Results showed that CALDE administration improves gastric emptying and accelerates gastrointestinal transit. Pretreatment with LOP altered the defecation parameters and generated an oxidative status in healthy rats. In contrast, CALDE coadministration protected against the deregulation of intestinal motor function and frequency of defecation and significantly re-established oxidative marker levels. CALDE treatment demonstrated significant protection against experimental oxidative stress and constipation induced by LOP. Therefore, it can be considered a pharmacological drug to treat these gastrointestinal troubles.
Collapse
Affiliation(s)
- Houcem Sammari
- Laboratory of Functional Physiology and Valorization, Higher Institute of Biotechnology of Béja, University of Jendouba, Béja, Tunisia
| | - Anouar Abidi
- Laboratory of Functional Physiology and Valorization, Higher Institute of Biotechnology of Béja, University of Jendouba, Béja, Tunisia
| | - Saber Jedidi
- Laboratory of Functional Physiology and Valorization, Higher Institute of Biotechnology of Béja, University of Jendouba, Béja, Tunisia
- National Institute of Technologies and Sciences of Kef (INTeK), University of Jendouba, Kef, Tunisia
| | - Nourhène Dhawefi
- Laboratory of Functional Physiology and Valorization, Higher Institute of Biotechnology of Béja, University of Jendouba, Béja, Tunisia
| | - Hichem Sebai
- Laboratory of Functional Physiology and Valorization, Higher Institute of Biotechnology of Béja, University of Jendouba, Béja, Tunisia
| |
Collapse
|
6
|
Ortega-Tinoco S, Padilla-Orozco M, Hernández-Vázquez F, Garduño J, Mondragón-García A, Ramírez-Sánchez E, Bargas J, Hernández-López S. PACAP induces increased excitability in D1- and D2-expressing nucleus accumbens medium spiny neurons. Brain Res Bull 2025; 224:111323. [PMID: 40147707 DOI: 10.1016/j.brainresbull.2025.111323] [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: 11/27/2024] [Revised: 03/14/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
One of the main eating disorders associated with overweight and obesity is binge eating disorder. Binge eating is characterized by excessive consumption of high-calorie foods over a short period of time, approximately 2 hours. The nucleus accumbens (NAc) plays a key role in modulating the hedonic value of high-calorie foods, commonly referred to as palatable foods. Specific subregions of the shell portion of the NAc (NAcSh), known as hedonic hot spots, may play an important role in the motivational aspect of food consumption. Previous work has shown that the pituitary adenylate cyclase-activating polypeptide (PACAP) injected into the NAc reduces palatable food intake, suggesting that this peptide could be a potential tool for treating binge eating. However, the mechanisms of action of PACAP on the NAc are poorly understood. Here, we used whole-cell recording and calcium imaging techniques in NAcSh brain slices from D1-Cre and A2A-Cre mice to investigate PACAP modulation of medium spiny neuron (MSN) activity. We found that PACAP administration increased the firing rate of D1- and D2-expressing MSNs. In addition, in a binge-eating mouse model, nasal PACAP reduced binge-eating behavior.
Collapse
Affiliation(s)
- S Ortega-Tinoco
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Ciudad de México 04510, Mexico
| | - M Padilla-Orozco
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), Ciudad de México 04510, Mexico
| | - F Hernández-Vázquez
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Ciudad de México 04510, Mexico
| | - J Garduño
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Ciudad de México 04510, Mexico
| | - A Mondragón-García
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Ciudad de México 04510, Mexico
| | - E Ramírez-Sánchez
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Ciudad de México 04510, Mexico
| | - J Bargas
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), Ciudad de México 04510, Mexico
| | - S Hernández-López
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Ciudad de México 04510, Mexico.
| |
Collapse
|
7
|
Al-Karagholi MAM, Zhuang ZA, Beich S, Ashina H, Ashina M. PACAP38-induced migraine attacks are independent of CGRP signaling: a randomized controlled trial. J Headache Pain 2025; 26:79. [PMID: 40229719 PMCID: PMC11998216 DOI: 10.1186/s10194-025-02022-2] [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: 12/28/2024] [Accepted: 04/01/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Calcitonin gene-related peptide (CGRP) and pituitary adenylate cyclase-activating polypeptide-38 (PACAP38) are key pathogenic drivers of migraine. While CGRP has become the target of several mechanism-based therapies, less is known about PACAP38 signaling in migraine pathogenesis. Previous studies suggest that PACAP38 can modulate CGRP release, but it might also induce migraine attacks via CGRP-independent mechanisms. Whether PACAP38 signaling is independent of and parallel to CGRP signaling has implications for future therapeutic strategies. Here, we aimed to ascertain whether PACAP-38 can mediate migraine attacks independently of CGRP signaling by assessing the ability of eptinezumab to prevent PACAP38-induced migraine attacks. METHODS In a double-blind, placebo-controlled, parallel-group study, we randomly allocated adults with migraine without aura to receive either an intravenous infusion of 300-mg eptinezumab or matching placebo (isotonic saline) over 30 min. Two hours post-infusion, all participants were administered PACAP38 intravenously at 10 pmol/kg/min for 20 min. The primary endpoint was the incidence of migraine attacks during the 24-hour observational period post-infusion of eptinezumab or placebo. Key secondary endpoints included between-group differences in incidence of headache, and area under the curve (AUC) for headache intensity scores, diameter of the superficial temporal artery (STA) and facial skin blood flow. RESULTS A total of 38 participants were enrolled and completed the study. No difference was observed in the incidence of PACAP38-induced migraine attacks between the eptinezumab (10 [53%] of 19) and placebo (12 [63%] of 19) groups (Fisher's exact test: P = 0.74). Headache of any intensity was reported by 15 (79%) participants in the eptinezumab group, compared with 16 (84%) participants in the placebo group (Fisher's exact test: P > 0.99). The AUC for headache intensity scores did not differ between the two groups during the first 12 h post-infusion of PACAP38 (Mann-Whitney U-test: P = 0.96). No differences were observed in AUC between the eptinezumab and placebo groups with respect to changes in STA diameter and facial skin blood flow (P > 0.05). No serious adverse events occurred. CONCLUSIONS Our results suggest that PACAP38 may mediate its signaling independently of CGRP in migraine pathogenesis. Therapies targeting PACAP signaling are thus a promising new avenue for treating migraine. TRIAL REGISTRATION ClinicalTrials.gov, NCT05635604. Registered on November 15 2022.
Collapse
Affiliation(s)
| | - Zixuan Alice Zhuang
- Department of Neurology, Danish Headache Center, Copenhagen University Hospital- Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Signe Beich
- Department of Neurology, Danish Headache Center, Copenhagen University Hospital- Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Håkan Ashina
- Department of Neurology, Danish Headache Center, Copenhagen University Hospital- Rigshospitalet, Copenhagen, Denmark
- Translational Research Center, Copenhagen University Hospital- Rigshospitalet, Copenhagen, Denmark
| | - Messoud Ashina
- Department of Neurology, Danish Headache Center, Copenhagen University Hospital- Rigshospitalet, Copenhagen, Denmark.
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
8
|
Asano S, Ozasa K, Uehara T, Yokoyama R, Nakazawa T, Yanamoto S, Ago Y. Dimerisation of the VIP receptor VIPR2 is essential to its binding VIP and Gα i proteins, and to its functions in breast cancer cells. Br J Pharmacol 2025. [PMID: 40203889 DOI: 10.1111/bph.70039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 02/02/2025] [Accepted: 03/04/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND AND PURPOSE Several G protein-coupled receptors (GPCRs) are known to homodimerise. Dimeric GPCRs may have different properties from their monomers, but the molecular basis and functional significance of GPCR dimerisation remain largely unknown. We recently found that signalling by the vasoactive intestinal peptide receptor, VIPR2, regulates breast cancer cell migration and proliferation. However, it is unclear whether VIPR2 monomers directly interact with each other and what function the dimeric receptor has. Here, we showed that VIPR2 dimerises and investigated their role in breast cancer progression. EXPERIMENTAL APPROACH Dimerisation of VIPR2 was assessed by fluorescence resonance energy transfer (FRET) analysis and a pull-down assay. Breast cancer progression was analysed by orthotopic growth and metastasis of human breast cancers into proper axillary and subiliac lymph-nodes in mice. KEY RESULTS VIPR2 monomers directly interacted with each other through transmembrane domains (TM)3-4. FRET analysis revealed that VIPR2 moved further apart in cells expressing TM3-4-peptides, suggesting that TM3-4 prevents VIPR2 dimerisation. Breast cancer cells stably expressing TM3-4 region exhibited suppressed tumour growth and lymph-node metastasis. Furthermore, ligand-receptor binding assays revealed that VIP-FITC bound to cells dose-dependently, and VIPR2 de-dimerisation by TM3-4 expression decreased VIP's affinity to cells. Additionally, TM3-4 expression decreased Gαi-VIPR2 interactions. CONCLUSION AND IMPLICATIONS Dimeric VIPR2 forms the minimal functional unit that effectively promotes growth and metastasis of breast cancer. Therefore, dimeric VIPR2 is a potential therapeutic target for breast cancer, and TM3-4-peptides are potential anti-cancer drug candidates to suppress cancer progression.
Collapse
Affiliation(s)
- Satoshi Asano
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- School of Dentistry, Hiroshima University, Hiroshima, Japan
| | - Kairi Ozasa
- School of Dentistry, Hiroshima University, Hiroshima, Japan
| | - Teru Uehara
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Department of Oral Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Rei Yokoyama
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- School of Dentistry, Hiroshima University, Hiroshima, Japan
| | - Takanobu Nakazawa
- Department of Bioscience, Graduate School of Life Sciences, Tokyo University of Agriculture, Tokyo, Japan
| | - Souichi Yanamoto
- School of Dentistry, Hiroshima University, Hiroshima, Japan
- Department of Oral Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yukio Ago
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- School of Dentistry, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
9
|
Wang YW, Yang XH, Zheng XH, Zhou GS, Zhao XX, Zhao YL, Wu SH. Unraveling the relationship between inflammation and cluster headache. Front Neurol 2025; 16:1548522. [PMID: 40248013 PMCID: PMC12003110 DOI: 10.3389/fneur.2025.1548522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/19/2025] [Indexed: 04/19/2025] Open
Abstract
Cluster headache (CH) is often referred to as the 'suicide headache.' Existing research suggests that the activation of the trigeminal-vascular system, increased sensitivity of nerve fibers, and the release and interaction of various neuropeptides and inflammatory mediators may contribute to neurogenic inflammation, which serves as a crucial pathophysiological basis for the development of CH. Additionally, some neuropeptides can modulate neuronal activity related to pain transmission and may increase pain perception by sensitizing central nerves. This review discusses the neuropeptides and inflammatory mediators associated with CH neuroinflammation, focusing on calcitonin gene-related peptide (CGRP), inflammatory cytokines and related signaling pathways, nitric oxide (NO), pituitary adenylate cyclase-activating peptide 38 (PACAP-38), and vasoactive intestinal peptide (VIP), incorporating both preclinical and clinical evidence to provide new insights into potential therapeutic targets for CH.
Collapse
Affiliation(s)
- Yu-Wen Wang
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xu-Hong Yang
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xin-Hui Zheng
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Gao-Shui Zhou
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xiao-Xia Zhao
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yi-Lan Zhao
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Shu-Hong Wu
- Chongqing Beibei District Hospital of Traditional Chinese Medicine, Chongqing, China
| |
Collapse
|
10
|
Mangutov E, Dripps I, Siegersma K, Zhang Y, Bocian R, Asif S, Halbesma T, Witkowski W, Pradhan AA. Activation of δ-opioid receptors blocks allodynia in a model of headache induced by PACAP. Br J Pharmacol 2025; 182:1630-1643. [PMID: 39797405 DOI: 10.1111/bph.17424] [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: 05/03/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND AND PURPOSE Pituitary adenylate cyclase activating polypeptide (PACAP) is a human migraine trigger that is being targeted for migraine. The δ-opioid receptor (δ-receptor) is a novel target for the treatment of migraine, but its mechanism remains unclear. The goals of this study were to develop a mouse PACAP-headache model using clinically significant doses of PACAP; determine the effects of δ-receptor activation in this model; and investigate the co-expression of δ-receptors, PACAP and PACAP-PAC1 receptor. EXPERIMENTAL APPROACH Cephalic allodynia to low doses of acute and chronic PACAP were tested. A triptan (sumatriptan) and a CGRP receptor antagonist (olcegepant) were tested in this model. The δ-receptor agonist SNC80 was tested in PACAP and CGRP-induced headache models. Expression of PACAP, PAC1, CRLR and δ-receptors was determined using in situ hybridisation. KEY RESULTS Low doses of PACAP produced dose-dependent acute and chronic cephalic allodynia, blocked by sumatriptan but not by olcegepant. The PAC1 antagonist (M65) did not inhibit CGRP-induced allodynia. There was moderate co-expression of PAC1 and CRLR transcripts in migraine-related regions. SNC80 blocked PACAP- and CGRP-induced allodynia. There was low co-expression of PACAP and δ-receptors in brain regions measured. However, there was high co-expression of PAC1 and δ-receptors in somatosensory cortex, hippocampus and trigeminal nucleus caudalis. CONCLUSION AND IMPLICATIONS We developed a translationally significant model of PACAP-induced headache, which was mechanistically distinct from CGRP. Activation of δ-receptors blocked PACAP- and CGRP-induced allodynia, and δ-receptors were highly co-expressed with the PACAP-ergic system. Future studies will examine the functional relationship between δ-receptors and PAC1.
Collapse
MESH Headings
- Animals
- Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, delta/genetics
- Hyperalgesia/chemically induced
- Hyperalgesia/metabolism
- Hyperalgesia/drug therapy
- Male
- Disease Models, Animal
- Mice
- Mice, Inbred C57BL
- Piperazines/pharmacology
- Piperazines/therapeutic use
- Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide, Type I/metabolism
- Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide, Type I/genetics
- Azepines/pharmacology
- Sumatriptan/pharmacology
- Migraine Disorders/drug therapy
- Migraine Disorders/chemically induced
- Migraine Disorders/metabolism
- Quinazolines/pharmacology
- Calcitonin Gene-Related Peptide Receptor Antagonists/pharmacology
- Benzamides/pharmacology
Collapse
Affiliation(s)
- Elizaveta Mangutov
- Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Isaac Dripps
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Kendra Siegersma
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Yanping Zhang
- Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Rebecca Bocian
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Sarah Asif
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Timothy Halbesma
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Wiktor Witkowski
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Amynah A Pradhan
- Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
11
|
Sato K, Chiba S, Hatakeyama T, Matsuwaki T, Watanabe G, Kawaguchi M. Administration of a Selective Antagonist for Pituitary Adenylate Cyclase-Activating Polypeptide Receptor in the Hippocampus Causes Anxiolytic Effects in the Male Rat. Neuropsychopharmacol Rep 2025; 45:e12514. [PMID: 39723441 DOI: 10.1002/npr2.12514] [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: 08/31/2024] [Revised: 11/15/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) affects rodents' stress-related behaviors, such as anxiety-like behavior or fear conditioning. However, previous studies have investigated the effect of intracerebroventricular, but not hippocampal, injection of this PAC1R-selective antagonist (PACAP-6-38) on anxiety-like behavior. However, it has been reported that administration of PACAP-6-38 to the dorsal hippocampus reduces the fear response in a fear conditioning test. Therefore, this study aimed to determine whether the effect of dorsal hippocampal PACAP-6-38 injection in Sprague-Dawley rats can affect anxiety-like behavior assessed by an elevated plus-maze test. As a result, rats treated with PACAP-6-38 spent longer time in open arms than those with saline, suggesting that this drug has an anxiolytic effect. In conclusion, the role of PACAP in the dorsal hippocampus can promote anxiety-like behavior.
Collapse
Affiliation(s)
- Kaito Sato
- Lab of Animal Behavior and Environmental Science, Graduate School of Agriculture, Meiji University, Kawasaki, Japan
| | - Shuichi Chiba
- Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Japan
| | - Taichi Hatakeyama
- Lab of Animal Behavior and Environmental Science, Graduate School of Agriculture, Meiji University, Kawasaki, Japan
- Organization for the Strategic Coordination of Research and Intellectual Property, Meiji University, Kawasaki, Japan
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Japan
| | - Takashi Matsuwaki
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Japan
| | - Gen Watanabe
- Lab of Animal Behavior and Environmental Science, Graduate School of Agriculture, Meiji University, Kawasaki, Japan
- Organization for the Strategic Coordination of Research and Intellectual Property, Meiji University, Kawasaki, Japan
| | - Maiko Kawaguchi
- Lab of Animal Behavior and Environmental Science, Graduate School of Agriculture, Meiji University, Kawasaki, Japan
| |
Collapse
|
12
|
Carpenter BA, Pirino BE, Jennings MC, Samels SB, Shah KS, Maser J, Gupta M, Barson JR. Pituitary adenylate cyclase-activating polypeptide (PACAP)overexpression in the paraventricular nucleus of the thalamus alters motivated and affective behavior in female rats. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2025; 49:539-550. [PMID: 39746847 PMCID: PMC11928257 DOI: 10.1111/acer.15525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/17/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Pituitary adenylate cyclase-activating polypeptide (PACAP) has been found to be involved in a wide range of motivated and affective behaviors. While the PACAP-38 isoform is more densely expressed than PACAP-27 in most of the brain, PACAP-27 is more highly expressed in the rodent paraventricular nucleus of the thalamus (PVT), where females also have greater expression than males. Notably, the role of PACAP-27 expression in cells of the PVT has not been explored. METHODS Adult, female Long-Evans rats were injected in the PVT with an adeno-associated virus (AAV) to increase expression of PACAP or a control AAV. They were then investigated for subsequent ethanol drinking and preference; sucrose drinking and preference; or locomotor activity in a novel chamber, behavior in a light-dark box, behavior in a novelty suppression of feeding test, locomotor activity in a familiar activity chamber, and behavior in a forced swim test. RESULTS Injection with the PACAP AAV resulted in a specific increase in levels of PACAP-27. Rats injected with the PACAP AAV demonstrated reduced drinking and preference for ethanol under the intermittent-access procedure compared to those injected with the control AAV. In contrast, rats injected with the PACAP AAV showed no significant difference in drinking or preference for sucrose, or in any affective behavior tested, except that they spent less time swimming in the forced swim test. CONCLUSIONS In light of the low overall level of expression of PACAP-27 in the brain, the ability of PACAP-27 in the PVT to control ethanol drinking, with minimal effects on other motivated or affective behaviors, supports the idea that compounds related to PACAP-27 should be investigated as potential therapeutics for the treatment of alcohol use disorder.
Collapse
Affiliation(s)
- Brody A. Carpenter
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129
| | - Breanne E. Pirino
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129
| | - Malcolm C. Jennings
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129
| | - Shanna B. Samels
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129
| | - Krisha S. Shah
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129
| | - Joya Maser
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129
| | - Medha Gupta
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129
| | - Jessica R. Barson
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129
| |
Collapse
|
13
|
Ebner K, Fontebasso V, Ferro F, Singewald N, Hannibal J. PACAP regulates neuroendocrine and behavioral stress responses via CRF-containing neurons of the rat hypothalamic paraventricular nucleus. Neuropsychopharmacology 2025; 50:519-530. [PMID: 39472527 PMCID: PMC11735793 DOI: 10.1038/s41386-024-02016-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 10/08/2024] [Accepted: 10/11/2024] [Indexed: 01/18/2025]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide widely distributed in the brain including the hypothalamic paraventricular nucleus (PVN) implying a regulatory role in stress function. Recent evidence indicates that one of the main targets of PACAP within the PVN are corticotropin-releasing factor (CRF) neurons, which are key regulators of the hypothalamic-pituitary-adrenal (HPA) axis. However, the neural correlates that mediate PACAP effects on stress function are not fully understood. In the present study, we characterized the neuronal mechanism by which PACAP regulates neuroendocrine and behavioral stress responses in rats. We found that intracerebroventricular administration of PACAP increased the swim stress-induced c-Fos expression in distinct brain areas of the stress and anxiety circuitry including the parvocellular part of the PVN and changed behavioral stress coping during forced swimming to a more passive coping style (i.e., indicated by increased floating and reduced struggling behavior). Subsequently, PACAP administration directly into the PVN mimicked these behavioral effects and potentiated the plasma ACTH response to forced swim stress suggesting an excitatory role of PACAP on HPA stress axis reactivity. In addition, immunohistochemical analysis revealed a considerable portion of stress-activated CRF neurons in the medial parvocellular part of the PVN that co-localized PAC1 receptors suggesting that PACAP-induced effects on stress function are likely mediated directly by activation of CRF neurons in the PVN. Thus, these findings suggest that the PVN may represent one of the key areas where PACAP regulates the neuroendocrine and behavioral stress response.
Collapse
Affiliation(s)
- Karl Ebner
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria.
| | - Veronica Fontebasso
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Federico Ferro
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Nicolas Singewald
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Jens Hannibal
- Faculty of Health and Medical Sciences, Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Faculty of Health Sciences, Bispebjerg and Frederiksberg Hospital, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
14
|
Cengiz TB, Kulkarni R, Corbett V, Ghesani NV, Wolin E, Ghesani MV. Case report: Resolution of VIPoma-related symptoms with peptide receptor radionuclide therapy. Front Oncol 2025; 14:1432758. [PMID: 39845312 PMCID: PMC11750990 DOI: 10.3389/fonc.2024.1432758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 10/21/2024] [Indexed: 01/24/2025] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) is used for the management of neuroendocrine tumors (NETs) not responsive to somatostatin analogs. In this case series, we report two patients with pancreatic vasoactive intestinal peptide (VIP)-secreting NETs (VIPomas) not responsive to any other therapies who achieved symptomatic control and a significant decrease in serum VIP levels with PRRT during their hospital stay. Two patients with VIPomas were admitted to the hospital with multiple prior hospital admissions after going through multiple lines of therapy. The first patient was a 47-year-old woman with a grade 2 pancreatic VIP-secreting NET. She was treated with somatostatin analogs and chemotherapy; however, she experienced recurrent symptoms and complications leading to two hospital admissions, one of which included an intensive care unit (ICU) admission. The patient was treated with lutetium-177 DOTATATE while in the hospital, which led to the resolution of the symptoms and a marked decline in serum VIP levels, and she was able to be discharged from the hospital after 147 days of hospitalization (16 days after PRRT). The second patient was a 27-year-old man who was diagnosed with a well-differentiated grade 3 pancreatic VIPoma. He also suffered from severe diarrhea and was not responsive to any form of therapy, including liver embolization. He was also treated with PRRT on admission, which led to immediate resolution of his symptoms and a decrease in tumor markers. In conclusion, 177Lu-DOTATATE is an effective treatment for highly symptomatic VIPoma. Inpatient administration of PRRT can rapidly reduce diarrhea and fluid and electrolyte complications of VIP secretion and may shorten hospital stays.
Collapse
Affiliation(s)
- Turgut Bora Cengiz
- Department of Nuclear Medicine, Mount Sinai Hospital at Icahn School of Medicine, New York, NY, United States
| | - Raksha Kulkarni
- Department of Nuclear Medicine, Mount Sinai Hospital at Icahn School of Medicine, New York, NY, United States
| | - Virginia Corbett
- Department of Hematology and Oncology, Mount Sinai Hospital at Icahn School of Medicine, New York, NY, United States
| | - Nasrin V. Ghesani
- Department of Nuclear Medicine, Mount Sinai Hospital at Icahn School of Medicine, New York, NY, United States
| | - Edward Wolin
- Department of Hematology and Oncology, Mount Sinai Hospital at Icahn School of Medicine, New York, NY, United States
| | - Munir V. Ghesani
- Department of Nuclear Medicine, Mount Sinai Hospital at Icahn School of Medicine, New York, NY, United States
| |
Collapse
|
15
|
He C, Wang Q, Gao J, Chen H, Tong P. Neuro-immune regulation in allergic Diseases: Role of neuropeptides. Int Immunopharmacol 2025; 145:113771. [PMID: 39667047 DOI: 10.1016/j.intimp.2024.113771] [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: 09/11/2024] [Revised: 11/05/2024] [Accepted: 11/30/2024] [Indexed: 12/14/2024]
Abstract
The role of neuro-immune interaction in allergic diseases, a group of common immune system diseases, has garnered increasing attention. Neuropeptides, as a crucial component of neuro-immune crosstalk with local neuroendocrine and signaling functions, play a significant role that must not be overlooked. Neuropeptides are released by neurons and even some immune cells, and mediate neuro-immune crosstalk by activating relevant specific receptors on immune cells. Recent studies have found that neuropeptides have a certain regulatory effect on allergic diseases, which could be beneficial or detrimental for the development of allergic diseases. Nevertheless, the precise mechanism of neuropeptides in allergic diseases remains unclear, particularly in the context of food allergy where their role is poorly understood. This review summarized the interplay between neuropeptides and different immune cells, as well as their current research progress in several common allergic diseases: atopic dermatitis, allergic asthma, and food allergy. It is evident that neuropeptides such as substance P, calcitonin gene-related peptide, vasoactive intestinal peptide, and neuromedin U, exert important regulatory effects on allergic diseases, yet further investigation is required to fully elucidate their mechanisms of action, which may contribute to better understanding of the onset and progression of allergic diseases and finding better immunomodulatory strategies.
Collapse
Affiliation(s)
- Cuiying He
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China; College of Food Science & Technology, Nanchang University, Nanchang 330047, China
| | - Qian Wang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China; College of Food Science & Technology, Nanchang University, Nanchang 330047, China
| | - Jinyan Gao
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China; College of Food Science & Technology, Nanchang University, Nanchang 330047, China; Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, 330047, China
| | - Hongbing Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China; Sino-German Joint Research Institute (Jiangxi-OAI), Nanchang University, Nanchang 330047, China; Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, 330047, China
| | - Ping Tong
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China; Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, 330047, China
| |
Collapse
|
16
|
Incontro S, Musella ML, Sammari M, Di Scala C, Fantini J, Debanne D. Lipids shape brain function through ion channel and receptor modulations: physiological mechanisms and clinical perspectives. Physiol Rev 2025; 105:137-207. [PMID: 38990068 DOI: 10.1152/physrev.00004.2024] [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: 01/16/2024] [Revised: 05/28/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024] Open
Abstract
Lipids represent the most abundant molecular type in the brain, with a fat content of ∼60% of the dry brain weight in humans. Despite this fact, little attention has been paid to circumscribe the dynamic role of lipids in brain function and disease. Membrane lipids such as cholesterol, phosphoinositide, sphingolipids, arachidonic acid, and endocannabinoids finely regulate both synaptic receptors and ion channels that ensure critical neural functions. After a brief introduction on brain lipids and their respective properties, we review here their role in regulating synaptic function and ion channel activity, action potential propagation, neuronal development, and functional plasticity and their contribution in the development of neurological and neuropsychiatric diseases. We also provide possible directions for future research on lipid function in brain plasticity and diseases.
Collapse
Affiliation(s)
| | | | - Malika Sammari
- UNIS, INSERM, Aix-Marseille Université, Marseille, France
| | | | | | | |
Collapse
|
17
|
Garnica-Siqueira MC, Martins AB, Monteiro ÉCAM, Oliveira MHBD, Baratto CDR, Tsutsui FTK, Oliveira LLFD, Stopa LRDS, Souza CFD, Wunderlich ALM, Zaia DAM, Leite CM, Zaia CTBV, Uchoa ET. Estrogens impair hypophagia and hypothalamic cell activation induced by vasoactive intestinal peptide, but not by pituitary adenylate cyclase-activating polypeptide. Peptides 2025; 183:171325. [PMID: 39617208 DOI: 10.1016/j.peptides.2024.171325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 11/27/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
The neuropeptides vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating polypeptide (PACAP) act in arcuate (ARC) and paraventricular (PVN) hypothalamic nuclei, reducing food intake and changing plasma parameters. Estrogens (E) also regulate energy homeostasis, and loss of ovarian function leads to hyperphagia and body weight gain. This study aimed to evaluate the effects of estradiol (E) in a postmenopausal rat model, ovariectomy (OVX), on PAC1 and VPAC2 receptors in the PVN and ARC, as well as on food intake, plasma parameters, and PVN and ARC cell activation in response to intracerebroventricular microinjection of VIP and PACAP. For this, the rats underwent intracerebroventricular and OVX surgeries, being treated daily with subcutaneous injections of 0.2 mL of corn oil or 10 μg/0.2 mL of estradiol cypionate, comprising the OVX+O and OVX+E groups, respectively. OVX+E showed reduced VPAC2 mRNA expression in the PVN. PACAP reduced food intake in both groups, and VIP-induced hypophagia was not observed in OVX+E. VIP increased plasma glucose in both groups, and PACAP increased plasma glucose only in OVX+O. VIP decreased free fatty acids in OVX+E. Furthermore, PACAP increased ARC cell activation in both groups, and in the PVN only in OVX+O. Cell activation induced by VIP in ARC and PVN was blocked by estradiol. Therefore, estrogens disrupted the hypophagia induced by VIP, but not by PACAP, and these differences seem to be, at least in part, due to an impairment of the activation of the ARC-PVN pathway.
Collapse
Affiliation(s)
- Marcela Cristina Garnica-Siqueira
- Laboratório de Fisiologia, Hospital de Reabilitação de Anomalias Craniofaciais, Universidade de São Paulo, Bauru, SP, Brazil; Programa Multicêntrico de Pós-Graduação em Ciências Fisiológicas, Universidade Estadual de Londrina, Londrina, PR, Brazil
| | - Andressa Busetti Martins
- Programa Multicêntrico de Pós-Graduação em Ciências Fisiológicas, Universidade Estadual de Londrina, Londrina, PR, Brazil
| | - Érica Cristina Alves Munhoz Monteiro
- Programa de Iniciação Científica do Conselho Nacional de Desenvolvimento Científico e Tecnológico, Universidade Estadual de Londrina, Londrina, PR, Brazil
| | - Maria Heloisa Bernardes de Oliveira
- Programa de Iniciação Científica do Conselho Nacional de Desenvolvimento Científico e Tecnológico, Universidade Estadual de Londrina, Londrina, PR, Brazil
| | - Carolina Dos Reis Baratto
- Programa de Iniciação Científica do Conselho Nacional de Desenvolvimento Científico e Tecnológico, Universidade Estadual de Londrina, Londrina, PR, Brazil
| | | | | | - Larissa Rugila Dos Santos Stopa
- Programa Multicêntrico de Pós-Graduação em Ciências Fisiológicas, Universidade Estadual de Londrina, Londrina, PR, Brazil; Programa de Pós-Graduação em Ciências Fisiológicas, Universidade Estadual de Londrina, Londrina, PR, Brazil
| | - Camila Franciele de Souza
- Programa Multicêntrico de Pós-Graduação em Ciências Fisiológicas, Universidade Estadual de Londrina, Londrina, PR, Brazil; Programa de Pós-Graduação em Ciências Fisiológicas, Universidade Estadual de Londrina, Londrina, PR, Brazil
| | | | - Dimas Augusto Morozin Zaia
- Laboratório de Química Prebiótica, Departamento de Química, Universidade Estadual de Londrina, Londrina, PR, Brazil
| | | | - Cássia Thaïs Bussamra Vieira Zaia
- Programa Multicêntrico de Pós-Graduação em Ciências Fisiológicas, Universidade Estadual de Londrina, Londrina, PR, Brazil; Programa de Pós-Graduação em Ciências Fisiológicas, Universidade Estadual de Londrina, Londrina, PR, Brazil; Laboratório de Fisiologia Neuroendócrina e Metabolismo, Departamento de Ciências Fisiológicas, Universidade Estadual de Londrina, Londrina, PR, Brazil.
| | - Ernane Torres Uchoa
- Programa Multicêntrico de Pós-Graduação em Ciências Fisiológicas, Universidade Estadual de Londrina, Londrina, PR, Brazil; Programa de Pós-Graduação em Ciências Fisiológicas, Universidade Estadual de Londrina, Londrina, PR, Brazil; Laboratório de Fisiologia Neuroendócrina e Metabolismo, Departamento de Ciências Fisiológicas, Universidade Estadual de Londrina, Londrina, PR, Brazil.
| |
Collapse
|
18
|
Ren L, Fan Y, Luo H, Hu J, Hu J. PACAP/VIP in the prefrontal cortex mediates the rapid antidepressant effects of zhizichi decoction. JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118638. [PMID: 39084272 DOI: 10.1016/j.jep.2024.118638] [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: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Zhizichi decoction (ZZCD) is a traditional Chinese medicine formula that consists of Gardenia jasminoides J.Ellis (GJ) and Semen Sojae Praeparatum. It is used to treat insomnia and emotion-related disorders, such as irritability. Previous studies have found that GJ has a rapid antidepressant effect. The study found that ZZCD is safer than GJ at the same dosage. Consequently, ZZCD is a superior drug with quicker antidepressant effects than GJ. The rapid antidepressant effects of ZZCD were examined in this study, along with the components that make up this effect. It was determined that the activation of prefrontal Pituitary Adenylate Cyclase Activating Polypeptide (PACAP)/Vasoactive Intestinal Polypeptide (VIP) is essential for ZZCD's rapid antidepressant effects. AIM This study identified and discussed the rapid antidepressant effects and biological mechanisms of ZZCD. MATERIALS AND METHODS The tail suspension test (TST) and the forced swimming test (FST) were used to screen the effective dosage of ZZCD (0.67 g/kg, 1 g/kg, 4 g/kg). The effective dosage of ZZCD (1 g/kg) was tested in the TST conducted on Institute of Cancer Research (ICR) mice that were treated with lipopolysaccharide (LPS) at a concentration of 0.1 mg/mL. To confirm the expression of c-Fos, PACAP, and VIP in the prefrontal cortex (PFC), immunohistochemistry tests were conducted on mice following intragastric injection of ZZCD. Chemical characterization analysis and HPLC quality control analysis were conducted using UHPLC-Q-Obitrap-HRMS and chromatographic analysis. RESULTS The results showed that an acute administration of ZZCD (1 g/kg) decreased the immobility time of Kunming (KM) mice in TST and FST. Depressive behaviors in TST-induced ICR mice treated with LPS (0.1 mg/mL) were reversed by ZZCD (1 g/kg). The results of immunohistochemical experiments showed that ZZCD (1 g/kg) activated neurons in the PFC and PACAP/VIP in the PFC. In this study, 22 substances in ZZCD were identified. Five primary distinctive fingerprint peaks-geniposide, genistin, genipin-1-β-D-gentiobioside, glycitin, and daidzin-were found among the ten common peaks. CONCLUSION ZZCD (1 g/kg) had significant rapid antidepressant effects. PACAP/VIP in the PFC was found to mediate the rapid antidepressant effects of ZZCD.
Collapse
Affiliation(s)
- Li Ren
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Sichuan, Chengdu, 611137, China.
| | - Yue Fan
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Sichuan, Chengdu, 611137, China
| | - Huoqing Luo
- School of Life Science and Technology, ShanghaiTech University Shanghai, 100 Haike Road, Pudong New District, Shanghai, 201210, China
| | - Ji Hu
- School of Life Science and Technology, ShanghaiTech University Shanghai, 100 Haike Road, Pudong New District, Shanghai, 201210, China
| | - Jingqing Hu
- Institute of Basic Theory of Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, 16 Nanxiaojie, Dongzhimennei, Dongcheng District, Beijing, 100700, China.
| |
Collapse
|
19
|
Peres MFP. PACAP pathway: a new frontier in migraine prevention. Lancet Neurol 2024; 23:1181-1182. [PMID: 39577907 DOI: 10.1016/s1474-4422(24)00444-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 10/29/2024] [Indexed: 11/24/2024]
Affiliation(s)
- Mario Fernando Prieto Peres
- Hospital Israelita Albert Einstein, Sao Paulo, Brazil; Instituto de Psiquiatria, HCFMUSP, Sao Paulo, Brazil.
| |
Collapse
|
20
|
Wang Y, Sen-Majumdar A, Li JM, Sarkar S, Passang T, Li Y, Cohen J, Chen Z, Chaudagar K, Das PK, Wang S, Bruk N, Papadantonakis N, Giver CR, Waller EK. Identification and characterization of vasoactive intestinal peptide receptor antagonists with high-affinity and potent anti-leukemia activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.08.622716. [PMID: 39605448 PMCID: PMC11601233 DOI: 10.1101/2024.11.08.622716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Vasoactive intestinal peptide (VIP) is a neuropeptide involved in tumor growth and immune modulating functions. Previous research indicated that a VIP antagonist (VIPhyb) enhances T-cell activation and induces T-cell-dependent anti-leukemic activity in mice. We created a combinatorial library of VIPhyb C-terminal sequence variations to develop a more potent VIP-receptor (VIP-R) antagonist, hypothesizing that specific amino acid substitutions would improve receptor binding and plasma stability. In silico screening analyses identified sequences with improved docking scores predicting increased binding affinity to human VIP receptors VPAC1 and VPAC2. Fifteen peptides were synthesized and tested for their ability to potentiate activation of purified mouse and human T cells and enhance T cell-dependent anti-leukemia responses in murine models of acute myeloid leukemia. Treating C57Bl/6 mice engrafted with a C1498 myeloid leukemia cell line with daily subcutaneous injections of VIP-R antagonist peptides induced T cell activation resulting in specific anti-leukemia responses. Strikingly, the predicted binding affinity of the VIP-R antagonists to VIP receptors correlated positively with their ability to augment mouse T-cell proliferation and anti-leukemia activity. ANT308 and ANT195 emerged as top candidates due to their high predicted VIP-R binding, low EC 50 for in vitro T cell activation, and potent anti-leukemia activities. ANT308 decreased CREB phosphorylation, a downstream signaling pathway of the VIP receptor, and stimulated granzyme B and perforin expression in CD8+ T cells from AML patients. Combining in silico modeling, in vitro T cell activation properties, and in vivo anti-leukemia activity has identified promising VIP-R antagonist candidates for further development as novel immunotherapies for AML, especially for patients with relapsed disease.
Collapse
|
21
|
Nielipińska D, Rubiak D, Pietrzyk-Brzezińska AJ, Małolepsza J, Błażewska KM, Gendaszewska-Darmach E. Stapled peptides as potential therapeutics for diabetes and other metabolic diseases. Biomed Pharmacother 2024; 180:117496. [PMID: 39362065 DOI: 10.1016/j.biopha.2024.117496] [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: 07/04/2024] [Revised: 09/10/2024] [Accepted: 09/24/2024] [Indexed: 10/05/2024] Open
Abstract
The field of peptide drug research has experienced notable progress, with stapled peptides featuring stabilized α-helical conformation, emerging as a promising field. These peptides offer enhanced stability, cellular permeability, and binding affinity and exhibit potential in the treatment of diabetes and metabolic disorders. Stapled peptides, through the disruption of protein-protein interactions, present varied functionalities encompassing agonism, antagonism, and dual-agonism. This comprehensive review offers insight into the technology of peptide stapling and targeting of crucial molecular pathways associated with glucose metabolism, insulin secretion, and food intake. Additionally, we address the challenges in developing stapled peptides, including concerns pertaining to structural stability, peptide helicity, isomer mixture, and potential side effects.
Collapse
Affiliation(s)
- Dominika Nielipińska
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Poland.
| | - Dominika Rubiak
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Poland
| | - Agnieszka J Pietrzyk-Brzezińska
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Poland
| | - Joanna Małolepsza
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Poland
| | - Katarzyna M Błażewska
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Poland.
| | - Edyta Gendaszewska-Darmach
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Poland.
| |
Collapse
|
22
|
Arteaga-Blanco LA, Temerozo JR, Tiné LPS, Dantas-Pereira L, Sacramento CQ, Fintelman-Rodrigues N, Toja BM, Gomes Dias SS, de Freitas CS, Espírito-Santo CC, Silva YP, Frozza RL, Bozza PT, Menna-Barreto RFS, Souza TML, Bou-Habib DC. Extracellular vesicles from primary human macrophages stimulated with VIP or PACAP mediate anti-SARS-CoV-2 activities in monocytes through NF-κB signaling pathway. Microbes Infect 2024; 26:105400. [PMID: 39069117 DOI: 10.1016/j.micinf.2024.105400] [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: 01/30/2024] [Revised: 06/28/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Infection by SARS-CoV-2 is associated with uncontrolled inflammatory response during COVID-19 severe disease, in which monocytes are one of the main sources of pro-inflammatory mediators leading to acute respiratory distress syndrome. Extracellular vesicles (EVs) from different cells play important roles during SARS-CoV-2 infection, but investigations describing the involvement of EVs from primary human monocyte-derived macrophages (MDM) on the regulation of this infection are not available. Here, we describe the effects of EVs released by MDM stimulated with the neuropeptides VIP and PACAP on SARS-CoV-2-infected monocytes. MDM-derived EVs were isolated by differential centrifugation of medium collected from cells cultured for 24 h in serum-reduced conditions. Based on morphological properties, we distinguished two subpopulations of MDM-EVs, namely large (LEV) and small EVs (SEV). We found that MDM-derived EVs stimulated with the neuropeptides inhibited SARS-CoV-2 RNA synthesis/replication in monocytes, protected these cells from virus-induced cytopathic effects and reduced the production of pro-inflammatory mediators. In addition, EVs derived from VIP- and PACAP-treated MDM prevented the SARS-CoV-2-induced NF-κB activation. Overall, our findings suggest that MDM-EVs are endowed with immunoregulatory properties that might contribute to the antiviral and anti-inflammatory responses in SARS-CoV-2-infected monocytes and expand our knowledge of EV effects during COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Luis A Arteaga-Blanco
- Laboratory on Thymus Research, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil
| | - Jairo R Temerozo
- Laboratory on Thymus Research, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil; National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil
| | - Lucas P S Tiné
- Laboratory on Thymus Research, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil
| | - Luíza Dantas-Pereira
- Laboratory of Cellular Biology, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil
| | - Carolina Q Sacramento
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil; National Institute for Science and Technology on Innovation in Diseases of Neglected Populations, Center for Technological Development in Health, Fiocruz, Rio de Janeiro, 21040-360, Brazil
| | - Natalia Fintelman-Rodrigues
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil; National Institute for Science and Technology on Innovation in Diseases of Neglected Populations, Center for Technological Development in Health, Fiocruz, Rio de Janeiro, 21040-360, Brazil
| | - Beatriz M Toja
- Laboratory of Cellular and Molecular Cardiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, 21941-902, Brazil
| | - Suelen Silva Gomes Dias
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil
| | - Caroline S de Freitas
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil
| | | | - Ygor P Silva
- Laboratory on Thymus Research, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil
| | - Rudimar L Frozza
- Laboratory on Thymus Research, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil
| | - Patrícia T Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil
| | - Rubem F S Menna-Barreto
- Laboratory of Cellular Biology, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil
| | - Thiago Moreno L Souza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil; National Institute for Science and Technology on Innovation in Diseases of Neglected Populations, Center for Technological Development in Health, Fiocruz, Rio de Janeiro, 21040-360, Brazil
| | - Dumith Chequer Bou-Habib
- Laboratory on Thymus Research, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil; National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, 21040-360, Brazil.
| |
Collapse
|
23
|
Ashina H, Christensen RH, Hay DL, Pradhan AA, Hoffmann J, Reglodi D, Russo AF, Ashina M. Pituitary adenylate cyclase-activating polypeptide signalling as a therapeutic target in migraine. Nat Rev Neurol 2024; 20:660-670. [PMID: 39256637 DOI: 10.1038/s41582-024-01011-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2024] [Indexed: 09/12/2024]
Abstract
Migraine is a disabling neurological disorder that affects more than one billion people worldwide. The clinical presentation is characterized by recurrent headache attacks, which are often accompanied by photophobia, phonophobia, nausea and vomiting. Although the pathogenesis of migraine remains incompletely understood, mounting evidence suggests that specific signalling molecules are involved in the initiation and modulation of migraine attacks. These signalling molecules include pituitary adenylate cyclase-activating polypeptide (PACAP), a vasoactive peptide that is known to induce migraine attacks when administered by intravenous infusion to people with migraine. Discoveries linking PACAP to migraine pathogenesis have led to the development of drugs that target PACAP signalling, and a phase II trial has provided evidence that a monoclonal antibody against PACAP is effective for migraine prevention. In this Review, we explore the molecular and cellular mechanisms of PACAP signalling, shedding light on its role in the trigeminovascular system and migraine pathogenesis. We then discuss emerging therapeutic strategies that target PACAP signalling for the treatment of migraine and consider the research needed to translate the current knowledge into a treatment for migraine in the clinic.
Collapse
Affiliation(s)
- Håkan Ashina
- Department of Neurology, Danish Headache Center, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Translational Research Center, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Rune H Christensen
- Department of Neurology, Danish Headache Center, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Translational Research Center, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Debbie L Hay
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand
| | - Amynah A Pradhan
- Center for Clinical Pharmacology, Department of Anaesthesiology, Washington University in St Louis, St Louis, MO, USA
| | - Jan Hoffmann
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Dora Reglodi
- Department of Anatomy, Centre for Neuroscience, University of Pécs Medical School, Pécs, Hungary
| | - Andrew F Russo
- Department of Molecular Physiology and Biophysics, University of Iowa, Veterans Affairs Healthcare System, Iowa City, IA, USA
- Department of Neurology, University of Iowa, Veterans Affairs Healthcare System, Iowa City, IA, USA
| | - Messoud Ashina
- Department of Neurology, Danish Headache Center, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark.
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
24
|
Raggi A, Leonardi M, Arruda M, Caponnetto V, Castaldo M, Coppola G, Della Pietra A, Fan X, Garcia-Azorin D, Gazerani P, Grangeon L, Grazzi L, Hsiao FJ, Ihara K, Labastida-Ramirez A, Lange KS, Lisicki M, Marcassoli A, Montisano DA, Onan D, Onofri A, Pellesi L, Peres M, Petrušić I, Raffaelli B, Rubio-Beltran E, Straube A, Straube S, Takizawa T, Tana C, Tinelli M, Valeriani M, Vigneri S, Vuralli D, Waliszewska-Prosół M, Wang W, Wang Y, Wells-Gatnik W, Wijeratne T, Martelletti P. Hallmarks of primary headache: part 1 - migraine. J Headache Pain 2024; 25:189. [PMID: 39482575 PMCID: PMC11529271 DOI: 10.1186/s10194-024-01889-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/15/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND AND AIM Migraine is a common disabling conditions which, globally, affects 15.2% of the population. It is the second cause of health loss in terms of years lived with disability, the first among women. Despite being so common, it is poorly recognised and too often undertreated. Specialty centres and neurologists with specific expertise on headache disorders have the knowledge to provide specific care: however, those who do not regularly treat patients with migraine will benefit from a synopsis on the most relevant and updated information about this condition. This paper presents a comprehensive view on the hallmarks of migraine, from genetics and diagnostic markers, up to treatments and societal impact, and reports the elements that identify migraine specific features. MAIN RESULTS The most relevant hallmark of migraine is that it has common and individual features together. Besides the known clinical manifestations, migraine presentation is heterogeneous with regard to frequency of attacks, presence of aura, response to therapy, associated comorbidities or other symptoms, which likely reflect migraine heterogeneous genetic and molecular basis. The amount of therapies for acute and for prophylactic treatment is really wide, and one of the difficulties is with finding the best treatment for the single patient. In addition to this, patients carry out different daily life activities, and might show lifestyle habits which are not entirely adequate to manage migraine day by day. Education will be more and more important as a strategy of brain health promotion, because this will enable reducing the amount of subjects needing specialty care, thus leaving it to those who require it in reason of refractory condition or presence of comorbidities. CONCLUSIONS Recognizing the hallmarks of migraine and the features of single patients enables prescribing specific pharmacological and non-pharmacological treatments. Medical research on headaches today particularly suffers from the syndrome of single-disease approach, but it is important to have a cross-sectional and joint vision with other close specialties, in order to treat our patients with a comprehensive approach that a heterogeneous condition like migraine requires.
Collapse
Affiliation(s)
- Alberto Raggi
- Neurology, Public Health and Disability Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan, 20133, Italy.
| | - Matilde Leonardi
- Neurology, Public Health and Disability Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan, 20133, Italy
| | - Marco Arruda
- Department of Neuroscience, Glia Institute, Ribeirão Preto, Brazil
| | - Valeria Caponnetto
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Matteo Castaldo
- Department of Health Science and Technology, Faculty of Medicine, CNAP, Center for Sensory-Motor Interaction (SMI), Aalborg University, Gistrup, Denmark
- Department of Medicine and Surgery, Clinical Psychophysiology and Clinical Neuropsychology Labs, Parma University, Parma, Italy
| | - Gianluca Coppola
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome Polo Pontino ICOT, Latina, Italy
| | - Adriana Della Pietra
- Dept. Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Xiangning Fan
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - David Garcia-Azorin
- Department of Medicine, Toxicology and Dermatology, Faculty of Medicine, University of Valladolid, Valladolid, Spain
- Department of Neurology, Hospital Universitario Río Hortega, Valladolid, Spain
| | - Parisa Gazerani
- Department of Health Science and Technology, Faculty of Medicine, CNAP, Center for Sensory-Motor Interaction (SMI), Aalborg University, Gistrup, Denmark
- Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, Oslo, Norway
| | - Lou Grangeon
- Neurology Department, CHU de Rouen, Rouen, France
| | - Licia Grazzi
- Neuroalgology Unit and Headache Center, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Fu-Jung Hsiao
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Keiko Ihara
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
- Japanese Red Cross Ashikaga Hospital, Tochigi, Japan
| | - Alejandro Labastida-Ramirez
- Division of Neuroscience, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, UK
| | - Kristin Sophie Lange
- Department of Neurology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
| | - Marco Lisicki
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Alessia Marcassoli
- Neurology, Public Health and Disability Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan, 20133, Italy
| | - Danilo Antonio Montisano
- Neuroalgology Unit and Headache Center, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Dilara Onan
- Department of Physiotherapy and Rehabilitation, Faculty of Heath Sciences, Yozgat Bozok University, Yozgat, Turkey
| | - Agnese Onofri
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Lanfranco Pellesi
- Department of Public Health Clinical Pharmacology, Pharmacy and Environmental Medicine, University of Southern Denmark, Odense, Denmark
| | - Mario Peres
- Hospital Israelita Albert Einstein, São Paulo, Brazil
- Instituto de Psiquiatria; Hospital das Clínicas da Faculdade de Medicina da USP, Sao Paulo, Brazil
| | - Igor Petrušić
- Laboratory for Advanced Analysis of Neuroimages, Faculty of Physical Chemistry, University of Belgrade, Belgrade, Serbia
| | - Bianca Raffaelli
- Department of Neurology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
| | - Eloisa Rubio-Beltran
- Headache Group, Wolfson SPaRC, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Andreas Straube
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sebastian Straube
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
- School of Public Health, University of Alberta, Edmonton, AB, Canada
| | - Tsubasa Takizawa
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Claudio Tana
- Center of Excellence On Headache and Geriatrics Clinic, SS Annunziata Hospital of Chieti, Chieti, Italy
| | - Michela Tinelli
- Care Policy Evaluation Centre (CPEC), London School of Economics and Political Science, London, UK
| | - Massimiliano Valeriani
- Systems Medicine Department, University of Tor Vergata, Rome, Italy
- Developmental Neurology Unit, IRCSS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Simone Vigneri
- Neurology and Neurophysiology Service - Pain Medicine Unit, Santa Maria Maddalena Hospital, Occhiobello, Italy
| | - Doga Vuralli
- Department of Neurology and Algology, Neuropsychiatry Center, Neuroscience and Neurotechnology Center of Excellence (NÖROM), Gazi University Faculty of Medicine, Ankara, Türkiye
| | | | - Wei Wang
- Department of Neurology, Headache Center, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
- Department of Neurology, Headache Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yonggang Wang
- Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, China
| | | | - Tissa Wijeratne
- Department of Neurology, Sunshine Hospital, St Albans, VIC, Australia
- Australian Institute of Migraine, Pascoe Vale South, VIC, Australia
| | | |
Collapse
|
25
|
Xing L, Chen B, Qin Y, Li X, Zhou S, Yuan K, Zhao R, Qin D. The role of neuropeptides in cutaneous wound healing: a focus on mechanisms and neuropeptide-derived treatments. Front Bioeng Biotechnol 2024; 12:1494865. [PMID: 39539691 PMCID: PMC11557334 DOI: 10.3389/fbioe.2024.1494865] [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: 09/17/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
An extensive network of cutaneous nerves, neuropeptides, and specific receptors richly innervates the skin and influences a variety of physiological and pathological processes. The sensory and autonomic nerve fibers secrete a variety of neuropeptides that are essential to the different phases of wound healing. In addition to initiating a neurogenic inflammatory response in the early stages of healing, neuropeptides also control wound healing by influencing immune cells, repair cells, and the growth factor network. However, the precise mechanism by which they accomplish these roles in the context of cutaneous wound healing is still unknown. Investigating the mechanisms of action of neuropeptides in wound healing and potential therapeutic applications is therefore urgently necessary. The present review discusses the process of wound healing, types of neuropeptides, potential mechanisms underlying the role of neuropeptides in cutaneous wound healing, as well as some neuropeptide-derived treatment strategies, such as hydrogels, new dressings, electro stimulation, and skin-derived precursors. Future in-depth mechanistic studies of neuropeptides in cutaneous wound healing may provide opportunities to develop therapeutic technologies that harness the roles of neuropeptides in the wound healing process.
Collapse
Affiliation(s)
- Liwei Xing
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Bing Chen
- School of Medicine, Kunming University, Kunming, China
| | - Yuliang Qin
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
| | - Xinyao Li
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
| | - Sitong Zhou
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
| | - Kai Yuan
- Second Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Rong Zhao
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Dongdong Qin
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
| |
Collapse
|
26
|
Su L, Li G, Chow BKC, Cardoso JCR. Neuropeptides and receptors in the cephalochordate: A crucial model for understanding the origin and evolution of vertebrate neuropeptide systems. Mol Cell Endocrinol 2024; 592:112324. [PMID: 38944371 DOI: 10.1016/j.mce.2024.112324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/26/2024] [Accepted: 06/25/2024] [Indexed: 07/01/2024]
Abstract
Genomes and transcriptomes from diverse organisms are providing a wealth of data to explore the evolution and origin of neuropeptides and their receptors in metazoans. While most neuropeptide-receptor systems have been extensively studied in vertebrates, there is still a considerable lack of understanding regarding their functions in invertebrates, an extraordinarily diverse group that account for the majority of animal species on Earth. Cephalochordates, commonly known as amphioxus or lancelets, serve as the evolutionary proxy of the chordate ancestor. Their key evolutionary position, bridging the invertebrate to vertebrate transition, has been explored to uncover the origin, evolution, and function of vertebrate neuropeptide systems. Amphioxus genomes exhibit a high degree of sequence and structural conservation with vertebrates, and sequence and functional homologues of several vertebrate neuropeptide families are present in cephalochordates. This review aims to provide a comprehensively overview of the recent findings on neuropeptides and their receptors in cephalochordates, highlighting their significance as a model for understanding the complex evolution of neuropeptide signaling in vertebrates.
Collapse
Affiliation(s)
- Liuru Su
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Guang Li
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China.
| | - Billy K C Chow
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China.
| | - João C R Cardoso
- Comparative Endocrinology and Integrative Biology, Centre of Marine Sciences, Universidade do Algarve, 8005-139, Faro, Portugal.
| |
Collapse
|
27
|
Passang T, Wang S, Zhang H, Zeng F, Hsu PC, Wang W, Li JM, Liu Y, Ravindranathan S, Lesinski GB, Waller EK. VPAC2 Receptor Signaling Promotes Growth and Immunosuppression in Pancreatic Cancer. Cancer Res 2024; 84:2954-2967. [PMID: 38809694 PMCID: PMC11458156 DOI: 10.1158/0008-5472.can-23-3628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/29/2024] [Accepted: 05/20/2024] [Indexed: 05/31/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) harbors a complex tumor microenvironment, and cross-talk among cells in the tumor microenvironment can contribute to drug resistance and relapse. Vasoactive intestinal peptide (VIP) is overexpressed in PDAC, and VIP receptors expressed on T cells are a targetable pathway that sensitizes PDAC to immunotherapy. In this study, we showed that pancreatic cancer cells engage in autocrine VIP signaling through VIP receptor 2 (VPAC2). High coexpression of VIP with VPAC2 correlated with reduced relapse-free survival in patients with PDAC. VPAC2 activation in PDAC cells upregulated Piwi-like RNA-mediated gene silencing 2, which stimulated cancer cell clonogenic growth. In addition, VPAC2 signaling increased expression of TGFβ1 to inhibit T-cell function. Loss of VPAC2 on PDAC cells led to reduced tumor growth and increased sensitivity to anti-PD-1 immunotherapy in mouse models of PDAC. Overall, these findings expand our understanding of the role of VIP/VPAC2 signaling in PDAC and provide the rationale for developing potent VPAC2-specific antagonists for treating patients with PDAC. Significance: Autocrine VIP signaling via VPAC2 promotes cancer cell growth and inhibits T-cell function in pancreatic ductal adenocarcinoma, highlighting its potential as a therapeutic target to improve pancreatic cancer treatment.
Collapse
Affiliation(s)
- Tenzin Passang
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Shuhua Wang
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Hanwen Zhang
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Fanyuan Zeng
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Po-Chih Hsu
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Wenxi Wang
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Jian Ming Li
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Yuan Liu
- Winship Cancer Institute Emory University, Atlanta, GA, USA
| | - Sruthi Ravindranathan
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Gregory B. Lesinski
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Winship Cancer Institute Emory University, Atlanta, GA, USA
| | - Edmund K. Waller
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Winship Cancer Institute Emory University, Atlanta, GA, USA
| |
Collapse
|
28
|
Castorina A, Scheller J, Keay KA, Marzagalli R, Rose-John S, Campbell IL. Increased Expression of the Neuropeptides PACAP/VIP in the Brain of Mice with CNS Targeted Production of IL-6 Is Mediated in Part by Trans-Signalling. Int J Mol Sci 2024; 25:9453. [PMID: 39273398 PMCID: PMC11395455 DOI: 10.3390/ijms25179453] [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: 08/13/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
Inflammation with expression of interleukin 6 (IL-6) in the central nervous system (CNS) occurs in several neurodegenerative/neuroinflammatory conditions and may cause neurochemical changes to endogenous neuroprotective systems. Pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal polypeptide (VIP) are two neuropeptides with well-established protective and anti-inflammatory properties. Yet, whether PACAP and VIP levels are altered in mice with CNS-restricted, astrocyte-targeted production of IL-6 (GFAP-IL6) remains unknown. In this study, PACAP/VIP levels were assessed in the brain of GFAP-IL6 mice. In addition, we utilised bi-genic GFAP-IL6 mice carrying the human sgp130-Fc transgene (termed GFAP-IL6/sgp130Fc mice) to determine whether trans-signalling inhibition rescued PACAP/VIP changes in the CNS. Transcripts and protein levels of PACAP and VIP, as well as their receptors PAC1, VPAC1 and VPAC2, were significantly increased in the cerebrum and cerebellum of GFAP-IL6 mice vs. wild type (WT) littermates. These results were paralleled by a robust activation of the JAK/STAT3, NF-κB and ERK1/2MAPK pathways in GFAP-IL6 mice. In contrast, co-expression of sgp130Fc in GFAP-IL6/sgp130Fc mice reduced VIP expression and activation of STAT3 and NF-κB pathways, but it failed to rescue PACAP, PACAP/VIP receptors and Erk1/2MAPK phosphorylation. We conclude that forced expression of IL-6 in astrocytes induces the activation of the PACAP/VIP neuropeptide system in the brain, which is only partly modulated upon IL-6 trans-signalling inhibition. Increased expression of PACAP/VIP neuropeptides and receptors may represent a homeostatic response of the CNS to an uncontrolled IL-6 synthesis and its neuroinflammatory consequences.
Collapse
Affiliation(s)
- Alessandro Castorina
- Laboratory of Cellular and Molecular Neuroscience, School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia;
| | - Jurgen Scheller
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine University, 40225 Düsseldorf, Germany;
| | - Kevin A. Keay
- Discipline of Anatomy and Histology, School of Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia;
| | - Rubina Marzagalli
- Laboratory of Cellular and Molecular Neuroscience, School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia;
| | - Stefan Rose-John
- Institute of Biochemistry, Medical Faculty, Christian Albrechts University, 24098 Kiel, Germany;
| | - Iain L. Campbell
- School of Molecular Bioscience, University of Sydney, Sydney, NSW 2006, Australia;
| |
Collapse
|
29
|
Thakur M, Tomar VS, Dale E, Gomella LG, Solomides C, Kolesnikov O, Keith SW, Navarro HT, Dahlgren O, Chaga M, Trabulsi EJ. Targeting genomic receptors in voided urine for confirmation of benign prostatic hyperplasia. BJUI COMPASS 2024; 5:675-680. [PMID: 39022663 PMCID: PMC11250152 DOI: 10.1002/bco2.362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/27/2024] [Accepted: 03/18/2024] [Indexed: 07/20/2024] Open
Abstract
Objectives The objective of this study is to validate a hypothesis that a non-invasive optical imaging assay targeting genomic VPAC receptors on malignant cells shed in voided urine will represent either benign prostatic hyperplasia (BPH) or prostatic cancer (PCa). Risk for BPH in men 50-70 years old is 50-70% and PCa is 17%. BPH and PCa can coexist in 20% of men with BPH. Most commonly practiced methods to diagnose BPH do not distinguish BPH from PCa. Patients or Materials and Methods Males with BPH (N = 97, 60.8 ± 6.3 years, prostate-specific antigen 0.7 ± 0.4 ng/mL) and without oncologic disease (N = 35, 63.4 ± 5.8 years, prostate-specific antigen < 1.5 ng/mL) signed informed consent form and provided voided urine. Urine was cytocentrifuged, cells collected on glass slide, fixed, treated with VPAC specific fluorophore TP4303 (Kd 3.1 × 10-8M), washed, incubated with DAPI and observed using a fluorescence microscope. Cells with no VPAC did not fluoresce (BPH) and those with VPAC had red-orange fluorescence (PCa). Real-time polymerase chain reaction analyses for VPAC and NKX3.1 assay for cell origin were performed. Results Eighty-seven subjects were negative for VPAC expression. Positive VPAC expression was noted in 10 subjects. Patient chart review for clinical data on these 10 VPAC positive subjects showed five had nephrolithiasis, three had renal cysts, one had prostatitis and one was being treated with finasteride. Real-time polymerase chain reaction analysis-VPAC expressions for 7 normal and 12 BPH subjects were 1.31 ± 1.26 and 0.94 ± 0.89, respectively (P = 0.46). NKX3.1 showed cells of prostate origin for finasteride-treated patient. Specificity for VPAC urine assay for excluding prostate cancer in this BPH cohort was 88.5%, positive predictive value 0.00% and negative predictive value 100%. Conclusion VPAC assay may contribute extensively for BPH diagnosis and warrant continued investigation.
Collapse
Affiliation(s)
- Mathew Thakur
- Department of Radiology, Urology, Radiation Oncology and S. Kimmel Cancer CenterThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Vivek S. Tomar
- Department of RadiologyThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Emma Dale
- Department of RadiologyThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Leonard G. Gomella
- Department of Urology and S. Kimmel Cancer CenterThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Charalambos Solomides
- Department of Pathology and Genomic MedicineThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Oleksandr Kolesnikov
- Department of Urology and S. Kimmel Cancer CenterThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Scott W. Keith
- Division of Biostatistics and S. Kimmel Cancer CenterThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Hector T. Navarro
- Department of RadiologyThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Olivia Dahlgren
- Department of UrologyThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Michael Chaga
- Department of RadiologyThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Edouard J. Trabulsi
- Department of UrologyJefferson Einstein Medical Center and Thomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
30
|
Cardoso JCR, Mc Shane JC, Li Z, Peng M, Power DM. Revisiting the evolution of Family B1 GPCRs and ligands: Insights from mollusca. Mol Cell Endocrinol 2024; 586:112192. [PMID: 38408601 DOI: 10.1016/j.mce.2024.112192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 02/28/2024]
Abstract
Family B1 G protein-coupled receptors (GPCRs) are one of the most well studied neuropeptide receptor families since they play a central role in many biological processes including endocrine, gastrointestinal, cardiovascular and reproduction in animals. The genes for these receptors emerged from a common ancestral gene in bilaterian genomes and evolved via gene/genome duplications and deletions in vertebrate and invertebrate genomes. Their existence and function have mostly been characterized in vertebrates and few studies exist in invertebrate species. Recently, an increased interest in molluscs, means a series of genomes have become available, and since they are less modified than insect and nematode genomes, they are ideal to explore the origin and evolution of neuropeptide gene families. This review provides an overview of Family B1 GPCRs and their peptide ligands and incorporates new data obtained from Mollusca genomes and taking a comparative approach challenges existing models on their origin and evolution.
Collapse
Affiliation(s)
- João C R Cardoso
- Comparative Endocrinology and Integrative Biology, Centre of Marine Sciences, Universidade do Algarve, Campus de Gambelas, 8005-139, Faro, Portugal.
| | - Jennifer C Mc Shane
- Comparative Endocrinology and Integrative Biology, Centre of Marine Sciences, Universidade do Algarve, Campus de Gambelas, 8005-139, Faro, Portugal
| | - Zhi Li
- Comparative Endocrinology and Integrative Biology, Centre of Marine Sciences, Universidade do Algarve, Campus de Gambelas, 8005-139, Faro, Portugal
| | - Maoxiao Peng
- Comparative Endocrinology and Integrative Biology, Centre of Marine Sciences, Universidade do Algarve, Campus de Gambelas, 8005-139, Faro, Portugal
| | - Deborah M Power
- Comparative Endocrinology and Integrative Biology, Centre of Marine Sciences, Universidade do Algarve, Campus de Gambelas, 8005-139, Faro, Portugal; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China
| |
Collapse
|
31
|
Sallicandro L, Gliozheni E, Feudi D, Sabbatini P, Pellegrino RM, Alabed HBR, Baldini D, Gerli S, Alviggi C, Cascardi E, Cicinelli E, Malvasi A, Fioretti B. Increased Vasoactive Intestinal Peptide (VIP) in polycystic ovary syndrome patients undergoing IVF. Front Endocrinol (Lausanne) 2024; 15:1331282. [PMID: 38774232 PMCID: PMC11106456 DOI: 10.3389/fendo.2024.1331282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 02/28/2024] [Indexed: 05/24/2024] Open
Abstract
Introduction Polycystic ovary syndrome (PCOS) is a common multifactorial and polygenic disorder of the endocrine system, affecting up to 20% of women in reproductive age with a still unknown etiology. Follicular fluid (FF) represents an environment for the normal development of follicles rich in metabolites, hormones and neurotransmitters, but in some instances of PCOS the composition can be different. Vasoactive intestinal peptide (VIP) is an endogenous autonomic neuropeptide involved in follicular atresia, granulosa cell physiology and steroidogenesis. Methods ELISA assays were performed to measure VIP and estradiol levels in human follicular fluids, while AMH, FSH, LH, estradiol and progesterone in the plasma were quantified by chemiluminescence. UHPLC/QTOF was used to perform the untargeted metabolomic analysis. Results Our ELISA and metabolomic results show: i) an increased concentration of VIP in follicular fluid of PCOS patients (n=9) of about 30% with respect to control group (n=10) (132 ± 28 pg/ml versus 103 ± 26 pg/ml, p=0,03) in women undergoing in vitro fertilization (IVF), ii) a linear positive correlation (p=0.05, r=0.45) between VIP concentration and serum Anti-Müllerian Hormone (AMH) concentration and iii) a linear negative correlation between VIP and noradrenaline metabolism. No correlation between VIP and estradiol (E2) concentration in follicular fluid was found. A negative correlation was found between VIP and noradrenaline metabolite 3,4-dihydroxyphenylglycolaldehyde (DOPGAL) in follicular fluids. Conclusion VIP concentration in follicular fluids was increased in PCOS patients and a correlation was found with noradrenaline metabolism indicating a possible dysregulation of the sympathetic reflex in the ovarian follicles. The functional role of VIP as noradrenergic modulator in ovarian physiology and PCOS pathophysiology was discussed.
Collapse
Affiliation(s)
- Luana Sallicandro
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
- Department of Medicine and Surgery, Perugia Medical School, University of Perugia, Perugia, Italy
| | - Elko Gliozheni
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
- Department of Medicine and Surgery, Perugia Medical School, University of Perugia, Perugia, Italy
| | - Davide Feudi
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
| | - Paola Sabbatini
- Department of Medicine and Surgery, Perugia Medical School, University of Perugia, Perugia, Italy
| | | | - Husam B. R. Alabed
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
| | - Domenico Baldini
- In Vitro Fertilization (IVF) Center, Momo Fertilife, Bisceglie, Italy
| | - Sandro Gerli
- Department of Medicine and Surgery, Perugia Medical School, University of Perugia, Perugia, Italy
- Department of Obstetrics and Gynecology, Centre of Perinatal and Reproductive Medicine, University of Perugia, Perugia, Italy
| | - Carlo Alviggi
- Department of Clinical Gynecological Emergency, Obstetrics and Reproductive Medicine, University Federico II, Naples, Italy
| | - Eliano Cascardi
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Ettore Cicinelli
- Department of Interdisciplinary Medicine (DIM), University of Bari “Aldo Moro”, Bari, Italy
| | - Antonio Malvasi
- Department of Interdisciplinary Medicine (DIM), University of Bari “Aldo Moro”, Bari, Italy
| | - Bernard Fioretti
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
- Department of Obstetrics and Gynecology, Centre of Perinatal and Reproductive Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
32
|
Pellesi L, Ashina M, Martelletti P. Targeting the PACAP-38 pathway is an emerging therapeutic strategy for migraine prevention. Expert Opin Emerg Drugs 2024; 29:57-64. [PMID: 38337150 DOI: 10.1080/14728214.2024.2317778] [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: 01/13/2024] [Accepted: 02/08/2024] [Indexed: 02/12/2024]
Abstract
INTRODUCTION The pituitary adenylate cyclase-activating polypeptide-38 (PACAP-38) has emerged as a key mediator of migraine pathogenesis. PACAP-38 and its receptors are predominantly distributed in arteries, sensory and parasympathetic neurons of the trigeminovascular system. Phase 2 trials have tested human monoclonal antibodies designed to bind and inhibit PACAP-38 and the pituitary adenylate cyclase-activating polypeptide type I (PAC1) receptor for migraine prevention. AREAS COVERED This review focuses on the significance of the PACAP-38 pathway as a target in migraine prevention. English peer-reviewed articles were searched in PubMed, Scopus and ClinicalTrials.gov electronic databases. EXPERT OPINION A PAC1 receptor monoclonal antibody was not effective for preventing migraine in a proof-of-concept trial, paving the way for alternative strategies to be considered. Lu AG09222 is a humanized monoclonal antibody targeting PACAP-38 that was effective in preventing physiological responses of PACAP38 and reducing monthly migraine days in individuals with migraine. Further studies are necessary to elucidate the clinical utility, long-term safety and cost-effectiveness of therapies targeting the PACAP pathway.
Collapse
Affiliation(s)
- Lanfranco Pellesi
- Clinical Pharmacology, Pharmacy and Environmental Medicine, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Messoud Ashina
- Danish Headache Center, Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Paolo Martelletti
- School of Health Sciences, Unitelma Sapienza University of Rome, Rome, Italy
| |
Collapse
|
33
|
Asano S, Ono A, Baba K, Uehara T, Sakamoto K, Hayata-Takano A, Nakazawa T, Yanamoto S, Tanimoto K, Hashimoto H, Ago Y. Blockade of vasoactive intestinal peptide receptor 2 (VIPR2) signaling suppresses cyclin D1-dependent cell-cycle progression in MCF-7 cells. J Pharmacol Sci 2024; 154:139-147. [PMID: 38395514 DOI: 10.1016/j.jphs.2024.01.002] [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: 11/01/2023] [Revised: 12/28/2023] [Accepted: 01/05/2024] [Indexed: 02/25/2024] Open
Abstract
Vasoactive intestinal peptide (VIP) receptor 2 (VIPR2) is a G protein-coupled receptor that binds to Gαs, Gαi, and Gαq proteins to regulate various downstream signaling molecules, such as protein kinase A (PKA), phosphatidylinositol 3-kinase (PI3K), and phospholipase C. In this study, we examined the role of VIPR2 in cell cycle progression. KS-133, a newly developed VIPR2-selective antagonist peptide, attenuated VIP-induced cell proliferation in MCF-7 cells. The percentage of cells in the S-M phase was decreased in MCF-7 cells treated with KS-133. KS-133 in the presence of VIP decreased the phosphorylation of extracellular signal-regulated kinase (ERK), AKT, and glycogen synthase kinase-3β (GSK3β), resulting in a decrease in cyclin D1 levels. In MCF-7 cells stably-expressing VIPR2, KS-133 decreased PI3K activity and cAMP levels. Treatment with the ERK-specific kinase (MEK) inhibitor U0126 and the class I PI3K inhibitor ZSTK474 decreased the percentage of cells in the S phase. KS-133 reduced the percentage of cells in the S phase more than treatment with U0126 or ZSTK474 alone and did not affect the effect of the mixture of these inhibitors. Our findings suggest that VIPR2 signaling regulates cyclin D1 levels through the cAMP/PKA/ERK and PI3K/AKT/GSK3β pathways, and mediates the G1/S transition to control cell proliferation.
Collapse
Affiliation(s)
- Satoshi Asano
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan; School of Dentistry, Hiroshima University, Hiroshima, 734-8553, Japan.
| | - Ami Ono
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan; Department of Orthodontics and Craniofacial Developmental Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Kaede Baba
- School of Dentistry, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Teru Uehara
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan; Department of Oral Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Kotaro Sakamoto
- Research & Development Department, Ichimaru Pharcos Company Limited, 318-1 Asagi, Motosu, Gifu, 501-0475, Japan
| | - Atsuko Hayata-Takano
- Department of Pharmacology, Graduate School of Dentistry, Osaka University, Suita, Osaka, 565-0871, Japan; Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 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, University of Fukui, Osaka, 565-0871, Japan
| | - Takanobu Nakazawa
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan; Laboratory of Molecular Biology, Department of Bioscience, Graduate School of Life Sciences, Tokyo University of Agriculture, Tokyo, 156-8502, Japan
| | - Souichi Yanamoto
- School of Dentistry, Hiroshima University, Hiroshima, 734-8553, Japan; Department of Oral Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Kotaro Tanimoto
- School of Dentistry, Hiroshima University, Hiroshima, 734-8553, Japan; Department of Orthodontics and Craniofacial Developmental Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Hitoshi Hashimoto
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 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, University of Fukui, Osaka, 565-0871, Japan; Division of Bioscience, Institute for Datability Science, Osaka University, Osaka, 565-0871, Japan; Transdimensional Life Imaging Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, 565-0871, Japan; Department of Molecular Pharmaceutical Science, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
| | - Yukio Ago
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan; School of Dentistry, Hiroshima University, Hiroshima, 734-8553, Japan.
| |
Collapse
|
34
|
Akerman S, Goadsby PJ, Romero-Reyes M. PACAP-38 related modulation of the cranial parasympathetic projection: A novel mechanism and therapeutic target in severe primary headache. Br J Pharmacol 2024; 181:480-494. [PMID: 37706270 DOI: 10.1111/bph.16242] [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: 05/16/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND AND PURPOSE Little is known of how cranial autonomic symptoms (CAS) in cluster headache and migraine may contribute to their severe headache phenotype. This strong association suggests the involvement of the cranial parasympathetic efferent pathway. To investigate its contribution, we studied the role of pituitary adenylate cyclase activating polypeptide-38 (PACAP-38), a potent sensory and parasympathetic neuropeptide, in modulating pre- and post-ganglionic cranial parasympathetic projection neurons, and their influence on headache-related trigeminal-autonomic responses. EXPERIMENTAL APPROACH Using PACAP-38 and PACAP-38 responsive receptor antagonists, electrophysiological, behavioural and facial neurovascular-blood flow was measured in rats to probe trigeminal- and parasympathetic-neuronal, periorbital thresholds and cranial-autonomic outcomes, as they relate to primary headaches. KEY RESULTS Sumatriptan attenuated the development of PACAP-38 mediated activation and sensitization of trigeminocervical neurons and related periorbital allodynia. PACAP-38 also caused activation and enhanced responses of dural-responsive pre-ganglionic pontine-superior salivatory parasympathetic neurons. Further, the PACAP-38 responsive receptor antagonists dissected a role of VPAC1 and PAC1 receptors in attenuating cranial-autonomic and trigeminal-neuronal responses to activation of the cranial parasympathetic projection, which requires post-ganglionic parasympathetic neurotransmission. CONCLUSION AND IMPLICATIONS Given the prevailing view that sumatriptan acts to some degree via a peripheral mechanism, our data support that PACAP-38 mediated receptor activation modulates headache-related cranial-autonomic and trigeminovascular responses via peripheral and central components of the cranial parasympathetic projection. This provides a mechanistic rationale for the association of CAS with more severe headache phenotypes in cluster headache and migraine, and supports the cranial parasympathetic projection as a potential novel locus for treatment by selectively targeting PACAP-38 or PACAP-38 responsive VPAC1 /PAC1 receptors.
Collapse
Affiliation(s)
- Simon Akerman
- Department of Neural and Pain Sciences, University of Maryland Baltimore, Baltimore, Maryland, USA
| | - Peter J Goadsby
- Headache Group, Wolfson Sensory, Pain and Regeneration Research Centre (SPaRRC), Institute of Psychology, Psychiatry and Neuroscience, King's College London, London, UK
- Department of Neurology, University of California Los Angeles, Los Angeles, California, USA
| | - Marcela Romero-Reyes
- Department of Neural and Pain Sciences, University of Maryland Baltimore, Baltimore, Maryland, USA
| |
Collapse
|
35
|
Jia ZF, Wang JL, Pan W, Hu J. Croton tiglium L. seeds ameliorate loperamide-induced constipation via regulating gastrointestinal hormones and gut microbiota before and after processing. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117378. [PMID: 37923254 DOI: 10.1016/j.jep.2023.117378] [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: 05/16/2023] [Revised: 10/26/2023] [Accepted: 10/30/2023] [Indexed: 11/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Crotonis Fructus (CF), the seeds of Croton tiglium L., have been commonly used in the treatment of constipation for more than two thousand years in traditional Chinese medicine (TCM). CF needs to be processed before clinical use and Crotonis Semen Pulveratum (CP) is the processed cream of CF, which could reduce the drastic purgative action and gastrointestinal damages. However, the mechanism of CF and CP in the treatment of constipation is still unclear. AIM OF THE STUDY This study was to evaluate the effects of CF and CP on loperamide-induced constipation and the underlying mechanism. MATERIALS AND METHODS The chemical compositions of CF and CP were analyzed by UPLC-Q-TOF-MS. Constipated mouse model was established by loperamide (9.6 mg/kg, b.w., i.g.) for two weeks. After successful modeling, the mice were treated with CF or CP (45.5 and 136.5 mg/kg, b.w., i.g.) once a day for seven days. The physiological status, defecation indices, defecation time, and intestinal propulsion rate in mice were measured. Histopathologic examination and serum biochemical parameters were further estimated. 16S rDNA gene sequencing was carried out to characterize the effects of CF and CP on intestinal microbiome structure. Spearman correlation analysis was also performed to explore the association between gut microbiotic abundance and serum indices. RESULTS The results verified the therapeutic effects of CF and CP on loperamide-induced constipation. CF and CP could significantly ameliorate the reduction of fecal number, fecal weight, fecal water content, and intestinal propulsion rate in mice with constipation, and the first stool defecation time was also obviously reduced. Moreover, CF and CP could regulate the secretion of gastrointestinal hormones and inflammatory factors induced by constipation. Histopathologic examination showed that CP was superior to CF in relieving pathological injury and inflammatory cell infiltration. According to 16S rDNA sequencing, CF and CP treatment could improve gut microbiota disturbance in mice with constipation and the abundance of opportunistic pathogens such as Parabacteroides, Parasutterella and Bacillus remarkably declined, while the levels of beneficial bacterial such as Candidatus_Arthromitus significantly increased. Besides, CP may play a better role in correcting the intestinal flora disorder than CF, which was more obvious in the high-dose group. In addition, phytochemical analysis revealed the presence of diterpenoids and alkaloids in CF and CP. CONCLUSIONS CF and CP could ameliorate loperamide-induced constipation by regulating gastrointestinal hormones secretion, reducing the levels of inflammatory cytokines and improving the disturbance of gut microbiota. Moreover, CP was superior to CF in the enrichment of beneficial bacteria and reduction of harmful bacteria and histopathological damage induced by constipation, which may be related to the changes in the species and content of diterpenoids after processing. The study provides new evidence for the processing mechanism and clinical application of CF and CP.
Collapse
Affiliation(s)
- Ze-Fei Jia
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin, 301617, China
| | - Jia-Li Wang
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin, 301617, China
| | - Wen Pan
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin, 301617, China
| | - Jing Hu
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin, 301617, China.
| |
Collapse
|
36
|
Elhadi K, Daiwile AP, Cadet JL. Modeling methamphetamine use disorder and relapse in animals: short- and long-term epigenetic, transcriptional., and biochemical consequences in the rat brain. Neurosci Biobehav Rev 2023; 155:105440. [PMID: 38707245 PMCID: PMC11068368 DOI: 10.1016/j.neubiorev.2023.105440] [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: 07/24/2023] [Revised: 10/13/2023] [Accepted: 10/24/2023] [Indexed: 05/07/2024]
Abstract
Methamphetamine use disorder (MUD) is a neuropsychiatric disorder characterized by binge drug taking episodes, intervals of abstinence, and relapses to drug use even during treatment. MUD has been modeled in rodents and investigators are attempting to identify its molecular bases. Preclinical experiments have shown that different schedules of methamphetamine self-administration can cause diverse transcriptional changes in the dorsal striatum of Sprague-Dawley rats. In the present review, we present data on differentially expressed genes (DEGs) identified in the rat striatum following methamphetamine intake. These include genes involved in transcription regulation, potassium channel function, and neuroinflammation. We then use the striatal data to discuss the potential significance of the molecular changes induced by methamphetamine by reviewing concordant or discordant data from the literature. This review identified potential molecular targets for pharmacological interventions. Nevertheless, there is a need for more research on methamphetamine-induced transcriptional consequences in various brain regions. These data should provide a more detailed neuroanatomical map of methamphetamine-induced changes and should better inform therapeutic interventions against MUD.
Collapse
Affiliation(s)
- Khalid Elhadi
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, Baltimore, MD, 21224
| | - Atul P. Daiwile
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, Baltimore, MD, 21224
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, Baltimore, MD, 21224
| |
Collapse
|
37
|
Blümm C, Bonaterra GA, Schwarzbach H, Eiden LE, Weihe E, Kinscherf R. PAC1 deficiency reduces chondrogenesis in atherosclerotic lesions of hypercholesterolemic ApoE-deficient mice. BMC Cardiovasc Disord 2023; 23:566. [PMID: 37980508 PMCID: PMC10657554 DOI: 10.1186/s12872-023-03600-5] [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: 08/08/2023] [Accepted: 11/07/2023] [Indexed: 11/20/2023] Open
Abstract
BACKGROUND Induction of chondrogenesis is associated with progressive atherosclerosis. Deficiency of the ADCYAP1 gene encoding pituitary adenylate cyclase-activating peptide (PACAP) aggravates atherosclerosis in ApoE deficient (ApoE-/-) mice. PACAP signaling regulates chondrogenesis and osteogenesis during cartilage and bone development. Therefore, this study aimed to decipher whether PACAP signaling is related to atherogenesis-related chondrogenesis in the ApoE-/- mouse model of atherosclerosis and under the influence of a high-fat diet. METHODS For this purpose, PACAP-/-/ApoE-/-, PAC1-/-/ApoE-/-, and ApoE-/- mice, as well as wildtype (WT) mice, were studied under standard chow (SC) or cholesterol-enriched diet (CED) for 20 weeks. The amount of cartilage matrix in atherosclerotic lesions of the brachiocephalic trunk (BT) with maximal lumen stenosis was monitored by alcian blue and collagen II staining on deparaffinized cross sections. The chondrogenic RUNX family transcription factor 2 (RUNX2), macrophages [(MΦ), Iba1+], and smooth muscle cells (SMC, sm-α-actin) were immunohistochemically analyzed and quantified. RESULTS ApoE-/- mice fed either SC or CED revealed an increase of alcian blue-positive areas within the media compared to WT mice. PAC1-/-/ApoE-/- mice under CED showed a reduction in the alcian blue-positive plaque area in the BT compared to ApoE-/- mice. In contrast, PACAP deficiency in ApoE-/- mice did not affect the chondrogenic signature under either diet. CONCLUSIONS Our data show that PAC1 deficiency reduces chondrogenesis in atherosclerotic plaques exclusively under conditions of CED-induced hypercholesterolemia. We conclude that CED-related chondrogenesis occurs in atherosclerotic plaques via transdifferentiation of SMCs and MΦ, partly depending on PACAP signaling through PAC1. Thus, PAC1 antagonists or PACAP agonists may offer therapeutic potential against pathological chondrogenesis in atherosclerotic lesions generated under hypercholesterolemic conditions, especially in familial hypercholesterolemia. This discovery opens therapeutic perspectives to be used in the treatment against the progression of atherosclerosis.
Collapse
Affiliation(s)
- C Blümm
- Institute for Anatomy and Cell Biology, Department of Medical Cell Biology, University of Marburg, 35032, Marburg, Germany
| | - G A Bonaterra
- Institute for Anatomy and Cell Biology, Department of Medical Cell Biology, University of Marburg, 35032, Marburg, Germany.
| | - H Schwarzbach
- Institute for Anatomy and Cell Biology, Department of Medical Cell Biology, University of Marburg, 35032, Marburg, Germany
| | - L E Eiden
- Section on Molecular Neuroscience, Laboratory of Cellular and Molecular Regulation, National Institute of Mental Health Intramural Research Program, Bethesda, MD, 20814, USA
| | - E Weihe
- Institute for Anatomy and Cell Biology, Department of Medical Cell Biology, University of Marburg, 35032, Marburg, Germany
| | - R Kinscherf
- Institute for Anatomy and Cell Biology, Department of Medical Cell Biology, University of Marburg, 35032, Marburg, Germany
| |
Collapse
|
38
|
Gutierrez Cruz A, Aresta Branco MSL, Borhani Peikani M, Mutafova-Yambolieva VN. Differential Influences of Endogenous and Exogenous Sensory Neuropeptides on the ATP Metabolism by Soluble Ectonucleotidases in the Murine Bladder Lamina Propria. Int J Mol Sci 2023; 24:15650. [PMID: 37958631 PMCID: PMC10647406 DOI: 10.3390/ijms242115650] [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: 09/29/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
Bladder urothelium and suburothelium/lamina propria (LP) have prominent sensory and transducer functions with the active participation of afferent neurons and urothelium-derived purine mediators such as adenosine 5'-triphosphate (ATP), adenosine 5'-diphosphate (ADP), and adenosine (ADO). Effective concentrations of purines at receptor targets depend significantly on the extracellular degradation of ATP by ectonucleotidases (ENTDs). We recently reported the regulated release of soluble ENTDs (s-ENTDs) in the LP and the consequent degradation of ATP to ADP, AMP, and ADO. Afferent neurons in the LP can be activated by urothelial ATP and release peptides and other transmitters that can alter the activity of cells in their vicinity. Using a murine decentralized ex vivo detrusor-free bladder model, 1,N6-etheno-ATP (eATP) as substrate, and sensitive HPLC-FLD methodologies, we found that exogenous neuropeptides calcitonin gene-related peptide (CGRP), substance P (Sub P), neurokinin A (NKA), and pituitary adenylate cyclase-activating polypeptide [PACAP (1-38)] all increased the degradation of eATP by s-ENTDs that were released in the LP spontaneously and/or during bladder filling. Using antagonists of neuropeptide receptors, we observed that endogenous NKA did not modify the ATP hydrolysis by s-ENTDs, whereas endogenous Sub P increased both the constitutive and distention-induced release of s-ENTDs. In contrast, endogenous CGRP and PACAP (1-38) increased the distention-induced, but not the spontaneous, release of s-ENTDs. The present study puts forward the novel idea that interactions between peptidergic and purinergic signaling mechanisms in the LP have an impact on bladder excitability and functions by regulating the effective concentrations of adenine purines at effector cells in the LP.
Collapse
Affiliation(s)
| | | | | | - Violeta N. Mutafova-Yambolieva
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada Reno, Reno, NV 89557, USA; (A.G.C.); (M.B.P.)
| |
Collapse
|
39
|
Chen Q, Zhang XY, Wang YP, Fu YJ, Cao F, Xu YN, Kong JG, Tian NX, Xu Y, Wang Y. Unveiling adcyap1 as a protective factor linking pain and nerve regeneration through single-cell RNA sequencing of rat dorsal root ganglion neurons. BMC Biol 2023; 21:235. [PMID: 37880634 PMCID: PMC10601282 DOI: 10.1186/s12915-023-01742-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 10/17/2023] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND Severe peripheral nerve injury (PNI) often leads to significant movement disorders and intractable pain. Therefore, promoting nerve regeneration while avoiding neuropathic pain is crucial for the clinical treatment of PNI patients. However, established animal models for peripheral neuropathy fail to accurately recapitulate the clinical features of PNI. Additionally, researchers usually investigate neuropathic pain and axonal regeneration separately, leaving the intrinsic relationship between the development of neuropathic pain and nerve regeneration after PNI unclear. To explore the underlying connections between pain and regeneration after PNI and provide potential molecular targets, we performed single-cell RNA sequencing and functional verification in an established rat model, allowing simultaneous study of the neuropathic pain and axonal regeneration after PNI. RESULTS First, a novel rat model named spared nerve crush (SNC) was created. In this model, two branches of the sciatic nerve were crushed, but the epineurium remained unsevered. This model successfully recapitulated both neuropathic pain and axonal regeneration after PNI, allowing for the study of the intrinsic link between these two crucial biological processes. Dorsal root ganglions (DRGs) from SNC and naïve rats at various time points after SNC were collected for single-cell RNA sequencing (scRNA-seq). After matching all scRNA-seq data to the 7 known DRG types, we discovered that the PEP1 and PEP3 DRG neuron subtypes increased in crushed and uncrushed DRG separately after SNC. Using experimental design scRNA-seq processing (EDSSP), we identified Adcyap1 as a potential gene contributing to both pain and nerve regeneration. Indeed, repeated intrathecal administration of PACAP38 mitigated pain and facilitated axonal regeneration, while Adcyap1 siRNA or PACAP6-38, an antagonist of PAC1R (a receptor of PACAP38) led to both mechanical hyperalgesia and delayed DRG axon regeneration in SNC rats. Moreover, these effects can be reversed by repeated intrathecal administration of PACAP38 in the acute phase but not the late phase after PNI, resulting in alleviated pain and promoted axonal regeneration. CONCLUSIONS Our study reveals that Adcyap1 is an intrinsic protective factor linking neuropathic pain and axonal regeneration following PNI. This finding provides new potential targets and strategies for early therapeutic intervention of PNI.
Collapse
Affiliation(s)
- Qi Chen
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100083, China
| | - Xi-Yin Zhang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100083, China
| | - Yu-Pu Wang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100083, China
| | - Yun-Jie Fu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100083, China
| | - Feng Cao
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100083, China
| | - Yi-Nuo Xu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100083, China
| | - Jin-Ge Kong
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100083, China
| | - Na-Xi Tian
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100083, China
| | - Yu Xu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100083, China
| | - Yun Wang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100083, China.
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China.
| |
Collapse
|
40
|
Park H, Jeon H, Lee KJ, Kim CG, Shin D. Seaweed intake modulates the association between VIPR2 variants and the incidence of metabolic syndrome in middle-aged Koreans. Food Funct 2023; 14:9446-9456. [PMID: 37807848 DOI: 10.1039/d3fo02425c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Vasoactive intrinsic peptide receptor (VIPR2), a circadian gene, is involved in metabolic homeostasis and metabolic syndrome (MetS). Seaweeds contain polysaccharides that regulate metabolic homeostasis, possibly by altering the effects of VIPR2 variants. We examined the relationship between VIPR2 expression and the incidence of MetS based on seaweed consumption. This study included 4979 Koreans aged ≥40 years using data from the Ansan-Ansung cohort of the Korean Genome and Epidemiology Study. The total seaweeds included were laver, kelp, and sea mustard. A multivariable Cox proportional hazards model was used to analyze the interactions between the VIPR2 rs6950857 genotype associated with MetS incidence and seaweed intake after adjusting for covariates such as region. A total of 2134 patients with MetS were followed for an average of 8.9 years. In men with the GG genotype of rs6950857, the highest quintile of seaweed consumption was associated with a decreased incidence of MetS compared with that of the lowest quintile (hazard ratio, 0.78; 95% confidence interval, 0.62-0.98). We identified a unique association between the rs6950857 genotype, seaweed intake, and MetS. These findings highlight the importance of VIPR2 and the regulatory role of seaweed consumption in MetS incidence.
Collapse
Affiliation(s)
- Haeun Park
- Department of Food and Nutrition, Inha University, Incheon 22212, Republic of Korea.
| | - Hyunyu Jeon
- Department of Food and Nutrition, Inha University, Incheon 22212, Republic of Korea.
| | - Kyung Ju Lee
- Department of Women's Rehabilitation, National Rehabilitation Center, 58, Samgaksan-ro, Gangbuk-gu, Seoul 01022, Republic of Korea
| | - Choong-Gon Kim
- Ocean Climate Response & Ecosystem Research Department, Korea Institute of Ocean Science and Technology, 385, Haeyang-ro, Yeongdo-gu, Busan 49111, Republic of Korea
| | - Dayeon Shin
- Department of Food and Nutrition, Inha University, Incheon 22212, Republic of Korea.
| |
Collapse
|
41
|
Witzel R, Block A, Pollmann S, Oetzel L, Fleck F, Bonaterra GA, Kinscherf R, Schwarz A. PACAP regulates VPAC1 expression, inflammatory processes and lipid homeostasis in M1- and M2-macrophages. Front Cardiovasc Med 2023; 10:1264901. [PMID: 37900572 PMCID: PMC10611464 DOI: 10.3389/fcvm.2023.1264901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 09/12/2023] [Indexed: 10/31/2023] Open
Abstract
Background Pituitary adenylate cyclase-activating polypeptide (PACAP) acts as an anti-atherogenic neuropeptide and plays an important role in cytoprotective, as well as inflammatory processes, and cardiovascular regulation. Therefore, the aim of this study is to investigate the regulatory effects of PACAP and its receptor VPAC1 in relation to inflammatory processes and lipid homeostasis in different macrophage (MΦ) subtypes. Methods To investigate the role of PACAP deficiency in the pathogenesis of atherosclerosis under standard chow (SC) or cholesterol-enriched diet (CED) in vivo, PACAP-/- mice were crossbred with ApoE-/- to generate PACAP-/-/ApoE-/- mice. Lumen stenosis in the aortic arch and different MΦ-subtypes were analyzed in atherosclerotic plaques by quantitative immunohistochemistry. Undifferentiated bone marrow-derived cells (BMDC) from 30-weeks-old ApoE-/- and PACAP-/-/ApoE-/- mice were isolated, differentiated into BMDM1- and BMDM2-MΦ, and incubated with oxidized low-density lipoprotein (oxLDL). In addition, PMA-differentiated human THP-1 MΦ were further differentiated into M1-/M2-MΦ and subsequently treated with PACAP38, the VPAC1 agonist [(Ala11,22,28)VIP], the antagonist (PG 97-269), and/or oxLDL. Uptake/accumulation of oxLDL was analyzed by oxLDL-DyLight™488 and Bodipy™ 493/503. The mRNA expression was analyzed by qRT-PCR, protein levels by Western blot, and cytokine release by ELISA. Results In vivo, after 30 weeks of SC, PACAP-/-/ApoE-/- mice showed increased lumen stenosis compared with ApoE-/- mice. In atherosclerotic plaques of PACAP-/-/ApoE-/- mice under CED, immunoreactive areas of VPAC1, CD86, and CD163 were increased compared with ApoE-/- mice. In vitro, VPAC1 protein levels were increased in PACAP-/-/ApoE-/- BMDM compared with ApoE-/- BMDM, resulting in increased TNF-α mRNA expression in BMDM1-MΦ and decreased TNF-α release in BMDM2-MΦ. Concerning lipid homeostasis, PACAP deficiency decreased the area of lipid droplets in BMDM1-/M2-MΦ with concomitant increasing adipose differentiation-related protein level. In THP-1 M1-/M2-MΦ, the VPAC1 antagonist increased the uptake of oxLDL, whereas the VPAC1 agonist decreased the oxLDL-induced intracellular triglyceride content. Conclusion Our data suggest that PACAP via VPAC1 signaling plays an important regulatory role in inflammatory processes in atherosclerotic plaques and in lipid homeostasis in different MΦ-subtypes, thereby affecting foam cell formation. Therefore, VPAC1 agonists or PACAP may represent a new class of anti-atherogenic therapeutics.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Anja Schwarz
- Department of Medical Cell Biology, Institute of Anatomy and Cell Biology, Philipps-University of Marburg, Marburg, Germany
| |
Collapse
|
42
|
Cao Y, Chen H, Yang J. Neuroanatomy of lymphoid organs: Lessons learned from whole-tissue imaging studies. Eur J Immunol 2023; 53:e2250136. [PMID: 37377338 DOI: 10.1002/eji.202250136] [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: 12/16/2022] [Revised: 06/06/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023]
Abstract
Decades of extensive research have documented the presence of neural innervations of sensory, sympathetic, or parasympathetic origin in primary and secondary lymphoid organs. Such neural inputs can release neurotransmitters and neuropeptides to directly modulate the functions of various immune cells, which represents one of the essential aspects of the body's neuroimmune network. Notably, recent studies empowered by state-of-the-art imaging techniques have comprehensively assessed neural distribution patterns in BM, thymus, spleen, and LNs of rodents and humans, helping clarify several controversies lingering in the field. In addition, it has become evident that neural innervations in lymphoid organs are not static but undergo alterations in pathophysiological contexts. This review aims to update the current information on the neuroanatomy of lymphoid organs obtained through whole-tissue 3D imaging and genetic approaches, focusing on anatomical features that may designate the functional modulation of immune responses. Moreover, we discuss several critical questions that call for future research, which will advance our in-depth understanding of the importance and complexity of neural control of lymphoid organs.
Collapse
Affiliation(s)
- Ying Cao
- Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Hongjie Chen
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Jing Yang
- Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
- Shenzhen Bay Laboratory, Institute of Molecular Physiology, Shenzhen, China
| |
Collapse
|
43
|
Afonso-Oramas D, Santana-Cordón L, Lemus-Mesa A, Teixidó-Trujillo S, Rodríguez-Rodríguez AE, Cruz-Muros I, González-Gómez M, Barroso-Chinea P. Drastic decline in vasoactive intestinal peptide expression in the suprachiasmatic nucleus in obese mice on a long-term high-fat diet. Brain Res Bull 2023; 202:110756. [PMID: 37678442 DOI: 10.1016/j.brainresbull.2023.110756] [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: 07/20/2023] [Revised: 08/27/2023] [Accepted: 09/05/2023] [Indexed: 09/09/2023]
Abstract
The suprachiasmatic nucleus (SCN) is the main region for the regulation of circadian rhythms. Although the SCN contains a heterogeneous neurochemical phenotype with a wide variety of neuropeptides, a key role has been suggested for the vasoactive intestinal neuropeptide (VIP) as a modulator circadian, reproductive, and seasonal rhythms. VIP is a 28-amino acid polypeptide hormone that belongs to the secretin-glucagon peptide superfamily and shares 68 % homology with the pituitary adenylate cyclase-activating polypeptide (PACAP). VIP acts as an endogenous appetite inhibitor in the central nervous system, where it participates in the control of appetite and energy homeostasis. In recent years, significant efforts have been made to better understand the role of VIP in the regulation of appetite/satiety and energy balance. This study aimed to elucidate the long-term effect of an obesogenic diet on the distribution and expression pattern of VIP in the SCN and nucleus accumbens (NAc) of C57BL/6 mice. A total of 15 female C57BL/6J mice were used in this study. Female mice were fed ad libitum with water and, either a standard diet (SD) or a high-fat diet (HFD) to induce obesity. There were 7 female mice on the SD and 8 on the HFD. The duration of the experiment was 365 days. The morphological study was performed using immunohistochemistry and double immunofluorescence techniques to study the neurochemical profile of VIP neurons of the SCN of C57BL/6 mice. Our data show that HFD-fed mice gained weight and showed reduced VIP expression in neurons of the SCN and also in fibres located in the NAc. Moreover, we observed a loss of neuropeptide Y (NPY) expression in fibres surrounding the SCN. Our findings on VIP may contribute to the understanding of the pathophysiological mechanisms underlying obesity in regions associated with uncontrolled intake of high-fat foods and the reward system, thus facilitating the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Domingo Afonso-Oramas
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain; Instituto de Tecnologías Biomédicas de Canarias (ITB), Universidad de La Laguna, Tenerife, Spain; Instituto Universitario de Neurociencias. Universidad de La Laguna, Tenerife, Spain.
| | - Laura Santana-Cordón
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain
| | - Alejandro Lemus-Mesa
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain
| | - Silvia Teixidó-Trujillo
- Departamento de Medicina Interna, Dermatología y Psiquiatría. Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain
| | | | - Ignacio Cruz-Muros
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain; Instituto de Tecnologías Biomédicas de Canarias (ITB), Universidad de La Laguna, Tenerife, Spain
| | - Miriam González-Gómez
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain; Instituto de Tecnologías Biomédicas de Canarias (ITB), Universidad de La Laguna, Tenerife, Spain; Instituto Universitario de Neurociencias. Universidad de La Laguna, Tenerife, Spain
| | - Pedro Barroso-Chinea
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain; Instituto de Tecnologías Biomédicas de Canarias (ITB), Universidad de La Laguna, Tenerife, Spain; Instituto Universitario de Neurociencias. Universidad de La Laguna, Tenerife, Spain.
| |
Collapse
|
44
|
Rees TA, Labastida-Ramírez A, Rubio-Beltrán E. Calcitonin/PAC 1 receptor splice variants: a blind spot in migraine research. Trends Pharmacol Sci 2023; 44:651-663. [PMID: 37543479 PMCID: PMC10529278 DOI: 10.1016/j.tips.2023.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/08/2023] [Accepted: 07/08/2023] [Indexed: 08/07/2023]
Abstract
The neuropeptides calcitonin gene-related peptide (CGRP) and pituitary adenylate cyclase-activating polypeptide (PACAP) and their receptors are linked to migraine neurobiology. Recent antimigraine therapeutics targeting the signaling of these neuropeptides are effective; however, some patients respond suboptimally, indicating an incomplete understanding of migraine pathophysiology. The CGRP- and PACAP-responsive receptors can be differentially spliced. It is known that receptor splice variants can have different pathophysiological effects in other receptor-mediated pain pathways. Despite considerable knowledge on the structural and pharmacological differences of the CGRP- and PACAP-responsive receptor splice variants and their expression in migraine-relevant tissues, their role in migraine is rarely considered. Here we shine a spotlight on the calcitonin and PACAP (PAC1) receptor splice variants and examine what implications they may have for drug activity and design.
Collapse
Affiliation(s)
- Tayla A Rees
- School of Biological Sciences, University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand.
| | - Alejandro Labastida-Ramírez
- Headache Group, Wolfson Center for Age Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Eloisa Rubio-Beltrán
- Headache Group, Wolfson Center for Age Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
45
|
Langer G, Scott J, Lind C, Otto C, Bothe U, Laux-Biehlmann A, Müller J, le Roy B, Irlbacher H, Nowak-Reppel K, Schlüter A, Davenport AJ, Slack M, Bäurle S. Discovery and In Vitro Characterization of BAY 2686013, an Allosteric Small Molecule Antagonist of the Human Pituitary Adenylate Cyclase-Activating Polypeptide Receptor. Mol Pharmacol 2023; 104:105-114. [PMID: 37348913 DOI: 10.1124/molpharm.122.000662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 05/04/2023] [Accepted: 05/12/2023] [Indexed: 06/24/2023] Open
Abstract
The human pituitary adenylate cyclase-activating polypeptide receptor (hPAC1-R), a class B G-protein-coupled receptor (GPCR) identified almost 30 years ago, represents an important pharmacological target in the areas of neuroscience, oncology, and immunology. Despite interest in this target, only a very limited number of small molecule modulators have been reported for this receptor. We herein describe the results of a drug discovery program aiming for the identification of a potent and selective hPAC1-R antagonist. An initial high-throughput screening (HTS) screen of 3.05 million compounds originating from the Bayer screening library failed to identify any tractable hits. A second, completely revised screen using native human embryonic kidney (HEK)293 cells yielded a small number of hits exhibiting antagonistic properties (4.2 million compounds screened). BAY 2686013 (1) emerged as a promising compound showing selective antagonistic activity in the submicromolar potency range. In-depth characterization supported the hypothesis that BAY 2686013 blocks receptor activity in a noncompetitive manner. Preclinical, pharmacokinetic profiling indicates that BAY 2686013 is a valuable tool compound for better understanding the signaling and function of hPAC1-R. SIGNIFICANCE STATEMENT: Although the human pituitary adenylate cyclase-activating polypeptide receptor (hPAC1-R) is of major significance as a therapeutic target with a well documented role in pain signaling, only a very limited number of small-molecule (SMOL) compounds are known to modulate its activity. We identified and thoroughly characterized a novel, potent, and selective SMOL antagonist of hPAC1-R (acting in an allosteric manner). These characteristics make BAY 2686013 an ideal tool for further studies.
Collapse
Affiliation(s)
- Gernot Langer
- Bayer AG, Research & Development, Pharmaceuticals, Berlin, Germany (G.L., U.B., J.M., B.l.R., S.B.); Bayer AG, Research & Development, Pharmaceuticals, Wuppertal, Germany (C.O., A.L.-B.); Innovation Campus Berlin, a Nuvisan Company, Berlin, Germany (H.I., K.N.-R.); Evotec SE, Hamburg, Germany (A.S., M.S.); and Evotec (UK) Ltd, Abingdon, Oxfordshire, United Kingdom (J.S., C.L., A.J.D.)
| | - John Scott
- Bayer AG, Research & Development, Pharmaceuticals, Berlin, Germany (G.L., U.B., J.M., B.l.R., S.B.); Bayer AG, Research & Development, Pharmaceuticals, Wuppertal, Germany (C.O., A.L.-B.); Innovation Campus Berlin, a Nuvisan Company, Berlin, Germany (H.I., K.N.-R.); Evotec SE, Hamburg, Germany (A.S., M.S.); and Evotec (UK) Ltd, Abingdon, Oxfordshire, United Kingdom (J.S., C.L., A.J.D.)
| | - Christoffer Lind
- Bayer AG, Research & Development, Pharmaceuticals, Berlin, Germany (G.L., U.B., J.M., B.l.R., S.B.); Bayer AG, Research & Development, Pharmaceuticals, Wuppertal, Germany (C.O., A.L.-B.); Innovation Campus Berlin, a Nuvisan Company, Berlin, Germany (H.I., K.N.-R.); Evotec SE, Hamburg, Germany (A.S., M.S.); and Evotec (UK) Ltd, Abingdon, Oxfordshire, United Kingdom (J.S., C.L., A.J.D.)
| | - Christiane Otto
- Bayer AG, Research & Development, Pharmaceuticals, Berlin, Germany (G.L., U.B., J.M., B.l.R., S.B.); Bayer AG, Research & Development, Pharmaceuticals, Wuppertal, Germany (C.O., A.L.-B.); Innovation Campus Berlin, a Nuvisan Company, Berlin, Germany (H.I., K.N.-R.); Evotec SE, Hamburg, Germany (A.S., M.S.); and Evotec (UK) Ltd, Abingdon, Oxfordshire, United Kingdom (J.S., C.L., A.J.D.)
| | - Ulrich Bothe
- Bayer AG, Research & Development, Pharmaceuticals, Berlin, Germany (G.L., U.B., J.M., B.l.R., S.B.); Bayer AG, Research & Development, Pharmaceuticals, Wuppertal, Germany (C.O., A.L.-B.); Innovation Campus Berlin, a Nuvisan Company, Berlin, Germany (H.I., K.N.-R.); Evotec SE, Hamburg, Germany (A.S., M.S.); and Evotec (UK) Ltd, Abingdon, Oxfordshire, United Kingdom (J.S., C.L., A.J.D.)
| | - Alexis Laux-Biehlmann
- Bayer AG, Research & Development, Pharmaceuticals, Berlin, Germany (G.L., U.B., J.M., B.l.R., S.B.); Bayer AG, Research & Development, Pharmaceuticals, Wuppertal, Germany (C.O., A.L.-B.); Innovation Campus Berlin, a Nuvisan Company, Berlin, Germany (H.I., K.N.-R.); Evotec SE, Hamburg, Germany (A.S., M.S.); and Evotec (UK) Ltd, Abingdon, Oxfordshire, United Kingdom (J.S., C.L., A.J.D.)
| | - Jörg Müller
- Bayer AG, Research & Development, Pharmaceuticals, Berlin, Germany (G.L., U.B., J.M., B.l.R., S.B.); Bayer AG, Research & Development, Pharmaceuticals, Wuppertal, Germany (C.O., A.L.-B.); Innovation Campus Berlin, a Nuvisan Company, Berlin, Germany (H.I., K.N.-R.); Evotec SE, Hamburg, Germany (A.S., M.S.); and Evotec (UK) Ltd, Abingdon, Oxfordshire, United Kingdom (J.S., C.L., A.J.D.)
| | - Beau le Roy
- Bayer AG, Research & Development, Pharmaceuticals, Berlin, Germany (G.L., U.B., J.M., B.l.R., S.B.); Bayer AG, Research & Development, Pharmaceuticals, Wuppertal, Germany (C.O., A.L.-B.); Innovation Campus Berlin, a Nuvisan Company, Berlin, Germany (H.I., K.N.-R.); Evotec SE, Hamburg, Germany (A.S., M.S.); and Evotec (UK) Ltd, Abingdon, Oxfordshire, United Kingdom (J.S., C.L., A.J.D.)
| | - Horst Irlbacher
- Bayer AG, Research & Development, Pharmaceuticals, Berlin, Germany (G.L., U.B., J.M., B.l.R., S.B.); Bayer AG, Research & Development, Pharmaceuticals, Wuppertal, Germany (C.O., A.L.-B.); Innovation Campus Berlin, a Nuvisan Company, Berlin, Germany (H.I., K.N.-R.); Evotec SE, Hamburg, Germany (A.S., M.S.); and Evotec (UK) Ltd, Abingdon, Oxfordshire, United Kingdom (J.S., C.L., A.J.D.)
| | - Katrin Nowak-Reppel
- Bayer AG, Research & Development, Pharmaceuticals, Berlin, Germany (G.L., U.B., J.M., B.l.R., S.B.); Bayer AG, Research & Development, Pharmaceuticals, Wuppertal, Germany (C.O., A.L.-B.); Innovation Campus Berlin, a Nuvisan Company, Berlin, Germany (H.I., K.N.-R.); Evotec SE, Hamburg, Germany (A.S., M.S.); and Evotec (UK) Ltd, Abingdon, Oxfordshire, United Kingdom (J.S., C.L., A.J.D.)
| | - Anne Schlüter
- Bayer AG, Research & Development, Pharmaceuticals, Berlin, Germany (G.L., U.B., J.M., B.l.R., S.B.); Bayer AG, Research & Development, Pharmaceuticals, Wuppertal, Germany (C.O., A.L.-B.); Innovation Campus Berlin, a Nuvisan Company, Berlin, Germany (H.I., K.N.-R.); Evotec SE, Hamburg, Germany (A.S., M.S.); and Evotec (UK) Ltd, Abingdon, Oxfordshire, United Kingdom (J.S., C.L., A.J.D.)
| | - Adam J Davenport
- Bayer AG, Research & Development, Pharmaceuticals, Berlin, Germany (G.L., U.B., J.M., B.l.R., S.B.); Bayer AG, Research & Development, Pharmaceuticals, Wuppertal, Germany (C.O., A.L.-B.); Innovation Campus Berlin, a Nuvisan Company, Berlin, Germany (H.I., K.N.-R.); Evotec SE, Hamburg, Germany (A.S., M.S.); and Evotec (UK) Ltd, Abingdon, Oxfordshire, United Kingdom (J.S., C.L., A.J.D.)
| | - Mark Slack
- Bayer AG, Research & Development, Pharmaceuticals, Berlin, Germany (G.L., U.B., J.M., B.l.R., S.B.); Bayer AG, Research & Development, Pharmaceuticals, Wuppertal, Germany (C.O., A.L.-B.); Innovation Campus Berlin, a Nuvisan Company, Berlin, Germany (H.I., K.N.-R.); Evotec SE, Hamburg, Germany (A.S., M.S.); and Evotec (UK) Ltd, Abingdon, Oxfordshire, United Kingdom (J.S., C.L., A.J.D.)
| | - Stefan Bäurle
- Bayer AG, Research & Development, Pharmaceuticals, Berlin, Germany (G.L., U.B., J.M., B.l.R., S.B.); Bayer AG, Research & Development, Pharmaceuticals, Wuppertal, Germany (C.O., A.L.-B.); Innovation Campus Berlin, a Nuvisan Company, Berlin, Germany (H.I., K.N.-R.); Evotec SE, Hamburg, Germany (A.S., M.S.); and Evotec (UK) Ltd, Abingdon, Oxfordshire, United Kingdom (J.S., C.L., A.J.D.)
| |
Collapse
|
46
|
Rajbhandari AK, Barson JR, Gilmartin MR, Hammack SE, Chen BK. The functional heterogeneity of PACAP: Stress, learning, and pathology. Neurobiol Learn Mem 2023; 203:107792. [PMID: 37369343 PMCID: PMC10527199 DOI: 10.1016/j.nlm.2023.107792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 06/29/2023]
Abstract
Pituitary adenylate cyclase-activating peptide (PACAP) is a highly conserved and widely expressed neuropeptide that has emerged as a key regulator of multiple neural and behavioral processes. PACAP systems, including the various PACAP receptor subtypes, have been implicated in neural circuits of learning and memory, stress, emotion, feeding, and pain. Dysregulation within these PACAP systems may play key roles in the etiology of pathological states associated with these circuits, and PACAP function has been implicated in stress-related psychopathology, feeding and metabolic disorders, and migraine. Accordingly, central PACAP systems may represent important therapeutic targets; however, substantial heterogeneity in PACAP systems related to the distribution of multiple PACAP isoforms across multiple brain regions, as well as multiple receptor subtypes with several isoforms, signaling pathways, and brain distributions, provides both challenges and opportunities for the development of new clinically-relevant strategies to target the PACAP system in health and disease. Here we review the heterogeneity of central PACAP systems, as well as the data implicating PACAP systems in clinically-relevant behavioral processes, with a particular focus on the considerable evidence implicating a role of PACAP in stress responding and learning and memory. We also review data suggesting that there are sex differences in PACAP function and its interactions with sex hormones. Finally, we discuss both the challenges and promise of harnessing the PACAP system in the development of new therapeutic avenues and highlight PACAP systems for their critical role in health and disease.
Collapse
Affiliation(s)
| | - Jessica R Barson
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Marieke R Gilmartin
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, United States
| | - Sayamwong E Hammack
- Department of Psychological Science, University of Vermont, 2 Colchester Avenue, Burlington, VT, United States
| | - Briana K Chen
- Division of Systems Neuroscience, Research Foundation for Mental Hygiene, Inc. (RFMH) / New York State Psychiatric Institute (NYSPI), New York, NY, United States; Department of Psychiatry, Columbia University Irving Medical Center (CUIMC), New York, NY, United States.
| |
Collapse
|
47
|
Chuang HK, Hsieh AR, Ang TY, Chen SW, Yang YP, Huang HJ, Chiou SH, Lin TC, Chen SJ, Hsu CC, Hwang DK. TMEM132D and VIPR2 Polymorphisms as Genetic Risk Loci for Retinal Detachment: A Genome-Wide Association Study and Polygenic Risk Score Analysis. Invest Ophthalmol Vis Sci 2023; 64:17. [PMID: 37695605 PMCID: PMC10501492 DOI: 10.1167/iovs.64.12.17] [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: 02/14/2023] [Accepted: 07/12/2023] [Indexed: 09/12/2023] Open
Abstract
Purpose Retinal detachment (RD) is a sight-threatening ocular disease caused by separation of the neurosensory retina from the underlying retinal pigment epithelium layer. Its genetic basis is unclear because of a limited amount of data. In this study, we aimed to identify genetic risk loci associated with RD in participants without diabetes mellitus and to construct a polygenic risk score (PRS) to predict the risk of RD. Methods A genome-wide association study was conducted using data from the Taiwan Biobank to identify RD risk loci. A total of 1533 RD cases and 106,270 controls were recruited, all of whom were Han Chinese. Replication studies were performed using data from the UK Biobank and Biobank Japan. To construct the PRS, a traditional clumping and thresholding method was performed and validated by fivefold cross-validation. Results Two novel loci with significant associations were identified. These two genes were TMEM132D (lead single nucleotide polymorphism [SNP]: rs264498, adjusted-P = 7.18 × 10-9) and VIPR2 (lead SNP: rs3812305, adjusted-P = 8.38 × 10-9). The developed PRS was effective in discriminating individuals at high risk of RD with a dose-response relationship. The quartile with the highest risk had an odds ratio of 1244.748 compared to the lowest risk group (95% confidence interval, 175.174-8844.892). Conclusions TMEM132D and VIPR2 polymorphisms are genetic candidates linked to RD in Han Chinese populations. Our proposed PRS was effective at discriminating high-risk from low-risk individuals.
Collapse
Affiliation(s)
- Hao-Kai Chuang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ai-Ru Hsieh
- Department of Statistics, Tamkang University, New Taipei City, Taiwan
| | - Tien-Yap Ang
- Department of Statistics, Tamkang University, New Taipei City, Taiwan
| | - Szu-Wen Chen
- Department of Statistics, Tamkang University, New Taipei City, Taiwan
| | - Yi-Ping Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hung-Juei Huang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of General Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tai-Chi Lin
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shih-Jen Chen
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chih-Chien Hsu
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - De-Kuang Hwang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
48
|
Pandher PK, Rahim Y, Timms KP, Filatov E, Short LI, Gray SL. Reference gene recommendations and PACAP receptor expression in murine sympathetic ganglia of the autonomic nervous system that innervate adipose tissues after chronic cold exposure. J Neuroendocrinol 2023; 35:e13313. [PMID: 37404042 DOI: 10.1111/jne.13313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 06/01/2023] [Accepted: 06/04/2023] [Indexed: 07/06/2023]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is an important regulator of the stress response in mammals, influencing both the hypothalamic-pituitary-adrenal (HPA) axis and the sympathetic nervous system (SNS). PACAP has been reported to influence energy homeostasis, including adaptive thermogenesis, an energy burning process in adipose tissue regulated by the SNS in response to cold stress and overfeeding. While research suggests PACAP acts centrally at the level of the hypothalamus, knowledge of PACAP's role within the sympathetic nerves innervating adipose tissues in response to metabolic stressors is limited. This work shows, for the first time, gene expression of PACAP receptors in stellate ganglia and highlights some differential expression with housing temperature. Additionally, we present our dissection protocol, analysis of tyrosine hydroxylase gene expression as a molecular biomarker for catecholamine producing tissue and recommend three stable reference genes for the normalization of quantitative real time-polymerase chain reaction (qRT-PCR) data when working with this tissue. This study adds to information about neuropeptide receptor expression in peripheral ganglia of the sympathetic nervous system innervating adipose tissue and provides insight into PACAP's role in the regulation of energy metabolism.
Collapse
Affiliation(s)
- Parleen K Pandher
- Northern Medical Program, Division of Medical Sciences, University of Northern British Columbia, Prince George, British Columbia, Canada
| | - Yamna Rahim
- Northern Medical Program, Division of Medical Sciences, University of Northern British Columbia, Prince George, British Columbia, Canada
| | - Katherine P Timms
- Northern Medical Program, Division of Medical Sciences, University of Northern British Columbia, Prince George, British Columbia, Canada
| | - Ekaterina Filatov
- Northern Medical Program, Division of Medical Sciences, University of Northern British Columbia, Prince George, British Columbia, Canada
| | - Landon I Short
- Northern Medical Program, Division of Medical Sciences, University of Northern British Columbia, Prince George, British Columbia, Canada
| | - Sarah L Gray
- Northern Medical Program, Division of Medical Sciences, University of Northern British Columbia, Prince George, British Columbia, Canada
| |
Collapse
|
49
|
Silvestro M, Iannone LF, Orologio I, Tessitore A, Tedeschi G, Geppetti P, Russo A. Migraine Treatment: Towards New Pharmacological Targets. Int J Mol Sci 2023; 24:12268. [PMID: 37569648 PMCID: PMC10418850 DOI: 10.3390/ijms241512268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/28/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
Migraine is a debilitating neurological condition affecting millions of people worldwide. Until a few years ago, preventive migraine treatments were based on molecules with pleiotropic targets, developed for other indications, and discovered by serendipity to be effective in migraine prevention, although often burdened by tolerability issues leading to low adherence. However, the progresses in unravelling the migraine pathophysiology allowed identifying novel putative targets as calcitonin gene-related peptide (CGRP). Nevertheless, despite the revolution brought by CGRP monoclonal antibodies and gepants, a significant percentage of patients still remains burdened by an unsatisfactory response, suggesting that other pathways may play a critical role, with an extent of involvement varying among different migraine patients. Specifically, neuropeptides of the CGRP family, such as adrenomedullin and amylin; molecules of the secretin family, such as pituitary adenylate cyclase-activating peptide (PACAP) and vasoactive intestinal peptide (VIP); receptors, such as transient receptor potential (TRP) channels; intracellular downstream determinants, such as potassium channels, but also the opioid system and the purinergic pathway, have been suggested to be involved in migraine pathophysiology. The present review provides an overview of these pathways, highlighting, based on preclinical and clinical evidence, as well as provocative studies, their potential role as future targets for migraine preventive treatment.
Collapse
Affiliation(s)
- Marcello Silvestro
- Headache Centre, Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (I.O.); (A.T.); (G.T.)
- Advanced MRI Neuroimaging Centre, Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Luigi Francesco Iannone
- Headache Centre and Clinical Pharmacology Unit, Careggi University Hospital Florence, 50134 Florence, Italy; (L.F.I.); (P.G.)
| | - Ilaria Orologio
- Headache Centre, Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (I.O.); (A.T.); (G.T.)
| | - Alessandro Tessitore
- Headache Centre, Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (I.O.); (A.T.); (G.T.)
- Advanced MRI Neuroimaging Centre, Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Gioacchino Tedeschi
- Headache Centre, Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (I.O.); (A.T.); (G.T.)
- Advanced MRI Neuroimaging Centre, Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Pierangelo Geppetti
- Headache Centre and Clinical Pharmacology Unit, Careggi University Hospital Florence, 50134 Florence, Italy; (L.F.I.); (P.G.)
| | - Antonio Russo
- Advanced MRI Neuroimaging Centre, Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| |
Collapse
|
50
|
Sbei S, Moncrief T, Limjunyawong N, Zeng Y, Green DP. PACAP activates MRGPRX2 on meningeal mast cells to drive migraine-like pain. Sci Rep 2023; 13:12302. [PMID: 37516794 PMCID: PMC10387048 DOI: 10.1038/s41598-023-39571-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 07/27/2023] [Indexed: 07/31/2023] Open
Abstract
Migraine ranks among the most prevalent disorders worldwide, leading to disability and decreased quality of life in patients. Recently, neurogenic inflammation has been recognized as a potential underlying pathology contributing to the migraine pain pathway. Mast cells reside in the meninges and have been implicated in contributing to the pathophysiology of migraine. Here we report for the first time that the mouse Mas-Related G-protein-coupled Receptor B2 (MrgprB2), is expressed on meningeal connective tissue mast cells and contributes to Pituitary Adenylate Cyclase Activating Peptide (PACAP)-induced migraine-like pain behavior. We also found that PACAP was able to dose-dependently lead to enzyme release from human mast cells via activation of MRGPRX2; the human homolog of MrgprB2. Using a transgenic MRGPRX2 mouse, we observed significant increases in PACAP-induced migraine-like pain behavior in MRGPRX2+ mice vs mice lacking the receptor. These results reveal both MrgprB2 and MRGPRX2 as important contributors to neuropeptide-induced migraine pain.
Collapse
Affiliation(s)
- Sami Sbei
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Taylor Moncrief
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Nathachit Limjunyawong
- Center of Research Excellence in Allergy and Immunology, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Yaping Zeng
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Dustin P Green
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|