Next Article in Journal
Sustained Nrf2 Overexpression-Induced Metabolic Deregulation Can Be Attenuated by Modulating Insulin/Insulin-like Growth Factor Signaling
Next Article in Special Issue
Glycerol Trinitrate Acts Downstream of Calcitonin Gene-Related Peptide in Trigeminal Nociception—Evidence from Rodent Experiments with Anti-CGRP Antibody Fremanezumab
Previous Article in Journal
Mechanisms of Endothelial Cell Membrane Repair: Progress and Perspectives
Previous Article in Special Issue
Monoclonal Antibodies against Calcitonin Gene-Related Peptide for Migraine Prophylaxis: A Systematic Review of Real-World Data
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

From CGRP to PACAP, VIP, and Beyond: Unraveling the Next Chapters in Migraine Treatment

by
Masaru Tanaka
1,
Ágnes Szabó
2,3,
Tamás Körtési
1,4,5,
Délia Szok
2,
János Tajti
2 and
László Vécsei
1,2,*
1
HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Danube Neuroscience Research Laboratory, Tisza Lajos krt. 113, H-6725 Szeged, Hungary
2
Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
3
Doctoral School of Clinical Medicine, University of Szeged, Korányi fasor 6, H-6720 Szeged, Hungary
4
Faculty of Health Sciences and Social Studies, University of Szeged, Temesvári krt. 31, H-6726 Szeged, Hungary
5
Preventive Health Sciences Research Group, Incubation Competence Centre of the Centre of Excellence for Interdisciplinary Research, Development and Innovation of the University of Szeged, H-6720 Szeged, Hungary
*
Author to whom correspondence should be addressed.
Cells 2023, 12(22), 2649; https://doi.org/10.3390/cells12222649
Submission received: 5 September 2023 / Revised: 13 November 2023 / Accepted: 14 November 2023 / Published: 17 November 2023
(This article belongs to the Special Issue Migraine Neuroscience: From Experimental Models to Target Therapy)

Abstract

:
Migraine is a neurovascular disorder that can be debilitating for individuals and society. Current research focuses on finding effective analgesics and management strategies for migraines by targeting specific receptors and neuropeptides. Nonetheless, newly approved calcitonin gene-related peptide (CGRP) monoclonal antibodies (mAbs) have a 50% responder rate ranging from 27 to 71.0%, whereas CGRP receptor inhibitors have a 50% responder rate ranging from 56 to 71%. To address the need for novel therapeutic targets, researchers are exploring the potential of another secretin family peptide, pituitary adenylate cyclase-activating polypeptide (PACAP), as a ground-breaking treatment avenue for migraine. Preclinical models have revealed how PACAP affects the trigeminal system, which is implicated in headache disorders. Clinical studies have demonstrated the significance of PACAP in migraine pathophysiology; however, a few clinical trials remain inconclusive: the pituitary adenylate cyclase-activating peptide 1 receptor mAb, AMG 301 showed no benefit for migraine prevention, while the PACAP ligand mAb, Lu AG09222 significantly reduced the number of monthly migraine days over placebo in a phase 2 clinical trial. Meanwhile, another secretin family peptide vasoactive intestinal peptide (VIP) is gaining interest as a potential new target. In light of recent advances in PACAP research, we emphasize the potential of PACAP as a promising target for migraine treatment, highlighting the significance of exploring PACAP as a member of the antimigraine armamentarium, especially for patients who do not respond to or contraindicated to anti-CGRP therapies. By updating our knowledge of PACAP and its unique contribution to migraine pathophysiology, we can pave the way for reinforcing PACAP and other secretin peptides, including VIP, as a novel treatment option for migraines.

1. Introduction

Migraines are neurological disorders causing recurrent, severe headaches and other symptoms like sensitivity to light, sound, smell, or touch, and nausea or vomiting [1]. Their cause is unclear but involves genetic, environmental, and lifestyle factors [2,3,4,5]. Triggers vary among individuals and include stress, hormonal changes, certain diets, and sleep disturbances [6,7,8,9]. Identifying and managing triggers can be crucial in preventing the onset of migraine attacks and reducing their frequency, duration, and severity [10]. Migraines are complex neurological disorders that can significantly impact individuals’ quality of life [11,12]. Comprehending the distinct stages, symptoms, triggers, and treatment options is fundamental for healthcare professionals and researchers, as it facilitates enhanced management and support for individuals affected by migraines [10].
Neuropeptides like calcitonin gene-related peptide (CGRP), pituitary adenylate cyclase-activating polypeptide (PACAP), vasoactive intestinal polypeptide (VIP), islet amyloid polypeptide (IAPP)/amylin, substance P, and adrenomedullin (ADM) [13,14,15,16,17,18]. The secretin family of peptides, including CGRP, PACAP, ADM, and amylin, control G protein-coupled receptors (GPCR) activity. They share homology, receptor cross-reactivity, and similar biological actions, suggesting they belong to this family (Figure 1) [19]. These neuropeptides play diverse roles in migraine pathogenesis, contributing to our understanding of the disorder’s mechanisms [20,21].
CGRP and PACAP, two neuropeptides, are released during migraine and cluster headache attacks, acting as potent vasodilators that trigger migraine-like symptoms [22,23,24]. Their expression increases when the trigeminovascular system is activated, contributing to pain signal transmission and the development of mechanical hyperalgesia [25,26,27,28]. Despite their similar functions, PACAP and CGRP likely have distinct roles in causing migraine-like symptoms. In rodent models, their pathways seem to operate independently; therefore, elevated levels of these substances in peripheral blood during migraine attacks may serve as prospective biomarkers [29,30,31,32,33]. Different PACAP variants also contribute uniquely to migraine development [34,35,36,37]. The neuropeptide VIP, found in the trigeminal nerve, plays a key role in the progression of migraines development. It dilates blood vessels during attacks, influences neurotransmitter release, regulates inflammation and immune responses, and may affect migraine intensity and frequency by modulating pain signal sensitivity [38,39,40,41,42].
New drugs targeting the CGRP signaling pathway have been developed for migraine treatment and prevention. These include monoclonal antibodies (mAbs) directed at either CGRP ligand or receptor and CGRP receptor inhibitors [43,44,45,46,47]. While promising, these treatments have some downsides [48,49]. They have been shown to reduce the frequency of migraine attacks: according to reports from double-blind placebo-controlled clinical trials and open-label trials, the ≥50% responder rate for CGRP mAbs ranges from 27 to 62%, or 44.5 to 71.0%, respectively [50,51]. For CGRP receptor inhibitors, the ≥50% responder rate ranges from 56 to 61%, or 44.5 to 71.0%, in double-blind placebo-controlled clinical trials and open-label trials, respectively [52,53]. The relatively higher effective rates of open-label trials compared to double-blind placebo-controlled clinical trials could be attributed to the possibility that placebo plays a role in real life. Nevertheless, the responder rate varies depending on the type of and the duration of treatment, the response criteria, and the patient characteristics. Additionally, these drugs can be costly with limited insurance coverage [54]. While generally well-tolerated, CGRP-targeting mAbs can cause gastrointestinal disorders like constipation, and gepants can cause fatigue, nausea, dizziness, tiredness, and dry mouth [55].
Humans typically experience migraines, but preclinical research using animal models reveals the interaction of genetic and environmental factors contributing to neurological disorders like migraines [56,57,58,59,60,61,62,63,64,65,66,67]. These models simulate disease conditions, aiding in identifying pathogenic processes, evaluating symptoms and comorbidities, and discovering interventions [68,69,70,71,72,73,74]. The integration of preclinical and clinical research contributes to innovative therapeutics and personalized medicine [75,76,77,78]. This review discusses the pathogenesis of migraines and the need for new treatment targets. It highlights the potential of secretin family peptides’ ligands and receptors as novel targets. The importance of further research into the roles of PACAP and VIP in migraine pathophysiology is emphasized, along with the development of targeted therapies. The review also considers the pituitary adenylate cyclase-activating peptide 1 receptor and other emerging therapeutic targets, such as PACAP1–38. It explores the similarities between PACAP and VIP, which are involved in sleep regulation and circadian rhythm, suggesting their key roles in migraines.

2. Pituitary Adenylate Cyclase-Activating Peptide and Vasoactive Intestinal Peptide

PACAP is a multi-functional peptide that has therapeutic potential in a variety of pathophysiological conditions and represents a promising avenue for intervention. PACAP is a neuropeptide that plays a crucial role in both neural and endocrine functions [78]. This peptide is widely distributed throughout the body and is involved in diverse physiological processes, including circadian rhythm and immune system regulations, modulation of pain perception, and stress response [79]. PACAP also has neuroprotective effects and has been shown to support nerve cell survival and regeneration in various neurological disorders [80]. GPCRs control the signaling pathways and cause the activation of adenylate cyclase (AC), the release of cyclic AMP, and the activation of protein kinase A (PKA) and calcium channels [81,82]. PACAP is a multi-functional peptide that has therapeutic potential in a variety of pathophysiological conditions and represents a promising avenue for therapeutic intervention [83].

2.1. Background

PACAP was found in ovine hypothalamic extracts in 1989. It is a 38-amino acid peptide hormone that stimulates AC activity in the pituitary gland [84]. Subsequently, it was found to be widely distributed in the central and peripheral nervous systems, as well as in non-neural tissues, including the adrenal gland, pancreas, gut, and reproductive system [85]. PACAP exists in three biologically active forms: PACAP1–38, 6–38-amino acid form of PACAP (PACAP6–38), and PACAP1–27 [86]. PACAP-related peptide (PRP) is also a member of the PACAP family [87]. Radioimmunoassay demonstrated that PACAP1–38 levels were approximately 60 times greater than PACAP1–27 levels and 10 times greater than PRP levels [88].
Since its discovery, PACAP has been extensively studied for its potent neuroprotective effects against a diverse range of neurological disorders, including stroke, traumatic brain injury, Parkinson’s disease, and Alzheimer’s disease [89,90]. Recent findings suggest that PACAP may also play a key role in the regulation of immune cell function and cytokine production, highlighting its potential as a therapeutic target for immune-mediated diseases such as rheumatoid arthritis, multiple sclerosis, and asthma [91]. Furthermore, PACAP has been implicated in the regulation of energy metabolism, making it a promising therapeutic agent for the treatment of metabolic disorders such as obesity and diabetes [92]. Overall, the growing body of evidence on the multifunctional properties of PACAP highlights its potential as a novel therapeutic target for a wide range of diseases.
VIP is a 28-amino acid polypeptide that was first characterized in 1970. It is secreted by cells throughout the intestinal tract and is widespread in many internal organs and systems [93]. VIP plays important roles in many biological functions, such as stimulation of contractility in the heart, vasodilation, promoting neuroendocrine–immune communication, lowering arterial blood pressure, and anti-inflammatory and immune-modulatory activity [94]. VIP stimulates the secretion of electrolytes and water by the intestinal mucosa and acts as a neurotransmitter, inducing a relaxation effect in some tissues [95]. VIP is also involved in the pathophysiology of various diseases, including osteoarthritis, cancer, and autoimmune disorders [94]. Furthermore, VIP is implicated in the physiological and pathophysiological roles of migraine [96]. In this context, VIP has been studied for its potential therapeutic applications.

2.2. Receptor and Signaling Mechanisms of PACAP and VIP

PACAP plays an important role in a wide range of biological processes such as feeding behavior, stress response, neuroprotection, and regulation of neurotransmitter release. It activates three different GPCRs named PAC1, vasoactive intestinal peptide receptor (VPAC) 1, and VPAC2; these receptors are widely expressed in the central and peripheral nervous systems, endocrine systems, and immune systems [97]. The binding of PACAP to these receptors leads to the activation of multiple signaling mechanisms (Table 1) [98].
Activation of the PAC1 receptor by PACAP leads to the activation of the adenylyl cyclase enzyme, which in turn leads to the production of cyclic adenosine monophosphate (cAMP) and the activation of PKA [99]. It also triggers the activation of phospholipase C, which leads to the breakdown of phosphatidyl inositol 4,5-bisphosphate (PIP2) into inositol triphosphate and diacylglycerol (DAG), which activates protein kinase C (PKC) [100]. On the other hand, VPAC1 and VPAC2 receptor activation leads to AC enzyme activation, which leads to the generation of cAMP and the activation of PKA [101]. Also, PACAP signaling turns on calcium signaling, which causes intracellular calcium to be released and calcium/calmodulin-dependent kinase II to be activated [102]. PACAP signaling also activates the mitogen-activated protein kinase (MAPK), extracellular signal-regulated kinase (ERK), and jun N-terminal kinase signaling pathways [103]. These signaling mechanisms contribute to the diverse biological effects of PACAP on cellular functions. The regulation of PACAP gene expression is presented in Figure 2.
PACAP and VIP are neuropeptides that interact specifically with three receptors (VPAC1, VPAC2, and PAC1) from the class II B GPCR family [104]. The similarities between PACAP and VIP in receptor and signaling mechanisms include the following: PACAP and VIP share nearly 70% amino acid sequence identity; PACAP binds with high affinity to all three receptors, while VIP binds with high affinity to VPAC1 and VPAC2 receptors and has a thousand fold lower affinity for the PAC1 receptor compared to PACAP; both PACAP and VIP receptors are preferentially coupled to Gαs, leading to activation of AC, subsequent cAMP production, and activation of PKA; and PKA may in turn activate ERKs, PACAP and VIP receptor-mediated signaling pathways [105,106,107,108]. Due to the wide distribution of VIP and PACAP receptors in the body, potential therapeutic applications of drugs targeting these receptors, as well as expected unwanted side effects, are numerous [109]. Designing selective therapeutics targeting these receptors remains challenging due to their structural similarities.

2.3. Role of PACAP and VIP in Migraine

PACAP has been strongly associated with the pathophysiology of migraine. PACAP is found in high levels in the trigeminal nerve, which is known to play a critical role in this condition. PACAP is known to increase the sensitivity of the trigeminal nerve, cause dilation of blood vessels in the brain, and trigger inflammation. All these biological effects have been implicated in the development of migraine attacks [110]. Several studies have been conducted to investigate the role of PACAP in migraine. One study showed that PACAP levels in the blood are significantly higher in migraine patients during an attack compared to headache-free controls [111]. This study suggests that PACAP could be used as a potential biomarker for migraine. Another study demonstrated that the venous infusion of PACAP into migraine patients resulted in the development of migraine-like attacks [112]. This finding strongly supports the hypothesis that PACAP plays a crucial role in the pathophysiology of migraine and suggests that blocking PACAP could be a potential therapeutic target for the treatment of migraines. The role of PACAP in migraine pathology is well established, and there is strong evidence that this neuropeptide plays a crucial role in the development of migraine attacks. Further research is needed to better understand the mechanism of action of PACAP and to develop new pharmacological agents that target PACAP for the treatment of migraines.
Both CGRP and PACAP are multifunctional peptides with many roles in the nervous, cardiovascular, respiratory, gastrointestinal, and reproductive systems. They play a role in vasodilation, neurogenic inflammation, and nociception. While CGRP plays an integral role in migraine, PACAP is likely to play a similar but distinct role as CGRP based on similarities and differences observed in both clinical and preclinical studies [113]. In rodent models, the PACAP pathway appears to be independent of the CGRP pathway, suggesting that CGRP and PACAP act in parallel ways that cause a migraine-like symptom [114]. In migraine without aura, the first double-blinded placebo-controlled study reported that 33% of the patients developed delayed migraine attacks after CGRP administration [115]. The studies have identified the involvement of two endogenous neuropeptides, CGRP and PACAP, in the pathogenesis of migraines [116].
VIP has also been implicated in the pathophysiology of migraine [117]. The similarities between PACAP and VIP in their roles in pathogenesis include the following: PACAP and VIP are released in conjunction with migraine and cluster headache attacks [118]; PACAP and VIP are potent vasodilators and can cause migraine-like attacks when infused into people [119]; a 2-h infusion of VIP caused migraine attacks, indicating that VIP plays a significant role in pathophysiology and intravenous administration of PACAP-38 caused headaches in all healthy subjects and migraine-like attacks in 58% of patients with a history of migraine without aura [15,35]; PACAP and VIP receptors are preferentially coupled to Gαs, leading to activation of AC, subsequent cAMP production, and activation of PKA [120]; PKA may in turn activate ERKs [121]; PACAP and VIP receptor-mediated signaling pathways are shown to share activities, including vasodilation, neurogenic inflammation, and nociception in rodents [122]; PACAP and VIP receptors provide a rich set of targets to complement and augment the current CGRP-based migraine therapeutics; VPAC1 receptors play a dominant role in PACAP-induced vasorelaxation in female mice [123]. Also, PG 99-465, a selective VPAC2 receptor antagonist that has been used in a number of physiological studies, has been shown to have significant activity at VPAC1 and PAC1 receptors [124].

2.4. Preclinical Studies

In addition to in vitro systems, a variety of organisms are used in experimental medicine [125,126,127]. Understanding the effects of endogenous neuropeptides, neurohormones, and metabolites has advanced significantly thanks to the information gathered using laboratory animals [128,129,130,131,132,133]. Animal models are a crucial tool for bridging the knowledge gap between data- and hypothesis-driven benchwork and its application to clinical bedside management. PACAP has been extensively studied as a neuromodulator in the trigeminal nociceptive pathway [134]. Preclinical studies have shown that PACAP is involved in the transmission of pain signals from the periphery to the central nervous system and is therefore a potential target for the treatment of migraine and other headache disorders [135,136].
In animal models, PACAP has been shown to play a role in trigeminal sensitization, which is the process by which nociceptive signals become amplified and persistent, leading to chronic pain [137]. Studies have also found that PACAP is involved in the activation of inflammatory pathways in the trigeminal nerve, further contributing to pain and inflammation [138]. In addition, PACAP has been implicated in the regulation of blood flow to the brain, which may also play a role in headache pathophysiology [139] and other neurological [26] or neuropsychological conditions [88]. In an experimental model of migraine, intraperitoneal administration of nitroglycerol caused marked photophobia and meningeal vasodilatation, and increased the number of c-fos-positive activated neurons in the TNC in wild-type mice but not in PACAP1–38-deficient mice [140]. In line with this, an increased concentration of PACAP1–38 was detected in the TNC after the activation of the TS in different animal models [141,142].
PAC1 receptor antagonists include PACAP6–38, N-stearyl-[Nle17] neurotensin-(6-11)/VIP-(7-28), deletion mutants of maxadilan, M65, and Max.d.4, and synthesized small-molecule acyl hydrazides, including PG 97-269 [143]. PACAP6–38 has been used as a PAC1 receptor antagonist in many studies, but it has an affinity for VPAC2 receptors [144]. N-stearyl-[Nle17] neurotensin-(6-11)/VIP-(7-28) (SNV) is a chimeric peptide analog that antagonizes the VIP2/PACAP receptor subclass. SNV is a better mitogen for the keratinocytic cell line and can increase AC activity in rat brain membranes 100 times more than VIP1-28 [145,146]. No migraine-related studies have been documented. The maxadilan is a vasodilator peptide derived from the salivary glands of sandflies. Its deletion mutants, M65 and Max.d.4, have been reported to be selective PAC1 receptor antagonists but have not been extensively used due to problems of availability [147,148]. PG 97-269 is a selective VPAC1 receptor antagonist with negligible affinity for the PACAP1 receptor. It did not stimulate AC activity but inhibited competitively the effect of VIP on AC activity in cells expressing the VIP1 receptor [146]. VIP and PACAP-induced vasodilation were partially blocked by PG 97-269, indicating that PACAP and VIP may play a role in migraine pathophysiology and that PG 97-269 may have therapeutic potential for migraine [149] (Table 2). Thus, preclinical studies suggest that concentrating on the PACAP signaling pathways in the trigeminal nociceptive system could be an effective strategy for discovering novel treatments for headache disorders. However, more research is needed to fully understand the mechanisms underlying PACAPs’ role in headache pathophysiology and to develop effective and safe PACAP-targeted therapies.
VIP plays a key role in sensory processing and the modulation of pain pathways in the trigeminal system. In preclinical studies, VIP has been shown to change the activity of nociceptive neurons in the trigeminal ganglion and make the TNC more sensitive, which can cause chronic pain or migraines [150]. In response to noxious stimuli, the trigeminal sensory neurons release VIP. This can activate VIP receptors on nearby neurons and cause the release of a number of signaling molecules involved in pain amplification [151]. VIP-mediated sensitization of trigeminal neurons can lead to hyperexcitability and increased responsiveness to noxious stimuli, which may contribute to the development and maintenance of chronic pain or migraine [152]. Targeting VIP signaling pathways may therefore represent a promising approach for the development of novel therapies for chronic pain or migraine.

2.5. Clinical Studies

A growing body of clinical research suggests that PACAP plays an important role in migraine pathophysiology. Patients with migraines exhibit higher levels of PACAP compared to control groups [153]. PACAP is a neuropeptide recognized for its involvement in the activation of nociceptive pathways, contributing to the development of migraines. The high levels of PACAP in migraineurs have been associated with increased headache severity and frequency, and this has led to the exploration of PACAP as a therapeutic target for treatment [154]. In migraineurs without aura, the development of PACAP1–38-evoked migraine-like attacks was independent of the severity of family load [35,155]. In the same study, 90 min after the injection, the levels of numerous migraine-related molecular markers were increased in the plasma of patients [156]. Magnetic resonance imaging angiography examinations revealed that PACAP1–38-induced headache was associated with prolonged vasodilatation of the middle meningeal artery (MMA) but not the middle cerebral artery (MCA). Sumatriptan, an antimigraine medication, was able to alleviate the headache, which mirrored the contraction of the MMA but not the MCA, indicating that PACAP1–38-induced headaches may originate from extracerebral arteries [157].
An increasing number of clinical studies have shown that targeting PACAP signaling may be a promising therapeutic strategy for migraine treatment. In terms of safety, PACAP has been generally well tolerated in clinical trials [158]. One study found that PACAP induces headaches via sustained vasodilation and that targeting the PACAP pathway may be a promising approach for treatment [159]. AMG 301, a mAb that targets the PAC1 receptor, was administered to patients with episodic or chronic migraines in a randomized, double-blind, placebo-controlled phase 2 study. There was no significant difference between the AMG 301 group and the placebo group, suggesting that AMG 301 was ineffective for prevention [160,161]. On the other hand, the PACAP ligand mAb, Lu AG09222, was shown to reduce the number of monthly migraine days from baseline to weeks 1–4 of treatment statistically significantly more than placebo [162,163]. Additionally, the mAb targeting the PAC1 receptor, LY3451838, is currently undergoing phase 2 clinical trials for adults with treatment-resistant migraine. This trial is in progress, and the results are not yet available [164] (Table 3). Overall, the efficacy and safety of PACAP as a migraine treatment in clinical studies suggest that it is a promising option for patients with this debilitating condition. Further research is needed to fully understand the potential of PACAP as a treatment for migraines, but the current evidence is encouraging.
VIP infusion has been studied in the context of migraines, with a particular focus on its potential to provoke migraine attacks and its role in pathophysiology. A phase 2 clinical trial investigated the effects of a long-lasting infusion of VIP on headaches, cranial hemodynamics, and autonomic symptoms in episodic migraine patients without aura [165]. The study found that a 2-h infusion of VIP promoted long-lasting cranial vasodilation and delayed headaches in healthy volunteers, resembling the effect of prophylaxis. However, other studies have suggested that VIP infusions may actually provoke migraine attacks. For example, a randomized clinical trial found that a 2-h infusion of VIP caused migraine episodes, suggesting an important role of VIP in migraine pathophysiology [15]. It remains unclear whether the lack of migraine induction can be attributed to the only transient vasodilatory response after a 20-min infusion of VIP. Overall, the search results suggest that VIP infusion may have a role in migraine pathophysiology, but further research is needed to fully understand its effects and potential therapeutic applications.

3. Discussion

This review paper aims to provide insights into the roles of PACAP in migraine by comparing its actions with those of VIP. By analyzing existing studies, this paper hopes to shed light on the pathophysiology of migraines and pave the way towards more effective treatments. The ultimate goal of this review is to explore the potential of developing antimigraine drugs that target the PACAP pathways. Identifying and producing new ways to target the PACAP system may provide an alternative therapeutic option for migraineurs. The authors aim to consolidate the current evidence on the PACAP system’s role in migraines and evaluate potential drug targets within the pathway, hoping to pave the way for more extensive research to develop new and effective antimigraine drugs that target the PACAP pathways.
The PACAP system presents a significant challenge when it comes to targeted therapies due to its pleiotropic roles in the body, both physiologically and pathologically [78,79,80,81,82]. PACAP plays crucial roles in various aspects of the body, such as neural development, pain regulation, immune functions, and stress responses. These diverse roles make the PACAP system difficult to target effectively without affecting other physiological functions. Furthermore, PACAP signaling is often dysregulated in pathological conditions such as inflammatory disorders, neurodegenerative diseases, and cancers [91,92]. Conversely, PACAP has been shown to have protective effects in certain diseases, such as ischemic stroke and Alzheimer’s disease [89,90]. Thus, finding a balance between targeting the PACAP system to treat diseases while preserving its physiological functions remains a significant challenge in the field of medicine.
The PACAP system has emerged as a potential target for the treatment of migraines, especially after the discovery of the role of CGRP and its receptors in pathophysiology [110,111,112]. PACAP is a peptide that belongs to the family of CGRP peptides and is highly expressed in the TS. The TS is the neural network that causes migraine pain [137,141,142]. PACAP receptors have been found to be co-localized with CGRP receptors in the TS, suggesting that the two systems could be acting in a synergistic manner to induce migraine pain [113,114,115]. Therefore, targeting the PACAP system could provide an additional therapeutic approach for the treatment of migraine, and several drugs that inhibit PACAP or its receptors are currently under development.
The present review holds notable significance in shedding light on the critical role of PACAP in comparison with other neuropeptides like CGRP and VIP, which have been extensively studied as potential therapeutic targets for various neurological disorders. The differences in symptomatic manifestation observed in preclinical studies of CGRP, PACAP, and VIP are most likely due to their distinct roles in migraine physiology and pathophysiology [105,106,107,108,109,113,114,115]. Thus, elucidating the mechanisms of those neuropeptides may not only lead to a better understanding of the etiology of migraine but may also provide a variety of therapeutic targets, potentially supplying a more diverse palette of antimigraine regimens [150]. By thoroughly analyzing the preclinical studies, the review highlights the promising findings that suggest the potential translation of PACAP’s therapeutic benefits from laboratory settings to clinical practice. The authors’ critical evaluation and systematic compilation of the latest research on PACAP is bound to have a relevant impact on the scientific community and serve as a foundation for further clinical research. Ultimately, the knowledge and insights gained from this review will be instrumental in developing advanced treatments for a range of debilitating neurological conditions.
The difference between those two clinical outcomes of PACAP mAbs could be explained by the fact that mAbs are designed to target specific receptors or ligands with high selectivity. The difference in how mAbs target receptors or ligands can result in different outcomes due to a variety of factors. Initially, mAbs can bind to various receptors or ligands in a variety of ways, which can alter their efficacy and the biological effects that follow [166]. Secondly, mAbs can have a variety of mechanisms of action when interacting with their targets, such as inhibiting cell surface receptors or promoting target cell death [167]. Thirdly, biological and clinical activities can vary greatly depending on the target and antibody design. This includes differences in the efficacy of the treatment, the occurrence of adverse effects, and the overall health of the patient [166]. Fourthly, mAbs exhibit exceptional target selectivity, with the choice of target influencing the antibody’s specificity and safety profile. When mAbs interact with their targets, they can perform a variety of actions, such as inhibiting the action of other molecules, killing cells, or altering the immune system’s function [166,167]. The choice of target and antibody design is crucial in determining the therapeutic effectiveness of mAbs.
The review also highlights limitations and challenges in PACAP research, such as the complexity of its signaling mechanism, variations in its effects on different cell types, and the limited availability of specific antibodies against PACAP and its receptors. The high cost of producing PACAP analogs and the lack of standardized protocols for their synthesis and purification are also limitations. The scarcity of studies on PACAP and VIP is also a major challenge for this field. It is difficult to establish a general agreement on the preclinical results and their relevance for human trials. Meta-analyses could be helpful in this regard, but they require more studies to be published. Therefore, more clinical investigations are necessary to gather evidence and, hopefully, derive conclusions from the clinical research. These challenges and limitations make it difficult to fully understand the mechanisms of PACAP action and to develop effective therapeutic interventions.
The development of PACAP-based therapeutics for migraines will focus on two main approaches: targeting PACAP ligands and receptors. Studies using animal models of migraines have demonstrated that blocking the PACAP receptor reduces symptoms while inhibiting PACAP signaling reduces pain sensitivity. Currently, clinical trials are underway to assess the safety and effectiveness of various PACAP-based drugs for migraines in humans. PACAP-based therapies may offer an alternative to current treatments by targeting the underlying mechanisms of the disorder and reducing the risk of side effects. In addition, the role of additional secretin family peptides, ADM, and amylin in the pathogenesis of migraine remains to be investigated. Further research in this area could lead to the development of better treatments for migraines.
The future direction of migraine research holds great promise for advancing our understanding of this complex neurological disorder. The combination of preclinical and clinical data, along with computational tools, has provided invaluable insights into various aspects of diseases, including neurological and psychiatric disorders [168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183,184,185,186,187,188,189]. The use of preclinical models and clinical studies has shed light on the underlying mechanisms of migraine. These studies have contributed to the identification of structural and functional changes in the brain that occur in neurological and psychiatric disorders, such as migraine attacks, as well as conditions like depression and other mental health problems [190,191,192,193,194,195,196,197,198,199,200,201,202,203,204,205,206]. Understanding these changes is crucial for identifying biomarkers, developing targeted treatments, and improving diagnosis [207,208,209].
Migraine is not just a pain disorder, but it is also interrelated to emotional and cognitive domains [210]. This condition is commonly linked with a broad range of psychiatric comorbidities, especially among subjects with migraine with aura or chronic migraine [211]. The comorbidity between neurological and psychiatric disorders likely suggests multiple causes, such as unidirectional causal explanations or shared environmental and/or genetic risk factors, communication with other parts of the body, and their interaction on multiple levels [212,213,214,215,216,217,218,219,220,221,222,223,224,225,226]. Emotional distress is commonly recognized as a migraine trigger, and being affected by psychiatric disorders is considered an independent modifiable factor of progression toward chronification of migraine and a tendency to overuse medication [227]. Therefore, revealing the mechanisms of comorbidity between migraine and psychiatric disorders may lead to a clue to prevention and management. Many biological and neural aspects of the comorbidity need to be clarified in order to better understand the true nature of the migraine–psychiatric disorder association.
The integration of computational tools in migraine research has allowed for the testing and evaluation of potential treatments. These tools enable researchers to simulate the effects of different interventions, including brain stimulation, and assess their therapeutic efficacy [228,229,230,231,232]. This approach holds promise for the development of novel and more effective treatments. Advanced imaging techniques have played a crucial role in migraine research. Neuroimaging studies have revealed structural and functional brain changes associated with migraine [233,234,235,236,237,238,239,240]. These imaging techniques provide valuable insights into the pathophysiology of the disorder and can help identify unique clinical cases. The use of human brain organoids in migraine research is an emerging area of study. Brain organoids are three-dimensional models that mimic the structure and function of the human brain. They can be used to investigate altered neuronal pathways, protein expression, and metabolic pathways associated with migraines [241,242,243,244]. This approach offers a unique opportunity to study the disease in a more physiologically relevant system.

4. Conclusions

PACAP is a neuropeptide that has been linked to the pathophysiology of primary headaches such as migraine. The release of PACAP is associated with this condition and cluster headache attacks, and it has been shown to be a potent vasodilator that dilates cranial arteries and causes migraines when infused into patients. Like CGRP, PACAP is located near sensory nerve fibers and has nociceptive functions. Both peptides are promising targets for migraine therapeutics, and growing evidence supports the involvement of PACAP-related mechanisms in migraines. While CGRP and PACAP share similar functions, the PACAP pathway appears to be independent of the CGRP pathway, suggesting that they act in parallel ways to cause a migraine-like symptom. Therefore, a better understanding of the role of PACAP and other secretin family peptides, including VIP, in migraine pathogenesis could lead to new treatment options for this debilitating condition.

Author Contributions

Conceptualization, M.T. and L.V.; writing—original draft preparation, M.T.; writing—review and editing, M.T., Á.S., T.K., D.S., J.T. and L.V; visualization, Á.S. and T.K.; supervision, J.T. and L.V.; project administration, L.V.; funding acquisition, L.V. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by OTKA-138125-K, TUDFO/47138-1/2019-ITM, and the HUN-REN Hungarian Research Network.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ACadenylate cyclase
ADMadrenomedullin
cAMPcyclic adenosine monophosphate
CGRPcalcitonin gene-related peptide
DAGdiacylglycerol
ERKextracellular signal-regulated kinase
GPCRG protein-coupled receptors
IAPPislet amyloid polypeptide/amylin
mAbsmonoclonal antibodies
MAPKmitogen-activated protein kinase
MCAmiddle cerebral artery
MMAmiddle meningeal artery
MEK: mitogen-activated protein kinase kinase
PACAPpituitary adenylate cyclase-activating polypeptide
PACAP1–3838-amino acid form of PACAP
PACAP1–2727-amino acid form of PACAP
PACAP6–386–38-amino acid form of PACAP
PIP2phosphatidyl inositol 4,5-bisphosphate
PKAprotein kinase A
PKCactivates protein kinase C
PRPPACAP-related peptide
SNVN-stearyl-[Nle17] neurotensin-(6–11)/VIP-(7–28)
TNCtrigeminal nucleus caudalis
TStrigeminovascular system
VPACvasoactive intestinal peptide receptor
VIPvasoactive intestinal polypeptide

References

  1. IHS Classification ICHD-3. Available online: https://ichd-3.org/1-migraine/1-2-migraine-with-aura/#:~:text=Description%3A,headache%20and%20associated%20migraine%20symptoms (accessed on 12 October 2023).
  2. Amiri, P.; Kazeminasab, S.; Nejadghaderi, S.A.; Mohammadinasab, R.; Pourfathi, H.; Araj-Khodaei, M.; Sullman, M.J.M.; Kolahi, A.A.; Safiri, S. Migraine: A Review on Its History, Global Epidemiology, Risk Factors, and Comorbidities. Front. Neurol. 2022, 12, 800605. [Google Scholar] [CrossRef] [PubMed]
  3. Ferrari, M.D.; Goadsby, P.J.; Burstein, R.; Kurth, T.; Ayata, C.; Charles, A.; Ashina, M.; van den Maagdenberg, A.M.J.M.; Dodick, D.W. Migraine. Nat. Rev. Dis. Primers 2022, 8, 2. [Google Scholar] [CrossRef]
  4. Ashina, M. Migraine. N. Engl. J. Med. 2020, 383, 1866–1876. [Google Scholar] [CrossRef] [PubMed]
  5. Gasparini, C.F.; Sutherland, H.G.; Griffiths, L.R. Studies on the pathophysiology and genetic basis of migraine. Curr. Genom. 2013, 14, 300–315. [Google Scholar] [CrossRef] [PubMed]
  6. Burstein, R.; Noseda, R.; Borsook, D. Migraine: Multiple processes, complex pathophysiology. J. Neurosci. 2015, 35, 6619–6629. [Google Scholar] [CrossRef]
  7. Dodick, D.W. A Phase-by-Phase Review of Migraine Pathophysiology. Headache 2018, 58 (Suppl. S1), 4–16. [Google Scholar] [CrossRef]
  8. Schankin, C.J.; Viana, M.; Goadsby, P.J. Persistent and Repetitive Visual Disturbances in Migraine: A Review. Headache 2017, 57, 1–16. [Google Scholar] [CrossRef]
  9. Khan, J.; Asoom, L.I.A.; Sunni, A.A.; Rafique, N.; Latif, R.; Saif, S.A.; Almandil, N.B.; Almohazey, D.; AbdulAzeez, S.; Borgio, J.F. Genetics, pathophysiology, diagnosis, treatment, management, and prevention of migraine. Biomed. Pharmacother. 2021, 139, 111557. [Google Scholar] [CrossRef]
  10. Buse, D.C.; Rupnow, M.F.; Lipton, R.B. Assessing and managing all aspects of migraine: Migraine attacks, migraine-related functional impairment, common comorbidities, and quality of life. Mayo Clin. Proc. 2009, 84, 422–435. [Google Scholar] [CrossRef]
  11. Gupta, J.; Gaurkar, S.S. Migraine: An Underestimated Neurological Condition Affecting Billions. Cureus 2022, 14, e28347. [Google Scholar] [CrossRef]
  12. Villar-Martinez, M.D.; Goadsby, P.J. Pathophysiology and Therapy of Associated Features of Migraine. Cells 2022, 11, 2767. [Google Scholar] [CrossRef]
  13. Durham, P.L. Calcitonin gene-related peptide (CGRP) and migraine. Headache 2006, 46 (Suppl. S1), S3–S8. [Google Scholar] [CrossRef] [PubMed]
  14. Waschek, J.A.; Baca, S.M.; Akerman, S. PACAP and migraine headache: Immunomodulation of neural circuits in autonomic ganglia and brain parenchyma. J. Headache Pain 2018, 19, 23. [Google Scholar] [CrossRef] [PubMed]
  15. Pellesi, L.; Al-Karagholi, M.A.; De Icco, R.; Coskun, H.; Elbahi, F.A.; Lopez-Lopez, C.; Snellman, J.; Hannibal, J.; Amin, F.M.; Ashina, M. Effect of Vasoactive Intestinal Polypeptide on Development of Migraine Headaches: A Randomized Clinical Trial. JAMA Netw. Open 2021, 4, e2118543. [Google Scholar] [CrossRef] [PubMed]
  16. Ghanizada, H.; Al-Karagholi, M.A.; Walker, C.S.; Arngrim, N.; Rees, T.; Petersen, J.; Siow, A.; Mørch-Rasmussen, M.; Tan, S.; O’Carroll, S.J.; et al. Amylin Analog Pramlintide Induces Migraine-like Attacks in Patients. Ann. Neurol. 2021, 89, 1157–1171. [Google Scholar] [CrossRef] [PubMed]
  17. May, A.; Goadsby, P.J. Substance P receptor antagonists in the therapy of migraine. Expert Opin. Investig. Drugs 2001, 10, 673–678. [Google Scholar] [CrossRef]
  18. Petersen, K.A.; Birk, S.; Kitamura, K.; Olesen, J. Effect of adrenomedullin on the cerebral circulation: Relevance to primary headache disorders. Cephalalgia 2009, 29, 23–30. [Google Scholar] [CrossRef]
  19. Hay, D.L.; Garelja, M.L.; Poyner, D.R.; Walker, C.S. Update on the pharmacology of calcitonin/CGRP family of peptides: IUPHAR Review 25. Br. J. Pharmacol. 2018, 175, 3–17. [Google Scholar] [CrossRef]
  20. Poyner, D.R.; Hay, D.L. Secretin family (Class B) G protein-coupled receptors—From molecular to clinical perspectives. Br. J. Pharmacol. 2012, 166, 1–3. [Google Scholar] [CrossRef]
  21. Edvinsson, L.; Grell, A.S.; Warfvinge, K. Expression of the CGRP Family of Neuropeptides and their Receptors in the Trigeminal Ganglion. J. Mol. Neurosci. 2020, 70, 930–944. [Google Scholar] [CrossRef]
  22. Dux, M.; Vogler, B.; Kuhn, A.; Mackenzie, K.D.; Stratton, J.; Messlinger, K. The Anti-CGRP Antibody Fremanezumab Lowers CGRP Release from Rat Dura Mater and Meningeal Blood Flow. Cells 2022, 11, 1768. [Google Scholar] [CrossRef]
  23. Pavelic, A.R.; Wöber, C.; Riederer, F.; Zebenholzer, K. Monoclonal Antibodies against Calcitonin Gene-Related Peptide for Migraine Prophylaxis: A Systematic Review of Real-World Data. Cells 2023, 12, 143. [Google Scholar] [CrossRef] [PubMed]
  24. Körtési, T.; Spekker, E.; Vécsei, L. Exploring the Tryptophan Metabolic Pathways in Migraine-Related Mechanisms. Cells 2022, 11, 3795. [Google Scholar] [CrossRef] [PubMed]
  25. Iyengar, S.; Johnson, K.W.; Ossipov, M.H.; Aurora, S.K. CGRP and the Trigeminal System in Migraine. Headache 2019, 59, 659–681. [Google Scholar] [CrossRef] [PubMed]
  26. Edvinsson, L.; Tajti, J.; Szalárdy, L.; Vécsei, L. PACAP and its role in primary headaches. J. Headache Pain 2018, 19, 21. [Google Scholar] [CrossRef] [PubMed]
  27. Körtési, T.; Tuka, B.; Nyári, A.; Vécsei, L.; Tajti, J. The effect of orofacial complete Freund’s adjuvant treatment on the expression of migraine-related molecules. J. Headache Pain 2019, 20, 43. [Google Scholar] [CrossRef] [PubMed]
  28. Russo, A.F. Calcitonin gene-related peptide (CGRP): A new target for migraine. Annu. Rev. Pharmacol. Toxicol. 2015, 55, 533–552. [Google Scholar] [CrossRef]
  29. Kaiser, E.A.; Russo, A.F. CGRP and migraine: Could PACAP play a role too? Neuropeptides 2013, 47, 451–461. [Google Scholar] [CrossRef]
  30. Kuburas, A.; Mason, B.N.; Hing, B.; Wattiez, A.S.; Reis, A.S.; Sowers, L.P.; Moldovan Loomis, C.; Garcia-Martinez, L.F.; Russo, A.F. PACAP Induces Light Aversion in Mice by an Inheritable Mechanism Independent of CGRP. J. Neurosci. 2021, 41, 4697–4715. [Google Scholar] [CrossRef]
  31. Russo, A.F. CGRP as a neuropeptide in migraine: Lessons from mice. Br. J. Clin. Pharmacol. 2015, 80, 403–414. [Google Scholar] [CrossRef]
  32. Goadsby, P.J.; Edvinsson, L.; Ekman, R. Release of vasoactive peptides in the extracerebral circulation of humans and the cat during activation of the trigeminovascular system. Ann. Neurol. 1988, 23, 193–196. [Google Scholar] [CrossRef] [PubMed]
  33. Tuka, B.; Helyes, Z.; Markovics, A.; Bagoly, T.; Szolcsányi, J.; Szabó, N.; Tóth, E.; Kincses, Z.T.; Vécsei, L.; Tajti, J. Alterations in PACAP-38-like immunoreactivity in the plasma during ictal and interictal periods of migraine patients. Cephalalgia 2013, 33, 1085–1095. [Google Scholar] [CrossRef] [PubMed]
  34. Lassen, L.H.; Haderslev, P.A.; Jacobsen, V.B.; Iversen, H.K.; Sperling, B.; Olesen, J. CGRP may play a causative role in migraine. Cephalalgia 2002, 22, 54–61. [Google Scholar] [CrossRef]
  35. Schytz, H.W.; Birk, S.; Wienecke, T.; Kruuse, C.; Olesen, J.; Ashina, M. PACAP38 induces migraine-like attacks in patients with migraine without aura. Brain 2009, 132 Pt 1, 16–25. [Google Scholar] [CrossRef] [PubMed]
  36. Vollesen, A.L.H.; Amin, F.M.; Ashina, M. Targeted Pituitary Adenylate Cyclase-Activating Peptide Therapies for Migraine. Neurotherapeutics 2018, 15, 371–376. [Google Scholar] [CrossRef] [PubMed]
  37. Ghanizada, H.; Al-Karagholi, M.A.; Arngrim, N.; Olesen, J.; Ashina, M. PACAP27 induces migraine-like attacks in migraine patients. Cephalalgia 2020, 40, 57–67. [Google Scholar] [CrossRef] [PubMed]
  38. Cernuda-Morollón, E.; Martínez-Camblor, P.; Alvarez, R.; Larrosa, D.; Ramón, C.; Pascual, J. Increased VIP levels in peripheral blood outside migraine attacks as a potential biomarker of cranial parasympathetic activation in chronic migraine. Cephalalgia 2015, 35, 310–316. [Google Scholar] [CrossRef] [PubMed]
  39. Hery, M.; Faudon, M.; Hery, F. Effect of vasoactive intestinal peptide on serotonin release in the suprachiasmatic area of the rat. Modulation by oestradiol. Peptides 1984, 5, 313–317. [Google Scholar] [CrossRef]
  40. Pellesi, L.; Al-Karagholi, M.A.; De Icco, R.; Chaudhry, B.A.; Lopez, C.L.; Snellman, J.; Hannibal, J.; Amin, F.M.; Ashina, M. Plasma Levels of CGRP During a 2-h Infusion of VIP in Healthy Volunteers and Patients With Migraine: An Exploratory Study. Front. Neurol. 2022, 13, 871176. [Google Scholar] [CrossRef]
  41. De la Fuente, M.; Delgado, M.; Gomariz, R.P. VIP modulation of immune cell functions. Adv. Neuroimmunol. 1996, 6, 75–91. [Google Scholar] [CrossRef]
  42. Hoffmann, J.; Baca, S.M.; Akerman, S. Neurovascular mechanisms of migraine and cluster headache. J. Cereb. Blood. Flow Metab. 2019, 39, 573–594. [Google Scholar] [CrossRef]
  43. Ocheretyaner, E.R.; Kofman, M.; Quattrocchi, E. Calcitonin gene-related peptide (CGRP) receptor antagonists for the acute treatment of migraines in adults. Drugs Context. 2022, 11, 2022-3-5. [Google Scholar] [CrossRef] [PubMed]
  44. Wang, X.; Chen, Y.; Song, J.; You, C. Efficacy and Safety of Monoclonal Antibody Against Calcitonin Gene-Related Peptide or Its Receptor for Migraine: A Systematic Review and Network Meta-analysis. Front. Pharmacol. 2021, 12, 649143. [Google Scholar] [CrossRef] [PubMed]
  45. Berger, A.A.; Winnick, A.; Popovsky, D.; Kaneb, A.; Berardino, K.; Kaye, A.M.; Cornett, E.M.; Kaye, A.D.; Viswanath, O.; Urits, I. Lasmiditan for the Treatment of Migraines With or Without Aura in Adults. Psychopharmacol. Bull. 2020, 50 (Suppl. S1), 163–188. [Google Scholar] [PubMed]
  46. Rissardo, J.P.; Caprara, A.L.F. Gepants for Acute and Preventive Migraine Treatment: A Narrative Review. Brain Sci. 2022, 12, 1612. [Google Scholar] [CrossRef] [PubMed]
  47. Ibekwe, A.; Perras, C.; Mierzwinski-Urban, M. Monoclonal Antibodies to Prevent Migraine Headaches. In CADTH Issues in Emerging Health Technologies; Canadian Agency for Drugs and Technologies in Health: Ottawa, ON, Canada, 2016. Available online: https://www.ncbi.nlm.nih.gov/books/NBK538376/ (accessed on 28 August 2023).
  48. Nie, L.; Sun, K.; Gong, Z.; Li, H.; Quinn, J.P.; Wang, M. Src Family Kinases Facilitate the Crosstalk between CGRP and Cytokines in Sensitizing Trigeminal Ganglion via Transmitting CGRP Receptor/PKA Pathway. Cells 2022, 11, 3498. [Google Scholar] [CrossRef] [PubMed]
  49. Greco, R.; Demartini, C.; Francavilla, M.; Zanaboni, A.M.; Tassorelli, C. Antagonism of CGRP Receptor: Central and Peripheral Mechanisms and Mediators in an Animal Model of Chronic Migraine. Cells 2022, 11, 3092. [Google Scholar] [CrossRef]
  50. Diener, H.C.; Förderreuther, S.; Gaul, C.; Giese, F.; Hamann, T.; Holle-Lee, D.; Jürgens, T.P.; Kamm, K.; Kraya, T.; Lampl, C.; et al. Prevention of migraine with monoclonal antibodies against CGRP or the CGRP receptor: Addition to the S1 guideline: Therapy of migraine attacks and prevention of migraine. Recommendations of the Germany Society of Neurology and the German Migraine and Headache Society. Neurol. Res. Pract. 2020, 2, 11. [Google Scholar] [CrossRef]
  51. Raffaelli, B.; De Icco, R.; Corrado, M.; Terhart, M.; Ailani, J. Open-label trials for CGRP-targeted drugs in migraine prevention: A narrative review. Cephalalgia 2023, 43, 3331024221137091. [Google Scholar] [CrossRef]
  52. FDA Approves Oral CGRP Receptor Antagonist Atogepant for Preventive Migraine Treatment. Available online: https://www.pharmacytimes.com/view/fda-approves-oral-cgrp-receptor-antagonist-atogepant-for-preventive-migraine-treatment (accessed on 13 November 2023).
  53. FDA Approves QULIPTA™ (Atogepant), the First and Only Oral CGRP Receptor Antagonist Specifically Developed for the Preventive Treatment of Migraine. Available online: https://news.abbvie.com/news/press-releases/fda-approves-qulipta-atogepant-first-and-only-oral-cgrp-receptor-antagonist-specifically-developed-for-preventive-treatment-migraine.htm (accessed on 13 November 2023).
  54. Nguyen, J.L.; Munshi, K.; Peasah, S.K.; Swart, E.C.S.; Kohli, M.; Henderson, R.; Good, C.B. Trends in utilization and costs of migraine medications, 2017–2020. J. Headache Pain 2022, 23, 111. [Google Scholar] [CrossRef]
  55. Haanes, K.A.; Edvinsson, L.; Sams, A. Understanding side-effects of anti-CGRP and anti-CGRP receptor antibodies. J. Headache Pain 2020, 21, 26. [Google Scholar] [CrossRef] [PubMed]
  56. Telegdy, G.; Adamik, A.; Tanaka, M.; Schally, A.V. Effects of the LHRH antagonist Cetrorelix on affective and cognitive functions in rats. Regul. Pept. 2010, 159, 142–147. [Google Scholar] [CrossRef] [PubMed]
  57. Tanaka, M.; Schally, A.V.; Telegdy, G. Neurotransmission of the antidepressant-like effects of the growth hormone-releasing hormone antagonist MZ-4-71. Behav. Brain Res. 2012, 228, 388–391. [Google Scholar] [CrossRef] [PubMed]
  58. Tanaka, M.; Telegdy, G. Neurotransmissions of antidepressant-like effects of neuromedin U-23 in mice. Behav. Brain Res. 2014, 259, 196–199. [Google Scholar] [CrossRef]
  59. Tanaka, M.; Csabafi, K.; Telegdy, G. Neurotransmissions of antidepressant-like effects of kisspeptin-13. Regul. Pept. 2013, 180, 1–4. [Google Scholar] [CrossRef]
  60. Telegdy, G.; Tanaka, M.; Schally, A.V. Effects of the growth hormone-releasing hormone (GH-RH) antagonist on brain functions in mice. Behav. Brain Res. 2011, 224, 155–158. [Google Scholar] [CrossRef] [PubMed]
  61. Rákosi, K.; Masaru, T.; Zarándia, M.; Telegdy, G.; Tóth, G.K. Short analogs and mimetics of human urocortin 3 display antidepressant effects in vivo. Peptides 2014, 62, 59–66. [Google Scholar] [CrossRef]
  62. Tran, K.N.; Nguyen, N.P.K.; Nguyen, L.T.H.; Shin, H.-M.; Yang, I.-J. Screening for Neuroprotective and Rapid Antidepressant-like Effects of 20 Essential Oils. Biomedicines 2023, 11, 1248. [Google Scholar] [CrossRef]
  63. Tanaka, M.; Kádár, K.; Tóth, G.; Telegdy, G. Antidepressant-like effects of urocortin 3 fragments. Brain Res. Bull. 2011, 84, 414–418. [Google Scholar] [CrossRef]
  64. Baliellas, D.E.M.; Barros, M.P.; Vardaris, C.V.; Guariroba, M.; Poppe, S.C.; Martins, M.F.; Pereira, Á.A.F.; Bondan, E.F. Propentofylline Improves Thiol-Based Antioxidant Defenses and Limits Lipid Peroxidation following Gliotoxic Injury in the Rat Brainstem. Biomedicines 2023, 11, 1652. [Google Scholar] [CrossRef]
  65. Montanari, M.; Imbriani, P.; Bonsi, P.; Martella, G.; Peppe, A. Beyond the Microbiota: Understanding the Role of the Enteric Nervous System in Parkinson’s Disease from Mice to Human. Biomedicines 2023, 11, 1560. [Google Scholar] [CrossRef] [PubMed]
  66. Garifulin, R.; Davleeva, M.; Izmailov, A.; Fadeev, F.; Markosyan, V.; Shevchenko, R.; Minyazeva, I.; Minekayev, T.; Lavrov, I.; Islamov, R. Evaluation of the Autologous Genetically Enriched Leucoconcentrate on the Lumbar Spinal Cord Morpho-Functional Recovery in a Mini Pig with Thoracic Spine Contusion Injury. Biomedicines 2023, 11, 1331. [Google Scholar] [CrossRef] [PubMed]
  67. Bueno, C.R.d.S.; Tonin, M.C.C.; Buchaim, D.V.; Barraviera, B.; Ferreira Junior, R.S.; Santos, P.S.d.S.; Reis, C.H.B.; Pastori, C.M.; Pereira, E.d.S.B.M.; Nogueira, D.M.B.; et al. Morphofunctional Improvement of the Facial Nerve and Muscles with Repair Using Heterologous Fibrin Biopolymer and Photobiomodulation. Pharmaceuticals 2023, 16, 653. [Google Scholar] [CrossRef] [PubMed]
  68. Sojka, A.; Żarowski, M.; Steinborn, B.; Hedzelek, W.; Wiśniewska-Spychała, B.; Dorocka-Bobkowska, B. Temporomandibular disorders in adolescents with headache. Adv. Clin. Exp. Med. 2018, 27, 193–199. [Google Scholar] [CrossRef]
  69. Polyák, H.; Galla, Z.; Nánási, N.; Cseh, E.K.; Rajda, C.; Veres, G.; Spekker, E.; Szabó, Á.; Klivényi, P.; Tanaka, M.; et al. The Tryptophan-Kynurenine Metabolic System Is Suppressed in Cuprizone-Induced Model of Demyelination Simulating Progressive Multiple Sclerosis. Biomedicines 2023, 11, 945. [Google Scholar] [CrossRef]
  70. Tanaka, M.; Szabó, Á.; Vécsei, L. Preclinical modeling in depression and anxiety: Current challenges and future research directions. Adv. Clin. Exp. Med. 2023, 32, 505–509. [Google Scholar] [CrossRef]
  71. Chu, P.-C.; Huang, C.-S.; Chang, P.-K.; Chen, R.-S.; Chen, K.-T.; Hsieh, T.-H.; Liu, H.-L. Weak Ultrasound Contributes to Neuromodulatory Effects in the Rat Motor Cortex. Int. J. Mol. Sci. 2023, 24, 2578. [Google Scholar] [CrossRef]
  72. Gecse, K.; Édes, A.E.; Nagy, T.; Demeter, A.K.; Virág, D.; Király, M.; Dalmadi Kiss, B.; Ludányi, K.; Környei, Z.; Denes, A.; et al. Citalopram Neuroendocrine Challenge Shows Altered Tryptophan and Kynurenine Metabolism in Migraine. Cells 2022, 11, 2258. [Google Scholar] [CrossRef]
  73. Nasini, S.; Tidei, S.; Shkodra, A.; De Gregorio, D.; Cambiaghi, M.; Comai, S. Age-Related Effects of Exogenous Melatonin on Anxiety-like Behavior in C57/B6J Mice. Biomedicines 2023, 11, 1705. [Google Scholar] [CrossRef]
  74. Chen, W.-C.; Wang, T.-S.; Chang, F.-Y.; Chen, P.-A.; Chen, Y.-C. Age, Dose, and Locomotion: Decoding Vulnerability to Ketamine in C57BL/6J and BALB/c Mice. Biomedicines 2023, 11, 1821. [Google Scholar] [CrossRef]
  75. Statsenko, Y.; Habuza, T.; Smetanina, D.; Simiyu, G.L.; Meribout, S.; King, F.C.; Gelovani, J.G.; Das, K.M.; Gorkom, K.N.-V.; Zaręba, K.; et al. Unraveling Lifelong Brain Morphometric Dynamics: A Protocol for Systematic Review and Meta-Analysis in Healthy Neurodevelopment and Ageing. Biomedicines 2023, 11, 1999. [Google Scholar] [CrossRef] [PubMed]
  76. Dang, J.; Tao, Q.; Niu, X.; Zhang, M.; Gao, X.; Yang, Z.; Yu, M.; Wang, W.; Han, S.; Cheng, J.; et al. Meta-Analysis of Structural and Functional Brain Abnormalities in Cocaine Addiction. Front. Psychiatry 2022, 13, 927075. [Google Scholar] [CrossRef]
  77. Balogh, L.; Tanaka, M.; Török, N.; Vécsei, L.; Taguchi, S. Crosstalk between Existential Phenomenological Psychotherapy and Neurological Sciences in Mood and Anxiety Disorders. Biomedicines 2021, 9, 340. [Google Scholar] [CrossRef] [PubMed]
  78. Rassler, B.; Blinowska, K.; Kaminski, M.; Pfurtscheller, G. Analysis of Respiratory Sinus Arrhythmia and Directed Information Flow between Brain and Body Indicate Different Management Strategies of fMRI-Related Anxiety. Biomedicines 2023, 11, 1028. [Google Scholar] [CrossRef]
  79. Arimura, A. Perspectives on pituitary adenylate cyclase activating polypeptide (PACAP) in the neuroendocrine, endocrine, and nervous systems. Jpn. J. Physiol. 1998, 48, 301–331. [Google Scholar] [CrossRef] [PubMed]
  80. Holland, P.R.; Barloese, M.; Fahrenkrug, J. PACAP in hypothalamic regulation of sleep and circadian rhythm: Importance for headache. J. Headache Pain 2018, 19, 20. [Google Scholar] [CrossRef]
  81. Maugeri, G.; D’Amico, A.G.; Musumeci, G.; Reglodi, D.; D’Agata, V. Effects of PACAP on Schwann Cells: Focus on Nerve Injury. Int. J. Mol. Sci. 2020, 21, 8233. [Google Scholar] [CrossRef]
  82. Johnson, G.C.; May, V.; Parsons, R.L.; Hammack, S.E. Parallel signaling pathways of pituitary adenylate cyclase activating polypeptide (PACAP) regulate several intrinsic ion channels. Ann. N. Y. Acad. Sci. 2019, 1455, 105–112. [Google Scholar] [CrossRef]
  83. Clement, A.; Guo, S.; Jansen-Olesen, I.; Christensen, S.L. ATP-Sensitive Potassium Channels in Migraine: Translational Findings and Therapeutic Potential. Cells 2022, 11, 2406. [Google Scholar] [CrossRef]
  84. Miyata, A.; Arimura, A.; Dahl, R.R.; Minamino, N.; Uehara, A.; Jiang, L.; Culler, M.D.; Coy, D.H. Isolation of a novel 38 residue-hypothalamic polypeptide which stimulates adenylate cyclase in pituitary cells. Biochem. Biophys. Res. Commun. 1989, 164, 567–574. [Google Scholar] [CrossRef]
  85. Denes, V.; Geck, P.; Mester, A.; Gabriel, R. Pituitary Adenylate Cyclase-Activating Polypeptide: 30 Years in Research Spotlight and 600 Million Years in Service. J. Clin. Med. 2019, 8, 1488. [Google Scholar] [CrossRef] [PubMed]
  86. Vaudry, D.; Nakamachi, T.; Basille, M.; Wurtz, O.; Fournier, A.; Shoida, S. Hypophysis Adenylate Cyclase Activating Polypeptide. In Handbook of Biologically Active Peptides, 2nd ed.; Academic Press: Boston, MA, USA, 2013; Available online: https://www.sciencedirect.com/topics/pharmacology-toxicology-and-pharmaceutical-science/hypophysis-adenylate-cyclase-activating-polypeptide (accessed on 28 August 2023).
  87. Tam, J.K.; Lee, L.T.; Chow, B.K. PACAP-related peptide (PRP)—Molecular evolution and potential functions. Peptides 2007, 28, 1920–1929. [Google Scholar] [CrossRef]
  88. Köves, K.; Szabó, E.; Kántor, O.; Heinzlmann, A.; Szabó, F.; Csáki, Á. Current State of Understanding of the Role of PACAP in the Hypothalamo-Hypophyseal Gonadotropin Functions of Mammals. Front. Endocrinol. 2020, 11, 88. [Google Scholar] [CrossRef]
  89. Lee, E.H.; Seo, S.R. Neuroprotective roles of pituitary adenylate cyclase-activating polypeptide in neurodegenerative diseases. BMB Rep. 2014, 47, 369–375. [Google Scholar] [CrossRef] [PubMed]
  90. Sadanandan, N.; Cozene, B.; Park, Y.J.; Farooq, J.; Kingsbury, C.; Wang, Z.J.; Moscatello, A.; Saft, M.; Cho, J.; Gonzales-Portillo, B.; et al. Pituitary Adenylate Cyclase-Activating Polypeptide: A Potent Therapeutic Agent in Oxidative Stress. Antioxidants 2021, 10, 354. [Google Scholar] [CrossRef] [PubMed]
  91. Waschek, J.A. VIP and PACAP: Neuropeptide modulators of CNS inflammation, injury, and repair. Br. J. Pharmacol. 2013, 169, 512–523. [Google Scholar] [CrossRef]
  92. Xiao, X.; Qiu, P.; Gong, H.Z.; Chen, X.M.; Sun, Y.; Hong, A.; Ma, Y. PACAP ameliorates hepatic metabolism and inflammation through up-regulating FAIM in obesity. J. Cell Mol. Med. 2019, 23, 5970–5980. [Google Scholar] [CrossRef] [PubMed]
  93. Gonkowski, S. Vasoactive Intestinal Polypeptide in the Carotid Body—A History of Forty Years of Research. A Mini Review. Int. J. Mol. Sci. 2020, 21, 4692. [Google Scholar] [CrossRef]
  94. Jiang, W.; Wang, H.; Li, Y.S.; Luo, W. Role of vasoactive intestinal peptide in osteoarthritis. J. Biomed. Sci. 2016, 23, 63. [Google Scholar] [CrossRef]
  95. Cao, S.G.; Wu, W.C.; Han, Z.; Wang, M.Y. Effects of psychological stress on small intestinal motility and expression of cholecystokinin and vasoactive intestinal polypeptide in plasma and small intestine in mice. World J. Gastroenterol. 2005, 11, 737–740. [Google Scholar] [CrossRef]
  96. Jacobs, B.; Dussor, G. Neurovascular contributions to migraine: Moving beyond vasodilation. Neuroscience 2016, 338, 130–144. [Google Scholar] [CrossRef] [PubMed]
  97. Langer, I.; Jeandriens, J.; Couvineau, A.; Sanmukh, S.; Latek, D. Signal Transduction by VIP and PACAP Receptors. Biomedicines 2022, 10, 406. [Google Scholar] [CrossRef] [PubMed]
  98. Fizanne, L.; Sigaudo-Roussel, D.; Saumet, J.L.; Fromy, B. Evidence for the involvement of VPAC1 and VPAC2 receptors in pressure-induced vasodilatation in rodents. J. Physiol. 2004, 554 Pt 2, 519–528. [Google Scholar] [CrossRef]
  99. Parsons, R.L.; May, V. PACAP-Induced PAC1 Receptor Internalization and Recruitment of Endosomal Signaling Regulate Cardiac Neuron Excitability. J. Mol. Neurosci. 2019, 68, 340–347. [Google Scholar] [CrossRef]
  100. Bill, C.A.; Vines, C.M. Phospholipase C. Adv. Exp. Med. Biol. 2020, 1131, 215–242. [Google Scholar] [CrossRef] [PubMed]
  101. Barloese, M.; Chitgar, M.; Hannibal, J.; Møller, S. Pituitary adenylate cyclase-activating peptide: Potential roles in the pathophysiology and complications of cirrhosis. Liver Int. 2020, 40, 2578–2589. [Google Scholar] [CrossRef] [PubMed]
  102. Makhinson, M.; Chotiner, J.K.; Watson, J.B.; O’Dell, T.J. Adenylyl cyclase activation modulates activity-dependent changes in synaptic strength and Ca2+/calmodulin-dependent kinase II autophosphorylation. J. Neurosci. 1999, 19, 2500–2510. [Google Scholar] [CrossRef]
  103. Johnson, G.L.; Lapadat, R. Mitogen-activated protein kinase pathways mediated by ERK, JNK, and p38 protein kinases. Science 2002, 298, 1911–1912. [Google Scholar] [CrossRef]
  104. Lu, J.; Piper, S.J.; Zhao, P.; Miller, L.J.; Wootten, D.; Sexton, P.M. Targeting VIP and PACAP Receptor Signaling: New Insights into Designing Drugs for the PACAP Subfamily of Receptors. Int. J. Mol. Sci. 2022, 23, 8069. [Google Scholar] [CrossRef]
  105. Hirabayashi, T.; Nakamachi, T.; Shioda, S. Discovery of PACAP and its receptors in the brain. J. Headache Pain 2018, 19, 28. [Google Scholar] [CrossRef]
  106. Watanabe, J. Vasoactive Intestinal Polypeptide Receptor 1. In Methods in Enzymology; Academic Press: Boston, MA, USA, 2013; Available online: https://www.sciencedirect.com/topics/medicine-and-dentistry/vasoactive-intestinal-polypeptide-receptor-1 (accessed on 28 August 2023).
  107. Vasoactive Intestinal Polypeptide Receptor. Autonomic Neuroscience. 2007. Available online: https://www.sciencedirect.com/topics/neuroscience/vasoactive-intestinal-polypeptide-receptor (accessed on 28 August 2023).
  108. May, V.; Buttolph, T.R.; Girard, B.M.; Clason, T.A.; Parsons, R.L. PACAP-induced ERK activation in HEK cells expressing PAC1 receptors involves both receptor internalization and PKC signaling. Am. J. Physiol. Cell Physiol. 2014, 306, C1068–C1079. [Google Scholar] [CrossRef] [PubMed]
  109. Hou, X.; Yang, D.; Yang, G.; Li, M.; Zhang, J.; Zhang, J.; Zhang, Y.; Liu, Y. Therapeutic potential of vasoactive intestinal peptide and its receptor VPAC2 in type 2 diabetes. Front. Endocrinol. 2022, 13, 984198. [Google Scholar] [CrossRef] [PubMed]
  110. Sundrum, T.; Walker, C.S. Pituitary adenylate cyclase-activating polypeptide receptors in the trigeminovascular system: Implications for migraine. Br. J. Pharmacol. 2018, 175, 4109–4120. [Google Scholar] [CrossRef] [PubMed]
  111. Liu, J.; Wang, G.; Dan, Y.; Liu, X. CGRP and PACAP-38 play an important role in diagnosing pediatric migraine. J. Headache Pain 2022, 23, 68. [Google Scholar] [CrossRef]
  112. Schytz, H.W.; Olesen, J.; Ashina, M. The PACAP receptor: A novel target for migraine treatment. Neurotherapeutics 2010, 7, 191–196. [Google Scholar] [CrossRef]
  113. Kuburas, A.; Russo, A.F. Shared and independent roles of CGRP and PACAP in migraine pathophysiology. J. Headache Pain 2023, 24, 34. [Google Scholar] [CrossRef]
  114. Ernstsen, C.; Christensen, S.L.; Rasmussen, R.H.; Nielsen, B.S.; Jansen-Olesen, I.; Olesen, J.; Kristensen, D.M. The PACAP pathway is independent of CGRP in mouse models of migraine: Possible new drug target? Brain 2022, 145, 2450–2460. [Google Scholar] [CrossRef]
  115. Christensen, C.E.; Ashina, M.; Amin, F.M. Calcitonin Gene-Related Peptide (CGRP) and Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) in Migraine Pathogenesis. Pharmaceuticals 2022, 15, 1189. [Google Scholar] [CrossRef]
  116. Anapindi, K.D.B.; Yang, N.; Romanova, E.V.; Rubakhin, S.S.; Tipton, A.; Dripps, I.; Sheets, Z.; Sweedler, J.V.; Pradhan, A.A. PACAP and Other Neuropeptide Targets Link Chronic Migraine and Opioid-induced Hyperalgesia in Mouse Models. Mol. Cell Proteomics 2019, 18, 2447–2458. [Google Scholar] [CrossRef]
  117. Silvestro, M.; Iannone, L.F.; Orologio, I.; Tessitore, A.; Tedeschi, G.; Geppetti, P.; Russo, A. Migraine Treatment: Towards New Pharmacological Targets. Int. J. Mol. Sci. 2023, 24, 12268. [Google Scholar] [CrossRef]
  118. Pellesi, L.; Chaudhry, B.A.; Vollesen, A.L.H.; Snoer, A.H.; Baumann, K.; Skov, P.S.; Jensen, R.H.; Ashina, M. PACAP38- and VIP-induced cluster headache attacks are not associated with changes of plasma CGRP or markers of mast cell activation. Cephalalgia 2022, 42, 687–695. [Google Scholar] [CrossRef]
  119. Rasmussen, L.; Deligianni, C.; Christensen, C.E.; Karlsson, W.K.; Al-Khazali, H.M.; Van de Casteele, T.; Granhall, C.; Amin, F.M.; Ashina, M. The effect of Lu AG09222 on PACAP38- and VIP-induced vasodilation, heart rate increase, and headache in healthy subjects: An interventional, randomized, double-blind, parallel-group, placebo-controlled study. J. Headache Pain 2023, 24, 60. [Google Scholar] [CrossRef] [PubMed]
  120. Vu, J.P.; Germano, P.M.; Pisegna, J.R. Adenylate Cyclase. Adenylate Cyclases Are Enzymes That Catalyze the Conversion of ATP to cAMP and Pyrophosphate. In From: The Senses: A Comprehensive Reference; Academic Press: Boston, MA, USA, 2008; Available online: https://www.sciencedirect.com/topics/agricultural-and-biological-sciences/adenylate-cyclase (accessed on 29 August 2023).
  121. Roberts, R.E. The extracellular signal-regulated kinase (ERK) pathway: A potential therapeutic target in hypertension. J. Exp. Pharmacol. 2012, 4, 77–83. [Google Scholar] [CrossRef]
  122. Lund, A.M.; Hannibal, J. Localization of the neuropeptides pituitary adenylate cyclase-activating polypeptide, vasoactive intestinal peptide, and their receptors in the basal brain blood vessels and trigeminal ganglion of the mouse CNS; an immunohistochemical study. Front. Neuroanat. 2022, 16, 991403. [Google Scholar] [CrossRef] [PubMed]
  123. Ivic, I.; Balasko, M.; Fulop, B.D.; Hashimoto, H.; Toth, G.; Tamas, A.; Juhasz, T.; Koller, A.; Reglodi, D.; Solymár, M. VPAC1 receptors play a dominant role in PACAP-induced vasorelaxation in female mice. PLoS ONE 2019, 14, e0211433. [Google Scholar] [CrossRef] [PubMed]
  124. VIP and PACAP Receptors—IUPHAR/BPS Guide to PHARMACOLOGY. Available online: https://www.guidetopharmacology.org/GRAC/FamilyDisplayForward?familyId=67 (accessed on 29 August 2023).
  125. Datki, Z.; Sinka, R. Translational biomedicine-oriented exploratory research on bioactive rotifer-specific biopolymers. Adv. Clin. Exp. Med. 2022, 31, 931–935. [Google Scholar] [CrossRef]
  126. Palotai, M.; Telegdy, G.; Tanaka, M.; Bagosi, Z.; Jászberényi, M. Neuropeptide AF induces anxiety-like and antidepressant-like behavior in mice. Behav. Brain Res. 2014, 274, 264–269. [Google Scholar] [CrossRef]
  127. Lieb, A.; Thaler, G.; Fogli, B.; Trovato, O.; Posch, M.A.; Kaserer, T.; Zangrandi, L. Functional Characterization of Spinocerebellar Ataxia Associated Dynorphin A Mutant Peptides. Biomedicines 2021, 9, 1882. [Google Scholar] [CrossRef]
  128. Skobeleva, K.; Shalygin, A.; Mikhaylova, E.; Guzhova, I.; Ryazantseva, M.; Kaznacheyeva, E. The STIM1/2-Regulated Calcium Homeostasis Is Impaired in Hippocampal Neurons of the 5xFAD Mouse Model of Alzheimer’s Disease. Int. J. Mol. Sci. 2022, 23, 14810. [Google Scholar] [CrossRef]
  129. Martos, D.; Tuka, B.; Tanaka, M.; Vécsei, L.; Telegdy, G. Memory Enhancement with Kynurenic Acid and Its Mechanisms in Neurotransmission. Biomedicines 2022, 10, 849. [Google Scholar] [CrossRef]
  130. Tanaka, M.; Szabó, Á.; Spekker, E.; Polyák, H.; Tóth, F.; Vécsei, L. Mitochondrial Impairment: A Common Motif in Neuropsychiatric Presentation? The Link to the Tryptophan–Kynurenine Metabolic System. Cells 2022, 11, 2607. [Google Scholar] [CrossRef] [PubMed]
  131. Tanaka, M.; Bohár, Z.; Martos, D.; Telegdy, G.; Vécsei, L. Antidepressant-like effects of kynurenic acid in a modified forced swim test. Pharmacol. Rep. 2020, 72, 449–455. [Google Scholar] [CrossRef]
  132. Tanaka, M.; Telegdy, G. Involvement of adrenergic and serotonergic receptors in antidepressant-like effect of urocortin 3 in a modified forced swimming test in mice. Brain Res. Bull. 2008, 77, 301–305. [Google Scholar] [CrossRef] [PubMed]
  133. Tanaka, M.; Spekker, E.; Szabó, Á.; Polyák, H.; Vécsei, L. Modelling the neurodevelopmental pathogenesis in neuropsychiatric disorders. Bioactive kynurenines and their analogues as neuroprotective agents-in celebration of 80th birthday of Professor Peter Riederer. J. Neural. Transm. 2022, 129, 627–642. [Google Scholar] [CrossRef]
  134. Reducha, P.V.; Edvinsson, L.; Haanes, K.A. Could Experimental Inflammation Provide Better Understanding of Migraines? Cells 2022, 11, 2444. [Google Scholar] [CrossRef] [PubMed]
  135. Ojala, J.; Tooke, K.; Hsiang, H.; Girard, B.M.; May, V.; Vizzard, M.A. PACAP/PAC1 Expression and Function in Micturition Pathways. J. Mol. Neurosci. 2019, 68, 357–367. [Google Scholar] [CrossRef] [PubMed]
  136. Tamas, A.; Reglodi, D.; Farkas, O.; Kovesdi, E.; Pal, J.; Povlishock, J.T.; Schwarcz, A.; Czeiter, E.; Szanto, Z.; Doczi, T.; et al. Effect of PACAP in central and peripheral nerve injuries. Int. J. Mol. Sci. 2012, 13, 8430–8448. [Google Scholar] [CrossRef]
  137. Zhang, L.; Zhou, Y.; Yang, L.; Wang, Y.; Xiao, Z. PACAP6-38 improves nitroglycerin-induced central sensitization by modulating synaptic plasticity at the trigeminal nucleus caudalis in a male rat model of chronic migraine. J. Headache Pain 2023, 24, 66. [Google Scholar] [CrossRef]
  138. Takács-Lovász, K.; Kun, J.; Aczél, T.; Urbán, P.; Gyenesei, A.; Bölcskei, K.; Szőke, É.; Helyes, Z. PACAP-38 Induces Transcriptomic Changes in Rat Trigeminal Ganglion Cells Related to Neuroinflammation and Altered Mitochondrial Function Presumably via PAC1/VPAC2 Receptor-Independent Mechanism. Int. J. Mol. Sci. 2022, 23, 2120. [Google Scholar] [CrossRef]
  139. Frederiksen, S.D.; Haanes, K.A.; Warfvinge, K.; Edvinsson, L. Perivascular neurotransmitters: Regulation of cerebral blood flow and role in primary headaches. J. Cereb. Blood Flow Metab. 2019, 39, 610–632. [Google Scholar] [CrossRef]
  140. Markovics, A.; Kormos, V.; Gaszner, B.; Lashgarara, A.; Szoke, E.; Sandor, K.; Szabadfi, K.; Tuka, B.; Tajti, J.; Szolcsanyi, J.; et al. Pituitary adenylate cyclase-activating polypeptide plays a key role in nitroglycerol-induced trigeminovascular activation in mice. Neurobiol. Dis. 2012, 45, 633–644. [Google Scholar] [CrossRef] [PubMed]
  141. Edvinsson, L. PACAP and its receptors in migraine pathophysiology: Commentary on Walker et al. Br. J. Pharmacol. 171: 1521–1533. Br. J. Pharmacol. 2015, 172, 4782–4784. [Google Scholar] [CrossRef] [PubMed]
  142. Saposnik, G.; Montalban, X.; Selchen, D.; Terzaghi, M.A.; Bakdache, F.; Montoya, A.; Fruns, M.; Caceres, F.; Oh, J. Therapeutic Inertia in Multiple Sclerosis Care: A Study of Canadian Neurologists. Front. Neurol. 2018, 9, 781. [Google Scholar] [CrossRef]
  143. Harding, S.D.; Armstrong, J.F.; Faccenda, E.; Southan, C.; Alexander, S.P.H.; Davenport, A.P.; Pawson, A.J.; Spedding, M.; Davies, J.A.; NC-IUPHAR. The IUPHAR/BPS guide to PHARMACOLOGY in 2022: Curating pharmacology for COVID-19, malaria and antibacterials. Nucl. Acids Res. 2022, 50, D1282–D1294. [Google Scholar] [CrossRef]
  144. Guo, S.; Jansen-Olesen, I.; Olesen, J.; Christensen, S.L. Role of PACAP in migraine: An alternative to CGRP? Neurobiol. Dis. 2023, 176, 105946. [Google Scholar] [CrossRef] [PubMed]
  145. Granoth, R.; Fridkin, M.; Gozes, I. VIP and the potent analog, stearyl-Nle(17)-VIP, induce proliferation of keratinocytes. FEBS Lett. 2000, 475, 78–83. [Google Scholar] [CrossRef]
  146. Gourlet, P.; De Neef, P.; Cnudde, J.; Waelbroeck, M.; Robberecht, P. In vitro properties of a high affinity selective antagonist of the VIP1 receptor. Peptide 1997, 18, 1555–1560. [Google Scholar] [CrossRef]
  147. Beebe, X.; Darczak, D.; Davis-Taber, R.A.; Uchic, M.E.; Scott, V.E.; Jarvis, M.F.; Stewart, A.O. Discovery and SAR of hydrazide antagonists of the pituitary adenylate cyclase-activating polypeptide (PACAP) receptor type 1 (PAC1-R). Bioorg. Med. Chem. Lett. 2008, 18, 2162–2166. [Google Scholar] [CrossRef]
  148. Laburthe, M.; Couvineau, A.; Tan, V. Class II G protein-coupled receptors for VIP and PACAP: Structure, models of activation and pharmacology. Peptides 2007, 28, 1631–1639. [Google Scholar] [CrossRef]
  149. Tasma, Z.; Siow, A.; Harris, P.W.R.; Brimble, M.A.; Hay, D.L.; Walker, C.S. Characterisation of agonist signalling profiles and agonist-dependent antagonism at PACAP-responsive receptors: Implications for drug discovery. Br. J. Pharmacol. 2022, 179, 435–453. [Google Scholar] [CrossRef]
  150. Spekker, E.; Tanaka, M.; Szabó, Á.; Vécsei, L. Neurogenic Inflammation: The Participant in Migraine and Recent Advancements in Translational Research. Biomedicines 2022, 10, 76. [Google Scholar] [CrossRef] [PubMed]
  151. Pinho-Ribeiro, F.A.; Verri, W.A., Jr.; Chiu, I.M. Nociceptor Sensory Neuron-Immune Interactions in Pain and Inflammation. Trends Immunol. 2017, 38, 5–19. [Google Scholar] [CrossRef] [PubMed]
  152. Guo, S.; Vollesen, A.L.; Hansen, R.D.; Esserlind, A.L.; Amin, F.M.; Christensen, A.F.; Olesen, J.; Ashina, M. Part I: Pituitary adenylate cyclase-activating polypeptide-38 induced migraine-like attacks in patients with and without familial aggregation of migraine. Cephalalgia 2017, 37, 125–135. [Google Scholar] [CrossRef] [PubMed]
  153. ClinicalTrials.gov. PACAP Induced Migraine Attacks in Patients with High and Low Genetic Load. Available online: https://classic.clinicaltrials.gov/ct2/show/NCT02158221 (accessed on 29 August 2023).
  154. Togha, M.; Ghorbani, Z.; Ramazi, S.; Zavvari, F.; Karimzadeh, F. Evaluation of Serum Levels of Transient Receptor Potential Cation Channel Subfamily V Member 1, Vasoactive Intestinal Polypeptide, and Pituitary Adenylate Cyclase-Activating Polypeptide in Chronic and Episodic Migraine: The Possible Role in Migraine Transformation. Front. Neurol. 2021, 12, 770980. [Google Scholar] [CrossRef] [PubMed]
  155. Körtési, T.; Tuka, B.; Tajti, J.; Bagoly, T.; Fülöp, F.; Helyes, Z.; Vécsei, L. Kynurenic Acid Inhibits the Electrical Stimulation Induced Elevated Pituitary Adenylate Cyclase-Activating Polypeptide Expression in the TNC. Front. Neurol. 2018, 8, 745. [Google Scholar] [CrossRef]
  156. Guo, S.; Vollesen, A.L.; Hansen, Y.B.; Frandsen, E.; Andersen, M.R.; Amin, F.M.; Fahrenkrug, J.; Olesen, J.; Ashina, M. Part II: Biochemical changes after pituitary adenylate cyclase-activating polypeptide-38 infusion in migraine patients. Cephalalgia 2017, 37, 136–147. [Google Scholar] [CrossRef]
  157. Amin, F.M.; Asghar, M.S.; Guo, S.; Hougaard, A.; Hansen, A.E.; Schytz, H.W.; van der Geest, R.J.; de Koning, P.J.; Larsson, H.B.; Olesen, J.; et al. Headache and prolonged dilatation of the middle meningeal artery by PACAP38 in healthy volunteers. Cephalalgia 2012, 32, 140–149. [Google Scholar] [CrossRef] [PubMed]
  158. Maasz, G.; Zrinyi, Z.; Reglodi, D.; Petrovics, D.; Rivnyak, A.; Kiss, T.; Jungling, A.; Tamas, A.; Pirger, Z. Pituitary adenylate cyclase-activating polypeptide (PACAP) has a neuroprotective function in dopamine-based neurodegeneration in rat and snail parkinsonian models. Dis. Model Mech. 2017, 10, 127–139. [Google Scholar] [CrossRef]
  159. Rubio-Beltrán, E.; Correnti, E.; Deen, M.; Kamm, K.; Kelderman, T.; Papetti, L.; Vigneri, S.; MaassenVanDenBrink, A.; Edvinsson, L.; European Headache Federation School of Advanced Studies (EHF-SAS). PACAP38 and PAC1 receptor blockade: A new target for headache? J. Headache Pain 2018, 19, 64. [Google Scholar] [CrossRef]
  160. Ashina, M.; Doležil, D.; Bonner, J.H.; Zhou, L.; Klatt, J.; Picard, H.; Mikol, D.D. A phase 2, randomized, double-blind, placebo-controlled trial of AMG 301, a pituitary adenylate cyclase-activating polypeptide PAC1 receptor monoclonal antibody for migraine prevention. Cephalalgia 2021, 41, 33–44. [Google Scholar] [CrossRef]
  161. Study to Evaluate the Efficacy and Safety of AMG 301 in Migraine Prevention 2020. Available online: https://ClinicalTrials.gov/show/NCT03238781 (accessed on 4 September 2023).
  162. Lundbeck News Room: Lundbeck Announced the Start of a Phase II Clinical Study to Assess Lu AG09222 for Migraine Prevention. Available online: https://newsroom.lundbeckus.com/news-release/2021/lundbeck-announced-start-of-phase-ii-clinical-study-for-migraine-prevention (accessed on 29 August 2023).
  163. A Study With Lu AG09222 in Adults With Migraine Who Have Not Been Helped by Prior Preventive Treatments 2023. Available online: https://classic.clinicaltrials.gov/ct2/show/NCT05133323 (accessed on 4 September 2023).
  164. A Study of LY3451838 in Participants with Migraine 2023. Available online: https://ClinicalTrials.gov/show/NCT04498910 (accessed on 4 September 2023).
  165. ClinicalTrials.gov. The Effects of a Long-Lasting Infusion of Vasoactive Intestinal Peptide (VIP) in Episodic Migraine Patients. Available online: https://classic.clinicaltrials.gov/ct2/show/NCT04260035 (accessed on 29 August 2023).
  166. Terry, J.S.; Anderson, L.B.; Scherman, M.S.; McAlister, C.E.; Perera, R.; Schountz, T.; Geiss, B.J. Development of a SARS-CoV-2 nucleocapsid specific monoclonal antibody. Virology 2021, 558, 28–37. [Google Scholar] [CrossRef] [PubMed]
  167. Sgro, C. Side-effects of a monoclonal antibody, muromonab CD3/orthoclone OKT3: Bibliographic review. Toxicology 1995, 105, 23–29. [Google Scholar] [CrossRef] [PubMed]
  168. Tanaka, M.; Szabó, Á.; Vécsei, L. Integrating Armchair, Bench, and Bedside Research for Behavioral Neurology and Neuropsychiatry: Editorial. Biomedicines 2022, 10, 2999. [Google Scholar] [CrossRef] [PubMed]
  169. Gaebler, A.J.; Finner-Prével, M.; Sudar, F.P.; Langer, F.H.; Keskin, F.; Gebel, A.; Zweerings, J.; Mathiak, K. The Interplay between Vitamin D, Exposure of Anticholinergic Antipsychotics and Cognition in Schizophrenia. Biomedicines 2022, 10, 1096. [Google Scholar] [CrossRef] [PubMed]
  170. Castillo-Mariqueo, L.; Giménez-Llort, L. Impact of Behavioral Assessment and Re-Test as Functional Trainings That Modify Survival, Anxiety and Functional Profile (Physical Endurance and Motor Learning) of Old Male and Female 3xTg-AD Mice and NTg Mice with Normal Aging. Biomedicines 2022, 10, 973. [Google Scholar] [CrossRef]
  171. Lee, E.C.; Hong, D.-Y.; Lee, D.-H.; Park, S.-W.; Lee, J.Y.; Jeong, J.H.; Kim, E.-Y.; Chung, H.-M.; Hong, K.-S.; Park, S.-P.; et al. Inflammation and Rho-Associated Protein Kinase-Induced Brain Changes in Vascular Dementia. Biomedicines 2022, 10, 446. [Google Scholar] [CrossRef]
  172. Simonato, M.; Dall’Acqua, S.; Zilli, C.; Sut, S.; Tenconi, R.; Gallo, N.; Sfriso, P.; Sartori, L.; Cavallin, F.; Fiocco, U.; et al. Tryptophan Metabolites, Cytokines, and Fatty Acid Binding Protein 2 in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Biomedicines 2021, 9, 1724. [Google Scholar] [CrossRef]
  173. Smagin, D.A.; Kovalenko, I.L.; Galyamina, A.G.; Belozertseva, I.V.; Tamkovich, N.V.; Baranov, K.O.; Kudryavtseva, N.N. Chronic Lithium Treatment Affects Anxious Behaviors and theExpression of Serotonergic Genes in Midbrain Raphe Nuclei of Defeated Male Mice. Biomedicines 2021, 9, 1293. [Google Scholar] [CrossRef]
  174. Tanaka, M.; Tóth, F.; Polyák, H.; Szabó, Á.; Mándi, Y.; Vécsei, L. Immune Influencers in Action: Metabolites and Enzymes of the Tryptophan-Kynurenine Metabolic Pathway. Biomedicines 2021, 9, 734. [Google Scholar] [CrossRef]
  175. Tanaka, M.; Török, N.; Tóth, F.; Szabó, Á.; Vécsei, L. Co-Players in Chronic Pain: Neuroinflammation and the Tryptophan-Kynurenine Metabolic Pathway. Biomedicines 2021, 9, 897. [Google Scholar] [CrossRef]
  176. Vila-Merkle, H.; González-Martínez, A.; Campos-Jiménez, R.; Martínez-Ricós, J.; Teruel-Martí, V.; Blasco-Serra, A.; Lloret, A.; Celada, P.; Cervera-Ferri, A. The Oscillatory Profile Induced by the Anxiogenic Drug FG-7142 in the Amygdala–Hippocampal Network Is Reversed by Infralimbic Deep Brain Stimulation: Relevance for Mood Disorders. Biomedicines 2021, 9, 783. [Google Scholar] [CrossRef]
  177. Santana-Santana, M.; Bayascas, J.-R.; Giménez-Llort, L. Fine-Tuning the PI3K/Akt Signaling Pathway Intensity by Sex and Genotype-Load: Sex-Dependent Homozygotic Threshold for Somatic Growth but Feminization of Anxious Phenotype in Middle-Aged PDK1 K465E Knock-In and Heterozygous Mice. Biomedicines 2021, 9, 747. [Google Scholar] [CrossRef]
  178. Muntsant, A.; Giménez-Llort, L. Genotype Load Modulates Amyloid Burden and Anxiety-Like Patterns in Male 3xTg-AD Survivors despite Similar Neuro-Immunoendocrine, Synaptic and Cognitive Impairments. Biomedicines 2021, 9, 715. [Google Scholar] [CrossRef] [PubMed]
  179. Giménez-Llort, L.; Marin-Pardo, D.; Marazuela, P.; Hernández-Guillamón, M. Survival Bias and Crosstalk between Chronological and Behavioral Age: Age- and Genotype-Sensitivity Tests Define Behavioral Signatures in Middle-Aged, Old, and Long-Lived Mice with Normal and AD-Associated Aging. Biomedicines 2021, 9, 636. [Google Scholar] [CrossRef] [PubMed]
  180. Komatsu, H.; Watanabe, E.; Fukuchi, M. Psychiatric Neural Networks and Precision Therapeutics by Machine Learning. Biomedicines 2021, 9, 403. [Google Scholar] [CrossRef] [PubMed]
  181. Caruso, G.; Godos, J.; Castellano, S.; Micek, A.; Murabito, P.; Galvano, F.; Ferri, R.; Grosso, G.; Caraci, F. The Therapeutic Potential of Carnosine/Anserine Supplementation against Cognitive Decline: A Systematic Review with Meta-Analysis. Biomedicines 2021, 9, 253. [Google Scholar] [CrossRef]
  182. Correia, B.S.B.; Nani, J.V.; Waladares Ricardo, R.; Stanisic, D.; Costa, T.B.B.C.; Hayashi, M.A.F.; Tasic, L. Effects of Psychostimulants and Antipsychotics on Serum Lipids in an Animal Model for Schizophrenia. Biomedicines 2021, 9, 235. [Google Scholar] [CrossRef] [PubMed]
  183. Ikonnikova, A.; Anisimova, A.; Galkin, S.; Gunchenko, A.; Abdukhalikova, Z.; Filippova, M.; Surzhikov, S.; Selyaeva, L.; Shershov, V.; Zasedatelev, A.; et al. Genetic Association Study and Machine Learning to Investigate Differences in Platelet Reactivity in Patients with Acute Ischemic Stroke Treated with Aspirin. Biomedicines 2022, 10, 2564. [Google Scholar] [CrossRef]
  184. Fan, P.; Miranda, O.; Qi, X.; Kofler, J.; Sweet, R.A.; Wang, L. Unveiling the Enigma: Exploring Risk Factors and Mechanisms for Psychotic Symptoms in Alzheimer’s Disease through Electronic Medical Records with Deep Learning Models. Pharmaceuticals 2023, 16, 911. [Google Scholar] [CrossRef]
  185. Parolini, F.; Goethel, M.; Becker, K.; Fernandes, C.; Fernandes, R.J.; Ervilha, U.F.; Santos, R.; Vilas-Boas, J.P. Breaking Barriers: Artificial Intelligence Interpreting the Interplay between Mental Illness and Pain as Defined by the International Association for the Study of Pain. Biomedicines 2023, 11, 2042. [Google Scholar] [CrossRef]
  186. Tanaka, M.; Szabó, Á.; Vécsei, L.; Giménez-Llort, L. Emerging Translational Research in Neurological and Psychiatric Diseases: From In Vitro to In Vivo Models. Int. J. Mol. Sci. 2023, 24, 15739. [Google Scholar] [CrossRef]
  187. Tanaka, M.; Telegdy, G. Antidepressant-like effects of the CRF family peptides, urocortin 1, urocortin 2 and urocortin 3 in a modified forced swimming test in mice. Brain Res. Bull. 2008, 75, 509–512. [Google Scholar] [CrossRef] [PubMed]
  188. Homberg, J.R.; Kyzar, E.J.; Scattoni, M.L.; Norton, W.H.; Pittman, J.; Gaikwad, S.; Nguyen, M.; Poudel, M.K.; Ullmann, J.F.; Diamond, D.M.; et al. Genetic and environmental modulation of neurodevelopmental disorders: Translational insights from labs to beds. Brain Res. Bull. 2016, 125, 79–91. [Google Scholar] [CrossRef] [PubMed]
  189. Ren, J.; Xiao, H. Exercise for Mental Well-Being: Exploring Neurobiological Advances and Intervention Effects in Depression. Life 2023, 13, 1505. [Google Scholar] [CrossRef] [PubMed]
  190. Tanaka, M.; Diano, M.; Battaglia, S. Editorial: Insights into structural and functional organization of the brain: Evidence from neuroimaging and non-invasive brain stimulation techniques. Front. Psychiatry. 2023, 14, 1225755. [Google Scholar] [CrossRef] [PubMed]
  191. Simon, C.; Soga, T.; Ahemad, N.; Bhuvanendran, S.; Parhar, I. Kisspeptin-10 Rescues Cholinergic Differentiated SHSY-5Y Cells from α-Synuclein-Induced Toxicity In Vitro. Int. J. Mol. Sci. 2022, 23, 5193. [Google Scholar] [CrossRef]
  192. Okanda Nyatega, C.; Qiang, L.; Jajere Adamu, M.; Bello Kawuwa, H. Altered striatal functional connectivity and structural dysconnectivity in individuals with bipolar disorder: A resting state magnetic resonance imaging study. Front. Psychiatry. 2022, 13, 1054380. [Google Scholar] [CrossRef] [PubMed]
  193. Liu, N.; Li, Y.; Hong, Y.; Huo, J.; Chang, T.; Wang, H.; Huang, Y.; Li, W.; Zhang, Y. Altered brain activities in mesocorticolimbic pathway in primary dysmenorrhea patients of long-term menstrual pain. Front. Neurosci. 2023, 17, 1098573. [Google Scholar] [CrossRef] [PubMed]
  194. Du, H.; Yang, B.; Wang, H.; Zeng, Y.; Xin, J.; Li, X. The non-linear correlation between the volume of cerebral white matter lesions and incidence of bipolar disorder: A secondary analysis of data from a cross-sectional study. Front. Psychiatry 2023, 14, 1149663. [Google Scholar] [CrossRef]
  195. Chen, Y.; Yu, R.; DeSouza, J.F.X.; Shen, Y.; Zhang, H.; Zhu, C.; Huang, P.; Wang, C. Differential responses from the left postcentral gyrus, right middle frontal gyrus, and precuneus to meal ingestion in patients with functional dyspepsia. Front. Psychiatry 2023, 14, 1184797. [Google Scholar] [CrossRef]
  196. Adamu, M.J.; Qiang, L.; Nyatega, C.O.; Younis, A.; Kawuwa, H.B.; Jabire, A.H.; Saminu, S. Unraveling the pathophysiology of schizophrenia: Insights from structural magnetic resonance imaging studies. Front. Psychiatry 2023, 14, 1188603. [Google Scholar] [CrossRef] [PubMed]
  197. Battaglia, S.; Nazzi, C.; Thayer, J.F. Fear-induced bradycardia in mental disorders: Foundations, current advances, future perspectives. Neurosci. Biobehav. Rev. 2023, 149, 105163. [Google Scholar] [CrossRef] [PubMed]
  198. Battaglia, S.; Nazzi, C.; Thayer, J.F. Heart’s tale of trauma: Fear-conditioned heart rate changes in post-traumatic stress disorder. Acta Psychiatr. Scand. 2023, 148, 463–466, Advance online publication. [Google Scholar] [CrossRef] [PubMed]
  199. Battaglia, S.; Di Fazio, C.; Vicario, C.M.; Avenanti, A. Neuropharmacological Modulation of N-methyl-D-aspartate, Noradrenaline and Endocannabinoid Receptors in Fear Extinction Learning: Synaptic Transmission and Plasticity. Int. J. Mol. Sci. 2023, 24, 5926. [Google Scholar] [CrossRef]
  200. Battaglia, M.R.; Di Fazio, C.; Battaglia, S. Activated Tryptophan-Kynurenine metabolic system in the human brain is associated with learned fear. Front. Mol. Neurosci. 2023, 16, 1217090. [Google Scholar] [CrossRef]
  201. Battaglia, S.; Garofalo, S.; di Pellegrino, G.; Starita, F. Revaluing the Role of vmPFC in the Acquisition of Pavlovian Threat Conditioning in Humans. J. Neurosci. 2020, 40, 8491–8500. [Google Scholar] [CrossRef]
  202. Tanaka, M. Crosstalk between Depression, Anxiety, Dementia, and Chronic Pain: Comorbidity in Behavioral Neurology and Neuropsychiatry 2.0. Biomedicines 2023. [Google Scholar] [CrossRef]
  203. Tanaka, M.; Telegdy, G. Antidepressant-Like Effects of Neuropeptide SF (NPSF). arXiv 2005, arXiv.2005.13256. [Google Scholar] [CrossRef]
  204. Battaglia, S.; Harrison, B.J.; Fullana, M.A. Does the human ventromedial prefrontal cortex support fear learning, fear extinction or both? A commentary on subregional contributions. Mol. Psychiatry 2022, 27, 784–786. [Google Scholar] [CrossRef]
  205. Di Gregorio, F.; Battaglia, S. Advances in EEG-based functional connectivity approaches to the study of the central nervous system in health and disease. Adv. Clin. Exp. Med. 2023, 32, 607–612. [Google Scholar] [CrossRef]
  206. Tanaka, M.; Vécsei, L. Editorial of Special Issue “Crosstalk between Depression, Anxiety, and Dementia: Comorbidity in Behavioral Neurology and Neuropsychiatry”. Biomedicines 2021, 9, 517. [Google Scholar] [CrossRef] [PubMed]
  207. Tanaka, M.; Vécsei, L. Monitoring the kynurenine system: Concentrations, ratios or what else? Adv. Clin. Exp. Med. 2021, 30, 775–778. [Google Scholar] [CrossRef] [PubMed]
  208. Tanaka, M.; Vécsei, L. Monitoring the Redox Status in Multiple Sclerosis. Biomedicines 2020, 8, 406. [Google Scholar] [CrossRef] [PubMed]
  209. Tanaka, M.; Török, N.; Vécsei, L. Are 5-HT1 receptor agonists effective anti-migraine drugs? Expert. Opin. Pharmacother. 2021, 22, 1221–1225. [Google Scholar] [CrossRef]
  210. Vuralli, D.; Ayata, C.; Bolay, H. Cognitive dysfunction and migraine. J. Headache Pain 2018, 19, 109. [Google Scholar] [CrossRef]
  211. Minen, M.T.; Begasse De Dhaem, O.; Kroon Van Diest, A.; Powers, S.; Schwedt, T.J.; Lipton, R.; Silbersweig, D. Migraine and its psychiatric comorbidities. J. Neurol. Neurosurg. Psychiatry 2016, 87, 741–749. [Google Scholar] [CrossRef]
  212. Tanaka, M.; Chen, C. Editorial: Towards a mechanistic understanding of depression, anxiety, and their comorbidity: Perspectives from cognitive neuroscience. Front. Behav. Neurosci. 2023, 17, 1268156. [Google Scholar] [CrossRef]
  213. Gonzalez-Escamilla, G.; Dörfel, D.; Becke, M.; Trefz, J.; Bonanno, G.A.; Groppa, S. Associating Flexible Regulation of Emotional Expression With Psychopathological Symptoms. Front. Behav. Neurosci. 2022, 16, 924305. [Google Scholar] [CrossRef]
  214. Battaglia, S.; Cardellicchio, P.; Di Fazio, C.; Nazzi, C.; Fracasso, A.; Borgomaneri, S. The Influence of Vicarious Fear-Learning in “Infecting” Reactive Action Inhibition. Front. Behav. Neurosci. 2022, 16, 946263. [Google Scholar] [CrossRef]
  215. Battaglia, S.; Cardellicchio, P.; Di Fazio, C.; Nazzi, C.; Fracasso, A.; Borgomaneri, S. Stopping in (e)motion: Reactive action inhibition when facing valence-independent emotional stimuli. Front. Behav. Neurosci. 2022, 16, 998714. [Google Scholar] [CrossRef]
  216. Ironside, M.; DeVille, D.C.; Kuplicki, R.T.; Burrows, K.P.; Smith, R.; Teed, A.R.; Paulus, M.P.; Khalsa, S.S. The unique face of comorbid anxiety and depression: Increased interoceptive fearfulness and reactivity. Front. Behav. Neurosci. 2023, 16, 1083357. [Google Scholar] [CrossRef] [PubMed]
  217. Rajkumar, R.P. Comorbid depression and anxiety: Integration of insights from attachment theory and cognitive neuroscience, and their implications for research and treatment. Front. Behav. Neurosci. 2022, 16, 1104928. [Google Scholar] [CrossRef] [PubMed]
  218. Vila-Merkle, H.; González-Martínez, A.; Campos-Jiménez, R.; Martínez-Ricós, J.; Teruel-Martí, V.; Lloret, A.; Blasco-Serra, A.; Cervera-Ferri, A. Sex differences in amygdalohippocampal oscillations and neuronal activation in a rodent anxiety model and in response to infralimbic deep brain stimulation. Front. Behav. Neurosci. 2023, 17, 1122163. [Google Scholar] [CrossRef] [PubMed]
  219. Panov, G.; Panova, P. Obsessive-compulsive symptoms in patient with schizophrenia: The influence of disorganized symptoms, duration of schizophrenia, and drug resistance. Front. Psychiatry 2023, 14, 1120974. [Google Scholar] [CrossRef] [PubMed]
  220. Hakamata, Y.; Hori, H.; Mizukami, S.; Izawa, S.; Yoshida, F.; Moriguchi, Y.; Hanakawa, T.; Inoue, Y.; Tagaya, H. Blunted diurnal interleukin-6 rhythm is associated with amygdala emotional hyporeactivity and depression: A modulating role of gene-stressor interactions. Front. Psychiatry 2023, 14, 1196235. [Google Scholar] [CrossRef] [PubMed]
  221. Fraile-Ramos, J.; Garrit, A.; Reig-Vilallonga, J.; Giménez-Llort, L. Hepatic Oxi-Inflammation and Neophobia as Potential Liver–Brain Axis Targets for Alzheimer’s Disease and Aging, with Strong Sensitivity to Sex, Isolation, and Obesity. Cells 2023, 12, 1517. [Google Scholar] [CrossRef] [PubMed]
  222. Sobolewska-Nowak, J.; Wachowska, K.; Nowak, A.; Orzechowska, A.; Szulc, A.; Płaza, O.; Gałecki, P. Exploring the Heart–Mind Connection: Unraveling the Shared Pathways between Depression and Cardiovascular Diseases. Biomedicines 2023, 11, 1903. [Google Scholar] [CrossRef]
  223. Festa, F.; Medori, S.; Macrì, M. Move Your Body, Boost Your Brain: The Positive Impact of Physical Activity on Cognition across All Age Groups. Biomedicines 2023, 11, 1765. [Google Scholar] [CrossRef]
  224. Spekker, E.; Bohár, Z.; Fejes-Szabó, A.; Szűcs, M.; Vécsei, L.; Párdutz, Á. Estradiol Treatment Enhances Behavioral and Molecular Changes Induced by Repetitive Trigeminal Activation in a Rat Model of Migraine. Biomedicines 2022, 10, 3175. [Google Scholar] [CrossRef]
  225. Park, S.Y.; Lee, S.P.; Kim, D.; Kim, W.J. Gut Dysbiosis: A New Avenue for Stroke Prevention and Therapeutics. Biomedicines 2023, 11, 2352. [Google Scholar] [CrossRef]
  226. Cui, Y.; Zhang, H.; Wang, S.; Lu, J.; He, J.; Liu, L.; Liu, W. Stimulated Parotid Saliva Is a Better Method for Depression Prediction. Biomedicines 2022, 10, 2220. [Google Scholar] [CrossRef] [PubMed]
  227. Altamura, C.; Corbelli, I.; de Tommaso, M.; Di Lorenzo, C.; Di Lorenzo, G.; Di Renzo, A.; Filippi, M.; Jannini, T.B.; Messina, R.; Parisi, P.; et al. Pathophysiological Bases of Comorbidity in Migraine. Front. Hum. Neurosci. 2021, 15, 640574. [Google Scholar] [CrossRef] [PubMed]
  228. Schwedt, T.J.; Vargas, B. Neurostimulation for Treatment of Migraine and Cluster Headache. Pain Med. 2015, 16, 1827–1834. [Google Scholar] [CrossRef]
  229. de Albuquerque, L.L.; Pantovic, M.; Clingo, M.; Fischer, K.; Jalene, S.; Landers, M.; Mari, Z.; Poston, B. A Single Application of Cerebellar Transcranial Direct Current Stimulation Fails to Enhance Motor Skill Acquisition in Parkinson’s Disease: A Pilot Study. Biomedicines 2023, 11, 2219. [Google Scholar] [CrossRef] [PubMed]
  230. Senevirathne, D.K.L.; Mahboob, A.; Zhai, K.; Paul, P.; Kammen, A.; Lee, D.J.; Yousef, M.S.; Chaari, A. Deep Brain Stimulation beyond the Clinic: Navigating the Future of Parkinson’s and Alzheimer’s Disease Therapy. Cells 2023, 12, 1478. [Google Scholar] [CrossRef]
  231. Borgomaneri, S.; Battaglia, S.; Sciamanna, G.; Tortora, F.; Laricchiuta, D. Memories are not written in stone: Re-writing fear memories by means of non-invasive brain stimulation and optogenetic manipulations. Neurosci. Biobehav. Rev. 2021, 127, 334–352. [Google Scholar] [CrossRef]
  232. Adeel, M.; Chen, C.-C.; Lin, B.-S.; Chen, H.-C.; Liou, J.-C.; Li, Y.-T.; Peng, C.-W. Safety of Special Waveform of Transcranial Electrical Stimulation (TES): In Vivo Assessment. Int. J. Mol. Sci. 2022, 23, 6850. [Google Scholar] [CrossRef]
  233. Battaglia, S.; Schmidt, A.; Hassel, S.; Tanaka, M. Editorial: Case reports in neuroimaging and stimulation. Front. Psychiatry 2023, 14, 1264669. [Google Scholar] [CrossRef]
  234. Chang, C.H.; Wang, W.L.; Shieh, Y.H.; Peng, H.Y.; Ho, C.S.; Tsai, H.C. Case Report: Low-Frequency Repetitive Transcranial Magnetic Stimulation to Dorsolateral Prefrontal Cortex and Auditory Cortex in a Patient With Tinnitus and Depression. Front. Psychiatry 2022, 13, 847618. [Google Scholar] [CrossRef]
  235. Zakia, H.; Iskandar, S. Case report: Depressive disorder with peripartum onset camouflages suspected intracranial tuberculoma. Front. Psychiatry 2022, 13, 932635. [Google Scholar] [CrossRef]
  236. Nyatega, C.O.; Qiang, L.; Adamu, M.J.; Kawuwa, H.B. Gray matter, white matter and cerebrospinal fluid abnormalities in Parkinson’s disease: A voxel-based morphometry study. Front. Psychiatry 2022, 13, 1027907. [Google Scholar] [CrossRef] [PubMed]
  237. Rymaszewska, J.; Wieczorek, T.; Fila-Witecka, K.; Smarzewska, K.; Weiser, A.; Piotrowski, P.; Tabakow, P. Various neuromodulation methods including Deep Brain Stimulation of the medial forebrain bundle combined with psychopharmacotherapy of treatment-resistant depression-Case report. Front. Psychiatry 2023, 13, 1068054. [Google Scholar] [CrossRef] [PubMed]
  238. Liu, M.; Xie, X.; Xie, J.; Tian, S.; Du, X.; Feng, H.; Zhang, H. Early-onset Alzheimer’s disease with depression as the first symptom: A case report with literature review. Front. Psychiatry 2023, 14, 1192562. [Google Scholar] [CrossRef] [PubMed]
  239. Kim, B.H.; Kim, S.H.; Han, C.; Jeong, H.G.; Lee, M.S.; Kim, J. Antidepressant-induced mania in panic disorder: A single-case study of clinical and functional connectivity characteristics. Front. Psychiatry 2023, 14, 1205126. [Google Scholar] [CrossRef]
  240. Zhou, J.; Cao, Y.; Deng, G.; Fang, J.; Qiu, C. Transient splenial lesion syndrome in bipolar-II disorder: A case report highlighting reversible brain changes during hypomanic episodes. Front. Psychiatry 2023, 14, 1219592. [Google Scholar] [CrossRef]
  241. Logan, S.; Arzua, T.; Canfield, S.G.; Seminary, E.R.; Sison, S.L.; Ebert, A.D.; Bai, X. Studying Human Neurological Disorders Using Induced Pluripotent Stem Cells: From 2D Monolayer to 3D Organoid and Blood Brain Barrier Models. Compr. Physiol. 2019, 9, 565–611. [Google Scholar] [CrossRef]
  242. Gazerani, P. Human Brain Organoids in Migraine Research: Pathogenesis and Drug Development. Int. J. Mol. Sci. 2023, 24, 3113. [Google Scholar] [CrossRef]
  243. Jalink, P.; Caiazzo, M. Brain Organoids: Filling the Need for a Human Model of Neurological Disorder. Biology 2021, 10, 740. [Google Scholar] [CrossRef]
  244. Zhang, D.Y.; Song, H.; Ming, G.L. Modeling neurological disorders using brain organoids. Semin. Cell Dev. Biol. 2021, 111, 4–14. [Google Scholar] [CrossRef]
Figure 1. The amino acid sequence alignment analysis of the main secretin family peptides. Those amino acids with matching hues are identical amino acid sequences. The alignment similarity between peptides is displayed as a percentage next to the brackets.
Figure 1. The amino acid sequence alignment analysis of the main secretin family peptides. Those amino acids with matching hues are identical amino acid sequences. The alignment similarity between peptides is displayed as a percentage next to the brackets.
Cells 12 02649 g001
Figure 2. PACAP receptors signaling to ERK activation. AC, adenylate cylase; ATP: adenosine monophosphate; cAMP: cyclic adenosine monophosphate; DAG: diacylglycerol; ERK, extracellular signal-regulated kinase; Gs and Gq: stimulatory G protein; MEK: mitogen-activated protein kinase kinase; PKA: protein kinase A; PKC: protein kinase C; PACAP: pituitary adenylate cyclase-activating polypeptide; PAC1: PACAP 1 receptor; PIP2: phosphatidylinositol bisphosphate; VPAC1: vasoactive intestinal peptide receptor type 1; VPAC2: vasoactive intestinal peptide receptor type 2.
Figure 2. PACAP receptors signaling to ERK activation. AC, adenylate cylase; ATP: adenosine monophosphate; cAMP: cyclic adenosine monophosphate; DAG: diacylglycerol; ERK, extracellular signal-regulated kinase; Gs and Gq: stimulatory G protein; MEK: mitogen-activated protein kinase kinase; PKA: protein kinase A; PKC: protein kinase C; PACAP: pituitary adenylate cyclase-activating polypeptide; PAC1: PACAP 1 receptor; PIP2: phosphatidylinositol bisphosphate; VPAC1: vasoactive intestinal peptide receptor type 1; VPAC2: vasoactive intestinal peptide receptor type 2.
Cells 12 02649 g002
Table 1. The secretin family peptides, their receptors, and their binding affinity.
Table 1. The secretin family peptides, their receptors, and their binding affinity.
PeptidesReceptors
CGRPCLR
PACAP1–38>>PAC1, <VPAC1, <VPAC2
PACAP6–38?
PACAP1–27>PAC1, <VPAC1, <VPAC2
PRP?
VIP>VPAC1, >VPAC2, <PAC1
CGRP: calcitonin gene-related peptide; PACAP: pituitary adenylate cyclase-activating polypeptide: PRP: PACAP-related peptide; VIP: vasoactive intestinal peptide; CLR: calcitonin receptor-like receptor; PAC1: pituitary adenylate cyclase-activating polypeptide type I; VPAC: vasoactive intestinal peptide receptor; ?: unknown; >>: much higher; >: higher; <: lower.
Table 2. Preclinical findings of PACAP receptor antagonists.
Table 2. Preclinical findings of PACAP receptor antagonists.
AntagonistsCharacteristicsRef.
PACAP6–38PAC1 receptor antagonist, affinity for VPAC2 receptors[144]
N-stearyl-[Nle17] neurotensin-(6–11)/VIP-(7–28)VIP2/PACAP receptor antagonist, mitogen for the keratinocytic cell line and can increase AC activity[145,146]
Maxadilan mutantsPAC1 receptor antagonists, increased AC activity[147,148]
PG 97-269selective PAC1 receptor antagonists[146]
Table 3. Pituitary adenylate cyclase-activating polypeptide (PACAP) monoclonal antibodies under clinical trials.
Table 3. Pituitary adenylate cyclase-activating polypeptide (PACAP) monoclonal antibodies under clinical trials.
ClinicalTrials.gov IdentifierMonoclonal AntibodyTargetStatusRef.
NCT03238781AMG 301receptorNo benefit over placebo for migraine prevention[160,161]
NCT05133323Lu AG09222ligandNo results posted; the press release announced a decrease in the number of migraine days per month[162,163]
NCT04498910LY3451838receptorNo results posted[164]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Tanaka, M.; Szabó, Á.; Körtési, T.; Szok, D.; Tajti, J.; Vécsei, L. From CGRP to PACAP, VIP, and Beyond: Unraveling the Next Chapters in Migraine Treatment. Cells 2023, 12, 2649. https://doi.org/10.3390/cells12222649

AMA Style

Tanaka M, Szabó Á, Körtési T, Szok D, Tajti J, Vécsei L. From CGRP to PACAP, VIP, and Beyond: Unraveling the Next Chapters in Migraine Treatment. Cells. 2023; 12(22):2649. https://doi.org/10.3390/cells12222649

Chicago/Turabian Style

Tanaka, Masaru, Ágnes Szabó, Tamás Körtési, Délia Szok, János Tajti, and László Vécsei. 2023. "From CGRP to PACAP, VIP, and Beyond: Unraveling the Next Chapters in Migraine Treatment" Cells 12, no. 22: 2649. https://doi.org/10.3390/cells12222649

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop