Nothing Special   »   [go: up one dir, main page]

You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (140)

Search Parameters:
Keywords = PACAP

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
38 pages, 2505 KiB  
Review
Sudden Unexpected Death in Epilepsy: Central Respiratory Chemoreception
by Ayse S. Dereli, Auriane Apaire and Riem El Tahry
Int. J. Mol. Sci. 2025, 26(4), 1598; https://doi.org/10.3390/ijms26041598 - 13 Feb 2025
Abstract
Sudden unexpected death in epilepsy (SUDEP) is a critical concern for individuals suffering from epilepsy, with respiratory dysfunction playing a significant role in its pathology. Fatal seizures are often characterized by central apnea and hypercapnia (elevated CO2 levels), indicating a failure in [...] Read more.
Sudden unexpected death in epilepsy (SUDEP) is a critical concern for individuals suffering from epilepsy, with respiratory dysfunction playing a significant role in its pathology. Fatal seizures are often characterized by central apnea and hypercapnia (elevated CO2 levels), indicating a failure in ventilatory control. Research has shown that both human epilepsy patients and animal models exhibit a reduced hypercapnic ventilatory response in the interictal (non-seizure) period, suggesting an impaired ability to regulate breathing in response to high CO2 levels. This review examines the role of central chemoreceptors—specifically the retrotrapezoid nucleus, raphe nuclei, nucleus tractus solitarius, locus coeruleus, and hypothalamus in this pathology. These structures are critical for sensing CO2 and maintaining respiratory homeostasis. Emerging evidence also implicates neuropeptidergic pathways within these chemoreceptive regions in SUDEP. Neuropeptides like galanin, pituitary adenylate cyclase-activating peptide (PACAP), orexin, somatostatin, and bombesin-like peptides may modulate chemosensitivity and respiratory function, potentially exacerbating respiratory failure during seizures. Understanding the mechanisms linking central chemoreception, respiratory control, and neuropeptidergic signaling is essential to developing targeted interventions to reduce the risk of SUDEP in epilepsy patients. Full article
(This article belongs to the Special Issue Epilepsy: From Molecular Basis to Therapy)
Show Figures

Figure 1

Figure 1
<p>Fatal seizures are characterized by central apnea, hypoxia, and hypercapnia, and epilepsy is associated with an ablated hypercapnic ventilatory response (HCVR). (<b>a</b>) Percentage oxygen saturation (%O<sub>2</sub>, blue trace) decreases, and partial pressure of CO<sub>2</sub> (Pco<sub>2</sub>, red trace) increases during a seizure (pink box). Elevated Pco<sub>2</sub> persists for up to 4 h after the seizure, adapted from [<a href="#B37-ijms-26-01598" class="html-bibr">37</a>,<a href="#B45-ijms-26-01598" class="html-bibr">45</a>]. (<b>b</b>) A recovery seizure with apnea is followed by an increase in heart rate, adapted from [<a href="#B38-ijms-26-01598" class="html-bibr">38</a>]. (<b>c</b>) A terminal seizure, in which respiratory arrest is followed by secondary bradycardia, is shown, adapted from [<a href="#B45-ijms-26-01598" class="html-bibr">45</a>]. (<b>d</b>) HCVR is reduced in epileptic conditions, with more severe ablation observed in the presence of risk factors such as postictal timing, late-stage epileptogenesis, genetic factors, and nocturnal seizures. Summary trace gathered from [<a href="#B58-ijms-26-01598" class="html-bibr">58</a>,<a href="#B59-ijms-26-01598" class="html-bibr">59</a>,<a href="#B60-ijms-26-01598" class="html-bibr">60</a>,<a href="#B61-ijms-26-01598" class="html-bibr">61</a>].</p>
Full article ">Figure 2
<p>A schematic summary of the cardiorespiratory symptoms and the contribution of central chemoreception in the SUDEP cascade. Repetitive seizures induce alterations in the central chemoreceptive centers, leading to impaired chemoreflex. This dysfunction contributes to central apnea, exacerbating hypoxia and hypercapnia. Hypoxia is proposed to cause cardiac asystole, culminating in SUDEP.</p>
Full article ">Figure 3
<p>Schema depicting the role of central chemoreception and its dysfunction in the context of epilepsy, highlighting the involvement of neuropeptides. Central chemoreceptors, including the hypothalamus, and key brainstem regions, including the RTN, LC, medullary raphe, and caudal NTS, are activated in response to CO<sub>2</sub> (blue arrows). Signals from central chemoreceptors project to the VRC, which sends information to the phrenic nucleus, which drives increased ventilation in response (neuronal projections represented by purple arrows). Central chemoreceptors contain neuropeptides: galanin, PACAP, orexin, somatostatin, NMB, and GRP, which are involved in the mechanism of central chemoreception. In healthy individuals (green box), central chemoreceptors respond to increased levels of PCO<sub>2</sub> by enhancing respiratory drive, maintaining normocapnia. However, in epilepsy (red boxes), the impaired activation of these chemoreceptors can lead to reduced ventilatory responses, resulting in hypercapnia and an increased risk of central apnea, which is strongly associated with SUDEP (associations are indicated by black arrows). Abbreviations: LC: locus coeruleus, NTS: nucleus tractus solitarius, PACAP: pituitary adenylate cyclase-activating polypeptide, NMB: neuromedin B (NMB) and GRP: gastrin releasing peptide, PCO<sub>2</sub>: partial pressure of carbon dioxide, RTN: retrotrapezoid nucleus, SUDEP: sudden unexpected death in epilepsy, VRC: ventral respiratory column. Created with BioRender.</p>
Full article ">
13 pages, 3000 KiB  
Article
The Effect of GO Flake Size on Field-Effect Transistor (FET)-Based Biosensor Performance for Detection of Ions and PACAP 38
by Seungjun Lee, Jongdeok Park, Jaeyoon Song, Jae-Joon Lee and Jinsik Kim
Biosensors 2025, 15(2), 86; https://doi.org/10.3390/bios15020086 - 5 Feb 2025
Abstract
The performance development of rGO-FET biosensors by analyzing the influence of GO flake size on biosensing efficacy. GO flakes of varying sizes, from 1 µm to 20 µm, were prepared under controlled conditions, followed by characterization through SEM and XPS to evaluate their [...] Read more.
The performance development of rGO-FET biosensors by analyzing the influence of GO flake size on biosensing efficacy. GO flakes of varying sizes, from 1 µm to 20 µm, were prepared under controlled conditions, followed by characterization through SEM and XPS to evaluate their size, surface area, and C/O ratio. The biosensing performance was systematically assessed by rGO-FET biosensors, examining the effects of GO flake size, C/O ratio, and film thickness. PACAP38 was employed as a biomarker for receptor-mediated detection, while chlorine ions served as model analytes for receptor-free small molecule detection. The results indicate that decreasing the GO flake size enhanced the performance for both target biomolecules. These findings highlight the crucial importance of selecting GO flake sizes specific to target analytes and detection strategies, thereby optimizing biosensor efficiency. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic showing the utilization of ≈1, ≈10, and ≈20 μm size of graphene flakes to verify rGO-FET biosensor sensitivity differences by controlling the graphene flake size.</p>
Full article ">Figure 2
<p>Size analysis of as-prepared GO flakes. (<b>A</b>,<b>B</b>) show the size and area distributions of the ≈10 and ≈20 μm GO flakes, respectively. The scale bar of each SEM image is 10 μm. (<b>C</b>) Morphological characteristics of prepared GO flakes. Variation values of the major axis, minor axis, area, and aspect ratio of the GO flakes.</p>
Full article ">Figure 3
<p>PACAP38 and chlorine detection using a controlled GO flake. (<b>A</b>) Resistance changes according to receptor immobilization depending on the GO flake size. (<b>B</b>) PACAP38 detection in the concentration range of 10 pg/mL~10 ng/mL for GO flakes of different sizes. (<b>C</b>) Linear slope of chlorine detection within a certain concentration range. (<b>D</b>) PACAP38 and chlorine detection slope of the linear function analysis with ≈1, ≈10, and ≈20 μm size of GO flakes.</p>
Full article ">Figure 4
<p>PACAP38 and chlorine detection analysis by GO flake surface area and C/O ratio. (<b>A</b>) and (<b>B</b>) Normalized PACAP38 detection results by surface area and C/O ratio of GO flakes, respectively. (<b>C</b>) Normalized chlorine detection results based on the GO flake surface area. (<b>D</b>) The sensitivity from the normalized result of each GO flake.</p>
Full article ">Figure 5
<p>PACAP38 and chlorine detection with controlled GO flake 30 depositions. (<b>A</b>) Resistance changes according to receptor immobilization depending on GO flake size. (<b>B</b>) PACAP38 detection in the concentration range of 10~100 ng/mL in GO flakes of different sizes. (<b>C</b>) The linear function slope of chlorine detection in a certain range of concentrations. (<b>D</b>) PACAP38 and chlorine detection slope of linear function analysis with ≈1, ≈10, and ≈20 μm size of GO flakes.</p>
Full article ">Figure 6
<p>Thirty depositions condition of the graphene biosensor for PACAP38 and chlorine detection analysis based on GO flake surface area and C/O ratio. (<b>A</b>,<b>B</b>) Normalized PACAP38 detection result by surface area and C/O ratio of GO flake, respectively. (<b>C</b>) Normalized chlorine detection results for GO flake surface area. (<b>D</b>) The sensitivity from the normalized result of each GO flake.</p>
Full article ">Figure 7
<p>Analysis of the effect of the graphene C/O ratio or surface area through sensitivity derived from the measured values of PACAP38 and chlorine measured in 20 and 30 depositions condition sensors implemented using graphene flakes of sizes ≈1, ≈10, and ≈20 μm. The <span class="html-italic">p</span>-values based on the number of depositions and flake sizes were determined using <span class="html-italic">t</span>-tests and are represented as follows: *** <span class="html-italic">p</span> ≤ 0.001, ** <span class="html-italic">p</span> ≤ 0.01, * <span class="html-italic">p</span> ≤ 0.05, and ns (not significant) for <span class="html-italic">p</span> &gt; 0.05. (<b>A</b>,<b>B</b>) Analysis of the sensitivity values obtained by measuring PACAP38 in 20 and 30 deposition sensors as surface area and C/O ratio. (<b>C</b>,<b>D</b>) Sensitivity values obtained by measuring chlorine as surface area and C/O ratio analyzed by value.</p>
Full article ">
27 pages, 4623 KiB  
Review
Antimicrobial Neuropeptides and Their Receptors: Immunoregulator and Therapeutic Targets for Immune Disorders
by Kaiqi Chen, Xiaojun Wu, Xiaoke Li, Haoxuan Pan, Wenhui Zhang, Jinxi Shang, Yinuo Di, Ruonan Liu, Zhaodi Zheng and Xitan Hou
Molecules 2025, 30(3), 568; https://doi.org/10.3390/molecules30030568 - 27 Jan 2025
Abstract
The interaction between the neuroendocrine system and the immune system plays a key role in the onset and progression of various diseases. Neuropeptides, recognized as common biochemical mediators of communication between these systems, are receiving increasing attention because of their potential therapeutic applications [...] Read more.
The interaction between the neuroendocrine system and the immune system plays a key role in the onset and progression of various diseases. Neuropeptides, recognized as common biochemical mediators of communication between these systems, are receiving increasing attention because of their potential therapeutic applications in immune-related disorders. Additionally, many neuropeptides share significant similarities with antimicrobial peptides (AMPs), and evidence shows that these antimicrobial neuropeptides are directly involved in innate immunity. This review examines 10 antimicrobial neuropeptides, including pituitary adenylate cyclase-activating polypeptide (PACAP), vasoactive intestinal peptide (VIP), α-melanocyte stimulating hormone (α-MSH), ghrelin, adrenomedullin (AM), neuropeptide Y (NPY), urocortin II (UCN II), calcitonin gene-related peptide (CGRP), substance P (SP), and catestatin (CST). Their expression characteristics and the immunomodulatory mechanisms mediated by their specific receptors are summarized, along with potential drugs targeting these receptors. Future studies should focus on further investigating antimicrobial neuropeptides and advancing the development of related drugs in preclinical and/or clinical studies to improve the treatment of immune-related diseases. Full article
(This article belongs to the Section Medicinal Chemistry)
Show Figures

Figure 1

Figure 1
<p>Bidirectional communication between the neuroendocrine and immune system. An intricate interplay exists between the neuroendocrine system and the immune system. Firstly, pathogens can induce immune cells to produce cytokines and nerve cells to release neuromediators, thereby facilitating the exchange of these chemical signals between immune cells and nerve cells. Secondly, pathogens can also stimulate other cell types, such as epithelial cells, leading to the secretion of effector molecules including cytokines, chemokines, and growth factors. These factors can further activate immune cells and nerve cells through receptor-mediated responses to pathogen stimulation. Additionally, the involvement of the hypothalamic–autonomic nervous system axis (HANS) should be considered. For instance, the brain–gut axis has been shown to trigger hypothalamus–pituitary–adrenal function within the central nervous system (CNS). The diagram of the immune cells was refined based on Krause’s work [<a href="#B18-molecules-30-00568" class="html-bibr">18</a>]. The diagram of the brain, pathogens, and nerve cells was designed by pch.vector/Freepik.</p>
Full article ">Figure 2
<p>Antimicrobial neuropeptides are key players in anti-inflammation. Certain neuropeptides synthesized by both neuronal and immune cells effectively respond to pathogen stimuli and mediate anti-inflammatory effects. Firstly, neuropeptides can modulate T cell function by inhibiting the production of TH1-related factor IL-12, inducing CD86 expression, and promoting the generation and differentiation of TH2 cells. Additionally, neuropeptides can induce Treg cell production while suppressing autoreactive T cell activation through secretion of IL-10 and TGFβ. Secondly, neuropeptides directly influence macrophages by promoting their differentiation into the M2 phenotype. This enhances the expression of anti-inflammatory factors while suppressing pro-inflammatory factor expression. The red “X” means inhibit. The diagram of the immune cells was refined based on Krause’s work [<a href="#B18-molecules-30-00568" class="html-bibr">18</a>].</p>
Full article ">Figure 3
<p>Antimicrobial neuropeptides are key players in pro-inflammation. Certain neuropeptides play a pivotal role in mediating neurogenic inflammation and augmenting inflammatory responses. In the presence of inflammatory stimuli, mast cells undergo degranulation, and through the action of neuropeptides, they elicit arterial dilation and heightened venous permeability, thereby establishing an environment conducive to inflammatory processes. Moreover, mast cell-derived tryptase possesses the ability to cleave activated PAR-2, thereby facilitating the release of neuropeptides and instigating the inflammatory response. The diagram of the immune cells was refined based on Krause’s work [<a href="#B18-molecules-30-00568" class="html-bibr">18</a>]. The diagram of the nerve cell was designed by pch.vector/Freepik.</p>
Full article ">
12 pages, 1046 KiB  
Article
CamKIIα and VPAC1 Expressions in the Caudal Trigeminal Nucleus of Rats After Systemic Nitroglycerin Treatment: Interaction with Anandamide
by Gábor Nagy-Grócz, Eleonóra Spekker, Tamás Körtési, Klaudia Flóra Laborc, Zsuzsanna Bohár, Annamária Fejes-Szabó, László Vécsei and Árpád Párdutz
Life 2025, 15(2), 155; https://doi.org/10.3390/life15020155 - 22 Jan 2025
Viewed by 404
Abstract
Migraines are a frequently occurring neurological condition that affects up to 16% of the global population. The precise pathomechanism of the disease remains unknown, but from animal and human observations, it appears that calcium/calmodulin-dependent protein kinase II alpha (CamKIIα), pituitary adenylate cyclase-activating polypeptide [...] Read more.
Migraines are a frequently occurring neurological condition that affects up to 16% of the global population. The precise pathomechanism of the disease remains unknown, but from animal and human observations, it appears that calcium/calmodulin-dependent protein kinase II alpha (CamKIIα), pituitary adenylate cyclase-activating polypeptide (PACAP), and vasoactive intestinal polypeptide (VIP) are involved in its pathogenesis. One of the animal models of migraines uses the systemic administration of nitroglycerin (NTG), which, as a nitric oxide (NO) donor, initiates a self-amplifying process in the trigeminal system, leading to central sensitization. Endocannabinoids, such as anandamide (AEA), are thought to play a modulatory role in trigeminal activation and sensitization phenomena. In the present experiment, we aimed to investigate the effect of NTG and AEA on CamKIIα, PACAP/VIP, and vasoactive intestinal polypeptide type 1 receptor (VPAC1) expression levels in the upper cervical spinal cord (C1-C2) of rats, where trigeminal nociceptive afferents are clustered. Four groups of animals were formed: in the first group, the rats received only the vehicle; in the second group, they were treated with an intraperitoneal injection of NTG (10 mg/kg); animals in the third and fourth groups received AEA (2 × 5 mg/kg) half an hour before and one hour after the placebo or treatment with NTG. Four hours after the placebo/NTG injection, the animals were transcardially perfused, and the cervical spinal cords were removed for Western blot. Our results show that both NTG and AEA alone can increase the expression of CamKIIα and VPAC1 in the C1-C2 segments. Interestingly, the combination of NTG and AEA had no such effect on these markers, possibly due to various negative feedback mechanisms. Full article
(This article belongs to the Special Issue Migraine and Headache: From Pathophysiological Aspects)
Show Figures

Figure 1

Figure 1
<p>Effect of NTG and AEA on the expression of CamKIIα, Western blot data. (<b>A</b>). Western blotting of CamKIIα and GAPDH expression in the C1-C2. (<b>B</b>). Densitometry of the individual bands showed that in NTG- and AEA-treated animals, the expression of CamKIIα was significantly higher compared to the placebo group. No such effect was observed in the NTG + AEA-injected animals. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 2
<p>Effect of NTG and AEA on the expression of VPAC1, Western blot data. (<b>A</b>). Western blotting of VPAC1 and GAPDH expression in the C1-C2. (<b>B</b>). Quantitative data demonstrate, that the relative optical density of VPAC1 in the NTG and AEA groups is significantly higher than in the placebo group. NTG + AEA mitigated this effect. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p>Possible mechanisms of NTG and AEA on the CamKIIα and VPAC1 expression. Both NTG and AEA can increase the expression of CamKIIα and VPAC1, probably by generating NO production. NO can initiate the phosphorylation of CamKIIα and can activate adenylate cyclase, which also triggers the production of PACAP. In addition to this, AEA also promotes the formation of NO by the CB1 receptor. On the other hand, the combined treatment might cause a huge boost in NO production, which activates a negative feedback mechanism and decreases the levels of NO in the long run. Also, it is well known that AEA can also reduce the synthesis of NO via the CB2 receptor.</p>
Full article ">
12 pages, 5281 KiB  
Article
PAC1 Agonist Maxadilan Reduces Atherosclerotic Lesions in Hypercholesterolemic ApoE-Deficient Mice
by Lilli Mey, Gabriel A. Bonaterra, Joy Hoffmann, Hans Schwarzbach, Anja Schwarz, Lee E. Eiden, Eberhard Weihe and Ralf Kinscherf
Int. J. Mol. Sci. 2024, 25(24), 13245; https://doi.org/10.3390/ijms252413245 - 10 Dec 2024
Viewed by 569
Abstract
A possible involvement of immune- and vasoregulatory PACAP signaling at the PAC1 receptor in atherogenesis and plaque-associated vascular inflammation has been suggested. Therefore, we tested the PAC1 receptor agonist Maxadilan and the PAC1 selective antagonist M65 on plaque development and lumen stenosis in [...] Read more.
A possible involvement of immune- and vasoregulatory PACAP signaling at the PAC1 receptor in atherogenesis and plaque-associated vascular inflammation has been suggested. Therefore, we tested the PAC1 receptor agonist Maxadilan and the PAC1 selective antagonist M65 on plaque development and lumen stenosis in the ApoE−/− atherosclerosis model for possible effects on atherogenesis. Adult male ApoE−/− mice were fed a cholesterol-enriched diet (CED) or standard chow (SC) treated with Maxadilan, M65 or Sham. Effects of treatment on atherosclerotic plaques, lumen stenosis, apoptosis and pro-inflammatory signatures were analyzed in the brachiocephalic trunk (BT). The percentage of Maxadilan treated mice exhibiting plaques under SC and CED was lower than that of Sham or M65 treatment indicating opposite effects of Maxadilan and M65. Maxadilan application inhibited lumen stenosis in SC and CED mice compared to the Sham mice. In spite of increased cholesterol levels, lumen stenosis of Maxadilan-treated mice was similar under CED and SC. In contrast, M65 under SC or CED did not reveal a significant influence on lumen stenosis. Maxadilan significantly reduced the TNF-α-immunoreactive (TNF-α+) area in the plaques under CED, but not under SC. In contrast, the IL-1β+ area was reduced after Maxadilan treatment in SC mice but remained unchanged in CED mice compared to Sham mice. Maxadilan reduced caspase-3 immunoreactive (caspase-3+) in the tunica media under both, SC and CED without affecting lipid content in plaques. Despite persistent hypercholesterolemia, Maxadilan reduces lumen stenosis, apoptosis and TNF-α driven inflammation. Our data suggest that Maxadilan provides atheroprotection by acting downstream of hypercholesterolemia-induced vascular inflammation. This implicates the potential of PAC1-specific agonist drugs against atherosclerosis even beyond statins and PCSK9 (proprotein convertase subtilisin/kexin type 9) inhibitors. Full article
(This article belongs to the Special Issue Atherosclerosis: From Molecular Basis to Therapy)
Show Figures

Figure 1

Figure 1
<p>Percentage of mice exhibiting macroscopically visible atherosclerotic plaques in the ascending aorta (aao), descending aorta (dao), aortic arch (aa), and branches of Sham and Maxadilan-treated ApoE<sup>−/−</sup> mice under standard chow (SC) or cholesterol-enriched diet (CED): Brachiocephalic trunk (BT), right/left carotid artery (rca/lca), right/left subclavian artery (rsca/lsca). Intraluminal contrast staining with methylene blue enhances the visibility of white atherosclerosis plaques, indicated by black arrows. For statistical significance, see <span class="html-italic">p</span> values in the individual micrographs. The numbers in parentheses denote the numbers of animals.</p>
Full article ">Figure 2
<p>Lumen stenosis and plaque development in the brachiocephalic trunk (BT)of ApoE<sup>−/−</sup> mice at the age of 20 weeks, fed with standard chow (SC) or cholesterol-enriched diet (CED). Treated mice received PAC1 agonist Maxadilan (20 nmol/kg) or PAC1 antagonist M65 (20 nmol/kg), dissolved in physiological saline solution i.p., Sham groups received physiological saline solution. Cross sections of areas with maximum plaque sizes were prepared and stained with hematoxylin/eosin. Lumen stenosis was measured in %; data are provided as mean ± SEM. For statistical significance, see <span class="html-italic">p</span> values in the individual micrographs.</p>
Full article ">Figure 3
<p>Immunohistochemistry and quantification of the inflammatory markers TNF-α and IL-1β in the plaque and tunica media of BT in ApoE<sup>−/−</sup> mice under SC and CED, after Maxadilan treatment or Sham (<b>A</b>). The percentages of immunohistochemically positive-stained areas per plaque or media were evaluated. Black arrowheads indicate immunoreactive positive cells. Data (<b>B</b>) TNF-α and (<b>C</b>) IL-1β are provided as mean + SEM (n = 7–9). * <span class="html-italic">p</span> ≤ 0.05 vs. Sham under SC or CED. Scale bar: 100 µm.</p>
Full article ">Figure 4
<p>Immunohistochemistry and quantification of immunoreactive areas (in %) in relation to the whole plaque or media of cross sections of BT of ApoE<sup>−/−</sup> mice under SC or CED after Maxadilan treatment or Sham (<b>A</b>). Antibodies were directed against cyclooxygenase-2 (COX-2) and cleaved caspase-3 (cl. casp.3, apoptosis). Black arrowheads indicate immunoreactive positive cells. Data (<b>B</b>) COX-2 and (<b>C</b>) cleaved caspase-3 are provided as mean + SEM (n = 7–9). ** <span class="html-italic">p</span> ≤ 0.01 vs. Sham under SC; <sup>#</sup> <span class="html-italic">p</span> ≤ 0.05 <sup>###</sup> <span class="html-italic">p</span> ≤ 0.001 vs. Sham under CED. Scale bar: 100 µm.</p>
Full article ">Figure 5
<p>ORO (on the left) and CD68 (on the right) (immuno)histochemistry of atherosclerotic BT cross-sections of ApoE<sup>−/−</sup> mice under Maxadilan treatment or Sham SC or CED. Oil red positive lipid deposits are shown in red. (<b>A</b>,<b>B</b>) Lipid-loaden MΦ in the plaque-forming foam cells (black arrowheads) are distinguishable from intimal cells, smooth muscle cells of the tunica media with small lipid droplets (arrows) and cells of perivascular fat tissue showing distinct fat vacuoles. (<b>C</b>) CD68<sup>+</sup> Mo and MΦ (right side, white arrowheads) stained with HRP-DAB in brown. Data are provided as mean + SEM (n = 7–9). Diagrams show % of lipid or Mo/MΦ positive area in relation to the whole plaque area. Intima (i), media (m) and adventitia (a). Scale bar: 100 µm.</p>
Full article ">
2 pages, 1567 KiB  
Correction
Correction: Giunta et al. Ameliorative Effects of PACAP against Cartilage Degeneration. Morphological, Immunohistochemical and Biochemical Evidence from in Vivo and in Vitro Models of Rat Osteoarthritis. Int. J. Mol. Sci. 2015, 16, 5922–5944
by Salvatore Giunta, Alessandro Castorina, Rubina Marzagalli, Marta Anna Szychlinska, Karin Pichler, Ali Mobasheri and Giuseppe Musumeci
Int. J. Mol. Sci. 2024, 25(22), 12148; https://doi.org/10.3390/ijms252212148 - 12 Nov 2024
Viewed by 598
Abstract
In the original publication [...] Full article
(This article belongs to the Section Biochemistry)
Show Figures

Figure 2

Figure 2
<p>Histological and histochemical evaluation. (<b>A</b>,<b>B</b>) Histology (H&amp;E staining) demonstrated the absence of structural alterations in control groups (without anterior cruciate ligament transection (ACLT)). In the superficial zone, cells appear flat and small; in the middle and deep zone, cells are organized in columns. Magnification ×20; Scale bars: 100 µm; (<b>C</b>) Histology (H&amp;E staining) demonstrated evidence of structural alterations in cartilage with moderate signs of OA (with ACLT). The structural alterations included a reduction of cartilage thickness in the superficial and the middle zones. The tidemark is no longer intact and the subchondral bone shows fibrillation. Magnification ×20; Scale bars: 100 µm; (<b>D</b>) Histology (H&amp;E staining) demonstrated signs of structural alterations in severe Osteoarthritis (OA) (with ACLT). Severe OA cartilage shows deep surface clefts, disappearance of cells from the superficial zone, cloning, and a lack of cells in the intermediate and deep zone, which are not arranged in columns. The cartilage layers (superficial zone, middle and deep zone) are completely absent. Magnification ×20; Scale bars: 100 µm; (<b>E</b>,<b>F</b>) Histochemistry (toluidine blue staining) showed an absence of structural alterations and preserved GAG, in control groups (without ACLT), as indicated by the intense toluidine blue staining. Magnification ×20; Scale bars: 100 µm; (<b>G</b>) Histochemistry (toluidine blue staining) demonstrated signs of structural alterations in moderate and severe OA cartilage and loss of proteoglycans as evidenced by poor GAG preservation in the OA group (with ACLT), showing reduced toluidine blue staining. Magnification ×20; Scale bars: 100 µm.</p>
Full article ">
14 pages, 2063 KiB  
Article
Effects of PACAP Deficiency on Immune Dysfunction and Peyer’s Patch Integrity in Adult Mice
by Jason Sparks, Matyas Meggyes, Lilla Makszin, Viktoria Jehn, Hedvig Lugosi, Dora Reglodi and Laszlo Szereday
Int. J. Mol. Sci. 2024, 25(19), 10676; https://doi.org/10.3390/ijms251910676 - 3 Oct 2024
Viewed by 1277
Abstract
PACAP (pituitary adenylate cyclase activating polypeptide) is a widespread neuropeptide with cytoprotective and anti-inflammatory effects. It plays a role in innate and adaptive immunity, but data are limited about gut-associated lymphoid tissue. We aimed to reveal differences in Peyer’s patches between wild-type (WT) [...] Read more.
PACAP (pituitary adenylate cyclase activating polypeptide) is a widespread neuropeptide with cytoprotective and anti-inflammatory effects. It plays a role in innate and adaptive immunity, but data are limited about gut-associated lymphoid tissue. We aimed to reveal differences in Peyer’s patches between wild-type (WT) and PACAP-deficient (KO) mice. Peyer’s patch morphology from young (3-months-old) and aging (12–15-months-old) mice was examined, along with flow cytometry to assess immune cell populations, expression of checkpoint molecules (PD-1, PD-L1, TIM-3, Gal-9) and functional markers (CD69, granzyme B, perforin) in CD3+, CD4+, and CD8+ T cells. We found slight differences between aging, but not in young, WT, and KO mice. In WT mice, aging reduced CD8+ T cell numbers frequency and altered checkpoint molecule expression (higher TIM-3, granzyme B; lower Gal-9, CD69). CD4+ T cell frequency was higher with similar checkpoint alterations, indicating a regulatory shift. In PACAP KO mice, aging did not change cell population frequencies but led to higher TIM-3, granzyme B and lower PD-1, PD-L1, Gal-9, and CD69 expression in CD4+ and CD8+ T cells, with reduced overall T cell activity. Thus, PACAP deficiency impacts immune dysfunction by altering checkpoint molecules and T cell functionality, particularly in CD8+ T cells, suggesting complex immune responses by PACAP, highlighting its role in intestinal homeostasis and potential implications for inflammatory bowel diseases. Full article
(This article belongs to the Special Issue Molecular Biology of Senescence and Anti-Aging Strategies)
Show Figures

Figure 1

Figure 1
<p>Gating strategy to identify CD3+ T, CD8+ T, CD4+, and CD4+/CD8+ T cell populations. Flow cytometric analyses for determining lymphocyte subpopulations. Following a two-step doublet exclusion, the lymphocyte population was gated using FSC-A/SSC-A parameters. From the lymphogate, CD3+ T, CD8+ T, CD4+, and CD4+/CD8+ T cell subpopulations were detected.</p>
Full article ">Figure 2
<p>PD-1 expression by CD4+/CD8+ T-cells in young and aging WT and PACAP KO mice. The surface expression of PD-1 receptor by CD4+/CD8+ T-cells in young and aging WT and PACAP KO mice. The solid bars represent medians of 11, 13, 10, and 10 determinations, respectively. The boxes indicate the interquartile ranges, and the whiskers represent the variability of the minimum, maximum, and any outlier data points in comparison to the interquartile range. The middle square within the box represents the mean value. Significant differences with <span class="html-italic">p</span>-values &lt; 0.05 ** &lt; 0.001 *** are indicated.</p>
Full article ">Figure 3
<p>TIM-3 expression by CD3+, CD4+, and CD8+ T-cells in young and aging WT and PACAP KO mice. The surface expression of TIM-3 receptor by CD3+ (<b>A</b>), CD8+ T (<b>B</b>), and CD4+ (<b>C</b>) cells in young and aging WT and PACAP KO mice. The solid bars represent medians of 10, 10, 10, and 10 determinations, respectively. The boxes indicate the interquartile ranges, and the whiskers represent the variability of the minimum, maximum, and any outlier data points in comparison to the interquartile range. The middle square within the box represents the mean value. Significant differences with <span class="html-italic">p</span>-values &lt; 0.001 *** are indicated.</p>
Full article ">Figure 4
<p>Granzyme B expression by CD3+ T-cells in young and aging WT and PACAP KO mice. The intracellular expression of Granzyme B by CD3+ T-cells in young and aging WT and PACAP KO mice. The solid bars represent medians of 10, 9, 10, and 5 determinations, respectively. The boxes indicate the interquartile ranges, and the whiskers represent the variability of the minimum, maximum, and any outlier data points in comparison to the interquartile range. The middle square within the box represents the mean value. Significant differences with <span class="html-italic">p</span>-values &lt; 0.001 *** are indicated.</p>
Full article ">Figure 5
<p>CD69 expression by CD8+ and CD4+/CD8+ T cells in young and aging WT and PACAP KO mice. The surface expression of CD69 receptor by CD8+ (<b>A</b>) and CD4+/CD8+ (<b>B</b>) T cells in young and aging WT and PACAP KO mice. The solid bars represent medians of 10, 10, 10, and 10 determinations, respectively. The boxes indicate the interquartile ranges, and the whiskers represent the variability of the minimum, maximum, and any outlier data points in comparison to the interquartile range. The middle square within the box represents the mean value. Significant differences with <span class="html-italic">p</span>-values &lt; 0.01 ** &lt; 0.001 *** are indicated.</p>
Full article ">
8 pages, 236 KiB  
Review
Onabotulinumtoxin A for the Treatment of Post-Traumatic Headache: Is It Better than Anti-CGRP Antibodies?
by Lanfranco Pellesi, Dilara Onan and Paolo Martelletti
Toxins 2024, 16(10), 427; https://doi.org/10.3390/toxins16100427 - 2 Oct 2024
Viewed by 1430
Abstract
Post-traumatic headache (PTH) is a common and debilitating consequence of traumatic brain injury (TBI), often resembling migraine and tension-type headaches. Despite its prevalence, the optimal treatment for PTH remains unclear, with current strategies largely extrapolated from other headache disorders. This review evaluates the [...] Read more.
Post-traumatic headache (PTH) is a common and debilitating consequence of traumatic brain injury (TBI), often resembling migraine and tension-type headaches. Despite its prevalence, the optimal treatment for PTH remains unclear, with current strategies largely extrapolated from other headache disorders. This review evaluates the use of onabotulinumtoxin A (ONA) and anti-calcitonin gene-related peptide (CGRP) monoclonal antibodies (mAbs) in the treatment of PTH. A comprehensive literature search was conducted on PubMed, including studies published up to September 2024, focusing on the efficacy, safety, and mechanisms of onabotulinumtoxin A and anti-CGRP mAbs in PTH. Both clinical trials and observational studies were reviewed. ONA, widely recognized for its efficacy in chronic migraine, has shown limited benefits in PTH with only one trial involving abobotulinumtoxin A in a cohort of 40 subjects. A phase 2 trial with fremanezumab, an anti-CGRP monoclonal antibody, failed to demonstrate significant efficacy in PTH, raising questions about the utility of targeting CGRP in this condition. ONA may offer advantages over anti-CGRP mAbs, not only in terms of its broader mechanism of action but also in cost-effectiveness and higher patient adherence. Both ONA and anti-CGRP mAbs are potential options for the management of PTH, but the current evidence is insufficient to establish clear guidelines. The negative results from the fremanezumab trial suggest that CGRP inhibition may not be sufficient for treating PTH, whereas onabotulinumtoxin A’s ability to target multiple pain pathways may make it a more promising candidate. Full article
(This article belongs to the Special Issue Immunogenicity of Botulinum Toxin)
16 pages, 9707 KiB  
Article
Increased Expression of the Neuropeptides PACAP/VIP in the Brain of Mice with CNS Targeted Production of IL-6 Is Mediated in Part by Trans-Signalling
by Alessandro Castorina, Jurgen Scheller, Kevin A. Keay, Rubina Marzagalli, Stefan Rose-John and Iain L. Campbell
Int. J. Mol. Sci. 2024, 25(17), 9453; https://doi.org/10.3390/ijms25179453 - 30 Aug 2024
Viewed by 879
Abstract
Inflammation with expression of interleukin 6 (IL-6) in the central nervous system (CNS) occurs in several neurodegenerative/neuroinflammatory conditions and may cause neurochemical changes to endogenous neuroprotective systems. Pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal polypeptide (VIP) are two neuropeptides with well-established protective [...] Read more.
Inflammation with expression of interleukin 6 (IL-6) in the central nervous system (CNS) occurs in several neurodegenerative/neuroinflammatory conditions and may cause neurochemical changes to endogenous neuroprotective systems. Pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal polypeptide (VIP) are two neuropeptides with well-established protective and anti-inflammatory properties. Yet, whether PACAP and VIP levels are altered in mice with CNS-restricted, astrocyte-targeted production of IL-6 (GFAP-IL6) remains unknown. In this study, PACAP/VIP levels were assessed in the brain of GFAP-IL6 mice. In addition, we utilised bi-genic GFAP-IL6 mice carrying the human sgp130-Fc transgene (termed GFAP-IL6/sgp130Fc mice) to determine whether trans-signalling inhibition rescued PACAP/VIP changes in the CNS. Transcripts and protein levels of PACAP and VIP, as well as their receptors PAC1, VPAC1 and VPAC2, were significantly increased in the cerebrum and cerebellum of GFAP-IL6 mice vs. wild type (WT) littermates. These results were paralleled by a robust activation of the JAK/STAT3, NF-κB and ERK1/2MAPK pathways in GFAP-IL6 mice. In contrast, co-expression of sgp130Fc in GFAP-IL6/sgp130Fc mice reduced VIP expression and activation of STAT3 and NF-κB pathways, but it failed to rescue PACAP, PACAP/VIP receptors and Erk1/2MAPK phosphorylation. We conclude that forced expression of IL-6 in astrocytes induces the activation of the PACAP/VIP neuropeptide system in the brain, which is only partly modulated upon IL-6 trans-signalling inhibition. Increased expression of PACAP/VIP neuropeptides and receptors may represent a homeostatic response of the CNS to an uncontrolled IL-6 synthesis and its neuroinflammatory consequences. Full article
(This article belongs to the Special Issue New Mechanisms and Therapeutics in Neurological Diseases 3.0)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Enhanced PACAP and VIP levels in the cerebrum and cerebellum of GFAP-IL6 mice are partly rescued in bi-genic GFAP-IL6/sgp130Fc mice. PACAP and VIP protein concentrations in lysates from cerebra and cerebella of age-matched six-months-old wild-type (WT), monogenic GFAP-IL6 and bi-genic GFAP-IL6/sgp130Fc mice were determined using commercially available PACAP and VIP mouse ELISA Kits (for details refer to <a href="#sec4-ijms-25-09453" class="html-sec">Section 4</a>). Comparisons among WTs, monogenic and bi-genic mice groups (n = 3 per group) were conducted to assess the levels of PACAP or VIP in the cerebrum (<b>A</b>,<b>B</b>) and cerebellum (<b>C</b>,<b>D</b>). * <span class="html-italic">p</span> &lt; 0.05 or ** <span class="html-italic">p</span> &lt; 0.01 vs. WT. # <span class="html-italic">p</span> &lt; 0.05 or ## <span class="html-italic">p</span> &lt; 0.01 vs. GFAP-IL6 mice. n.s. = not significant. Tukey post-hoc test after analysis of variance.</p>
Full article ">Figure 2
<p>Comparative analyses of STAT3, NFκB and ERK1/2<sup>MAPK</sup> phosphorylation in the cerebrum of GFAP-IL6 and GFAP-IL6/sgp130Fc mice. Co-expression of sgp130Fc reduces steady-state p-STAT3<sup>(Y705)</sup> and p-NFκB<sup>(S536)</sup> but not p-ERK1/2<sup>MAPK</sup> in the cerebrum of GFAP-IL6 mice. (<b>A</b>,<b>C</b>) Tissue lysates (15 μg protein per lane) from cerebra of 6-month-old mice were subjected to SDS-PAGE followed by immunoblotting. (<b>B</b>,<b>D</b>), X-ray films were quantified by densitometry (OD) using NIH ImageJ software (version 1.52) in (<b>B</b>) for p-STAT3, STAT3, p-NFκB, NFκB or reported as a ratio between phospho-specific and pan proteins and in (<b>D</b>) for p-ERK1/2, ERK1/2 or as a ratio. GAPDH was used as loading control. Values represent the mean ± SEM with n = 3 brains per genotype. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 or *** <span class="html-italic">p</span> &lt; 0.001 vs. WT. # <span class="html-italic">p</span> &lt; 0.05 or ### <span class="html-italic">p</span> &lt; 0.001 vs. GFAP-IL6 mice. One-Way ANOVA followed Tukey post-hoc test.</p>
Full article ">Figure 3
<p>Comparative analyses of STAT3, NFκB and ERK1/2<sup>MAPK</sup> phosphorylation in the cerebellum of GFAP-IL6 and GFAP-IL6/sgp130Fc mice. Co-expression of sgp130Fc reduces steady-state p-STAT3<sup>(Y705)</sup> and p-NFκB<sup>(S536)</sup> but not p-ERK1/2<sup>MAPK</sup> in the cerebellum of GFAP-IL6 mice. (<b>A</b>,<b>C</b>) Tissue lysates (15 μg protein per lane) from cerebra of 6-month-old mice were subjected to SDS-PAGE followed by immunoblotting (<b>B</b>,<b>D</b>). X-ray films were quantified by densitometry (OD) using NIH ImageJ software in (<b>B</b>) for p-STAT3, STAT3, p-NFκB and NFκB or reported as a ratio between phospho-specific and pan proteins and in (<b>D</b>) for p-ERK1/2 and ERK1/2 or as a ratio. GAPDH was used as a loading control. Values represent the mean ± SEM, with n = 3 brains per genotype. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 or *** <span class="html-italic">p</span> &lt; 0.001 vs. WT. # <span class="html-italic">p</span> &lt; 0.05 or ## <span class="html-italic">p</span> &lt; 0.01 vs. GFAP-IL6 mice. One-Way ANOVA followed Tukey post-hoc test.</p>
Full article ">Figure 4
<p>Expression of PAC1, VPAC1 and VPAC2 receptors in the cerebrum of GFAP-IL6 and GFAP-IL6/sgp130Fc mice. The presence of sgp130Fc does not prevent the increase in PACAP/VIP receptors in the cerebrum of GFAP-IL6 mice. (<b>A</b>,<b>C</b>,<b>E</b>) Western blots and (<b>B</b>,<b>D</b>,<b>F</b>) densitometry of bands obtained from lysates of cerebra from 6-month-old mice (n = 3 × genotype) that were separated by SDS-PAGE and quantified by NIH ImageJ software (version 1.52). GAPDH was used as the loading control. (<b>G</b>,<b>I</b>,<b>K</b>) PAC1, VPAC1 and VPAC2 immunohistochemistry was performed on paraformaldehyde fixed, paraffin-embedded sections (5 µm) of brains prepared from 6-month-old mice. Scale bar, 50 µm. (<b>H</b>,<b>J</b>,<b>L</b>) Semi-quantitative analyses of immunoreactivities were performed on at least three blinded sections per brain and on a minimum of four brains × genotype. Values represent the mean ± SEM with n = 4 brains per genotype. ** <span class="html-italic">p</span> &lt; 0.01 or *** <span class="html-italic">p</span> &lt; 0.001 vs. WT mice. One-Way ANOVA followed Tukey post-hoc test.</p>
Full article ">Figure 5
<p>Expression of PAC1, VPAC1 and VPAC2 receptors in the cerebellum of GFAP-IL6 and GFAP-IL6/sgp130Fc mice. Inhibition of IL6 trans-signalling failed to reduce GFAP-IL6-driven induction of PACAP/VIP receptors in the mouse cerebellum. (<b>A</b>,<b>C</b>,<b>E</b>) Tissue lysates (15 µg protein per lane) from cerebellum of 6-month-old mice were subjected to SDS-PAGE followed by immunoblotting. (<b>B</b>,<b>D</b>,<b>F</b>) Quantification of band densities (n = 3 × genotype) by NIH ImageJ software. GAPDH was used as loading control. (<b>G</b>,<b>I</b>,<b>K</b>) PAC1, VPAC1 and VPAC2 immunohistochemistry was performed on paraformaldehyde fixed, paraffin-embedded sections (5 µm) of brains prepared from 6-month-old mice. Scale bar, 50 µm. (<b>H</b>,<b>J</b>,<b>L</b>) Stereological assessments of (<b>H</b>) PAC1<sup>+</sup>, (<b>J</b>) VPAC1<sup>+</sup> and (<b>L</b>) VPAC2<sup>+</sup> cells in each of the three cerebellar cortical layers was performed on at least three blinded sections per brain and using four brains × genotype. Values represent the mean ± SEM with n = 4 brains per genotype. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 or *** <span class="html-italic">p</span> &lt; 0.001 vs. WT mice. Western blots (<b>A</b>–<b>F</b>): One-Way ANOVA followed by Tukey post-hoc tests. Stereology (<b>G</b>–<b>L</b>): 2-Way ANOVA (factoring in both cell layers and genotypes) followed Tukey post-hoc tests. ML = Molecular layer; PC = Purkinje cells layer; GCL = Granular cell layer.</p>
Full article ">
14 pages, 4045 KiB  
Article
Dipeptidyl Peptidase (DPP)-4 Inhibitors and Pituitary Adenylate Cyclase-Activating Polypeptide, a DPP-4 Substrate, Extend Neurite Outgrowth of Mouse Dorsal Root Ganglia Neurons: A Promising Approach in Diabetic Polyneuropathy Treatment
by Masahiro Yamaguchi, Saeko Noda-Asano, Rieko Inoue, Tatsuhito Himeno, Mikio Motegi, Tomohide Hayami, Hiromi Nakai-Shimoda, Ayumi Kono, Sachiko Sasajima, Emiri Miura-Yura, Yoshiaki Morishita, Masaki Kondo, Shin Tsunekawa, Yoshiro Kato, Koichi Kato, Keiko Naruse, Jiro Nakamura and Hideki Kamiya
Int. J. Mol. Sci. 2024, 25(16), 8881; https://doi.org/10.3390/ijms25168881 - 15 Aug 2024
Cited by 1 | Viewed by 1255
Abstract
Individuals suffering from diabetic polyneuropathy (DPN) experience debilitating symptoms such as pain, paranesthesia, and sensory disturbances, prompting a quest for effective treatments. Dipeptidyl-peptidase (DPP)-4 inhibitors, recognized for their potential in ameliorating DPN, have sparked interest, yet the precise mechanism underlying their neurotrophic impact [...] Read more.
Individuals suffering from diabetic polyneuropathy (DPN) experience debilitating symptoms such as pain, paranesthesia, and sensory disturbances, prompting a quest for effective treatments. Dipeptidyl-peptidase (DPP)-4 inhibitors, recognized for their potential in ameliorating DPN, have sparked interest, yet the precise mechanism underlying their neurotrophic impact on the peripheral nerve system (PNS) remains elusive. Our study delves into the neurotrophic effects of DPP-4 inhibitors, including Diprotin A, linagliptin, and sitagliptin, alongside pituitary adenylate cyclase-activating polypeptide (PACAP), Neuropeptide Y (NPY), and Stromal cell-derived factor (SDF)-1a—known DPP-4 substrates with neurotrophic properties. Utilizing primary culture dorsal root ganglia (DRG) neurons, we meticulously evaluated neurite outgrowth in response to these agents. Remarkably, all DPP-4 inhibitors and PACAP demonstrated a significant elongation of neurite length in DRG neurons (PACAP 0.1 μM: 2221 ± 466 μm, control: 1379 ± 420, p < 0.0001), underscoring their potential in nerve regeneration. Conversely, NPY and SDF-1a failed to induce neurite elongation, accentuating the unique neurotrophic properties of DPP-4 inhibition and PACAP. Our findings suggest that the upregulation of PACAP, facilitated by DPP-4 inhibition, plays a pivotal role in promoting neurite elongation within the PNS, presenting a promising avenue for the development of novel DPN therapies with enhanced neurodegenerative capabilities. Full article
(This article belongs to the Special Issue Peripheral Neuropathies: Molecular Research and Novel Therapy)
Show Figures

Figure 1

Figure 1
<p>Expression and activity of DPP-4 in the peripheral nervous system. (<b>A</b>) RT-PCR of DPP-4 mRNA on several tissues: liver, dorsal root ganglia (DRG), sciatic nerve (SCN), heart, and skeletal muscle (SM). (<b>B</b>–<b>D</b>) Immunostaining of DPP-4 protein on DRG. (<b>B</b>): DPP-4. (<b>C</b>): DAPI. (<b>D</b>): Merge. Scale bar: 50 μm. (<b>E</b>) Quantified DPP-4 activity of DRG and liver, μU: microunits; mgPro: mg Protein; ns: not significant between DRG and liver.</p>
Full article ">Figure 2
<p>Neurite outgrowth of DRG neurons by Diprotin A. (<b>A</b>–<b>D</b>) Representative fluorescence figures of DRG neurons cultured in the absence (<b>A</b>) or presence (<b>B</b>–<b>D</b>) of Diprotin A ((<b>B</b>): 1 μM, (<b>C</b>): 10 μM, (<b>D</b>): 50 μM). Scale bar: 50 μm. (<b>E</b>) Quantified neurite length in each DRG neuron. *: <span class="html-italic">p</span> &lt; 0.05 versus control; ns: not significant versus control.</p>
Full article ">Figure 3
<p>Quantified neurite length in each DRG neuron with or without sitagliptin. ***: <span class="html-italic">p</span> &lt; 0.001, ****: <span class="html-italic">p</span> &lt; 0.0001 versus control.</p>
Full article ">Figure 4
<p>Neurite outgrowth of DRG neurons by linagliptin. (<b>A</b>–<b>C</b>) Representative fluorescence figures of DRG neurons cultured in the absence (<b>A</b>) or presence (<b>B</b>,<b>C</b>) of linagliptin ((<b>B</b>): 10 nM, (<b>C</b>): 100 nM). Scale bar: 50 μm. (<b>D</b>) Quantified neurite length in each DRG neuron. *: <span class="html-italic">p</span> &lt; 0.05; ns: not significant versus control.</p>
Full article ">Figure 5
<p>Neurite outgrowth of DRG neurons by PACAP. (<b>A</b>–<b>C</b>) Representative fluorescence figures of DRG neurons cultured in the absence (<b>A</b>) or presence (<b>B</b>,<b>C</b>) of PACAP ((<b>B</b>): 0.1 μM, (<b>C</b>): 1 μM). Scale bar: 50 μm. (<b>D</b>) Quantified neurite length in each DRG neuron. ***: <span class="html-italic">p</span> &lt; 0.001, ****: <span class="html-italic">p</span> &lt; 0.0001 versus control.</p>
Full article ">Figure 6
<p>Neurite outgrowth of DRG neurons by NPY and SDF-1α. (<b>A</b>–<b>C</b>) Representative fluorescence figures of DRG neurons cultured in the absence (<b>A</b>) or presence of (<b>B</b>) NPY 0.1 μM and (<b>C</b>) SDF-1α 0.1 μM. Scale bar: 50 μm. (<b>D</b>) Quantified neurite length in each DRG neuron. ns: not significant versus control.</p>
Full article ">Figure 7
<p>Cell viability in 50B11 cells treated with DPP-4 inhibitors and PACAP. H<sub>2</sub>O<sub>2</sub> (−): no supplementation of H<sub>2</sub>O<sub>2</sub>. 0.1 mM H<sub>2</sub>O<sub>2</sub> (+): supplementation of 0.1 mM H<sub>2</sub>O<sub>2</sub>. 100% cell viability: control without H<sub>2</sub>O<sub>2</sub>. H<sub>2</sub>O<sub>2</sub>: hydrogen peroxide. ***: <span class="html-italic">p</span> &lt; 0.001, ****: <span class="html-italic">p</span> &lt; 0.0001 versus control with 0.1 mM H<sub>2</sub>O<sub>2</sub>.</p>
Full article ">
24 pages, 16101 KiB  
Article
Differential Expression of PACAP/VIP Receptors in the Post-Mortem CNS White Matter of Multiple Sclerosis Donors
by Margo Iris Jansen, Giuseppe Musumeci and Alessandro Castorina
Int. J. Mol. Sci. 2024, 25(16), 8850; https://doi.org/10.3390/ijms25168850 - 14 Aug 2024
Viewed by 1003
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal peptide (VIP) are two neuroprotective and anti-inflammatory molecules of the central nervous system (CNS). Both bind to three G protein-coupled receptors, namely PAC1, VPAC1 and VPAC2, to elicit their beneficial effects in various CNS diseases, [...] Read more.
Pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal peptide (VIP) are two neuroprotective and anti-inflammatory molecules of the central nervous system (CNS). Both bind to three G protein-coupled receptors, namely PAC1, VPAC1 and VPAC2, to elicit their beneficial effects in various CNS diseases, including multiple sclerosis (MS). In this study, we assessed the expression and distribution of PACAP/VIP receptors in the normal-appearing white matter (NAWM) of MS donors with a clinical history of either relapsing–remitting MS (RRMS), primary MS (PPMS), secondary progressive MS (SPMS) or in aged-matched non-MS controls. Gene expression studies revealed MS-subtype specific changes in PACAP and VIP and in the receptors’ levels in the NAWM, which were partly corroborated by immunohistochemical analyses. Most PAC1 immunoreactivity was restricted to myelin-producing cells, whereas VPAC1 reactivity was diffused within the neuropil and in axonal bundles, and VPAC2 in small vessel walls. Within and around lesioned areas, glial cells were the predominant populations showing reactivity for the different PACAP/VIP receptors, with distinctive patterns across MS subtypes. Together, these data identify the differential expression patterns of PACAP/VIP receptors among the different MS clinical entities. These results may offer opportunities for the development of personalized therapeutic approaches to treating MS and/or other demyelinating disorders. Full article
Show Figures

Figure 1

Figure 1
<p>Representative lesions and normal-appearing white matter in human brain sections from donors with different MS subtypes. Luxol Fast Blue (LFB) staining shows the intense blue staining of myelinated fibers in the white matter (WM) of (<b>A</b>) non-MS donors, differentiating it from the less myelinated grey matter (GM). Evident discoloring of lesioned areas (indicated by black arrowheads) can be appreciated in sections from (<b>B</b>) RRMS, (<b>C</b>) PPMS and (<b>D</b>) SPMS cases. Myelin is stained blue, resulting in a clear distinction between GM and WM. Scale bar in (<b>A</b>) 200 µm, (<b>B</b>,<b>C</b>) 500 µm, (<b>D</b>) 2000 µm and NAWM (panels on the right) 25 µm. MS = multiple sclerosis, RRMS = relapsing–remitting MS, PPMS = primary-progressive MS, SPMS = secondary-progressive MS, GM = grey matter, WM = white matter.</p>
Full article ">Figure 2
<p>Differential expression of PACAP and VIP neuropeptide genes in the normal-appearing white matter of MS donors. (<b>A</b>) PACAP (gene name = ADCYAP1) expression was measured using RT-qPCR, comparing non-MS and MS cases. Further stratification of cases by clinical course, showing the expression levels of ADCYAP1 in (<b>A′</b>) non-MS vs. RRMS, (<b>A″</b>) non-MS vs. PPMS and (<b>A‴</b>) non-MS vs. SPMS. (<b>B</b>) VIP gene expression in non-MS vs. MS cases. A stratification similar to that in A demonstrates relative changes in the transcript levels between non-MS and (<b>B′</b>) RRMS, (<b>B″</b>) PPMS and (<b>B‴</b>) SPMS cases. The data shown are the mean fold change ± SEM, obtained from <span class="html-italic">n</span> = 6 (non-MS), <span class="html-italic">n</span> = 5 (RRMS), <span class="html-italic">n</span> = 6 (SPMS) and <span class="html-italic">n</span> = 4 (PPMS) cases. <span class="html-italic">p</span>-values &gt; 0.05 are also shown. * <span class="html-italic">p</span> &lt; 0.05 or *** <span class="html-italic">p</span> &lt; 0.001 vs. non-MS, as determined by unpaired <span class="html-italic">t</span>-test. VIP = vasoactive intestinal peptide, PACAP = pituitary adenylate cyclase activating polypeptide, MS = multiple sclerosis, NAWM = normal-appearing white matter, RRMS = relapsing–remitting MS, PPMS = primary progressive MS, SPMS = secondary progressive MS.</p>
Full article ">Figure 3
<p>Differential expression of PAC1, VPAC1 and VPAC2 genes in the normal-appearing white matter of MS donors. Gene expression of (<b>A</b>) ADCYAP1R1 (aka PAC1), (<b>B</b>) VIPR1 (VPAC1) and (<b>C</b>) VIPR2 (VPAC2) in the NAWM of non-MS vs. MS donors. Upon stratification based on the clinical MS course, the gene expression levels of ADCYAP1R1, VIPR1 and VIPR2 were determined for (<b>A′</b>–<b>C′</b>) RRMS, (<b>A″</b>–<b>C″</b>) PPMS and (<b>A‴</b>–<b>C‴</b>) SPMS cases. The data shown are the mean fold change ± SEM, obtained from <span class="html-italic">n</span> = 6 (non-MS), <span class="html-italic">n</span> = 5 (RRMS), <span class="html-italic">n</span> = 6 (SPMS) and <span class="html-italic">n</span> = 4 (PPMS) cases. <span class="html-italic">p</span>-values &gt; 0.05 are also shown. * <span class="html-italic">p</span> &lt; 0.05 or *** <span class="html-italic">p</span> &lt; 0.001 vs. non-MS, as determined by unpaired <span class="html-italic">t</span>-test. ADCYAP1R1 = Pituitary adenylate cyclase-activating polypeptide type I receptor, VIPR1 = Vasoactive intestinal polypeptide receptor 1, VIPR2 = Vasoactive intestinal polypeptide receptor 2, MS = multiple sclerosis, NAWM = normal-appearing white matter, RRMS = relapsing–remitting MS, PPMS = primary progressive MS, SPMS = secondary progressive MS.</p>
Full article ">Figure 4
<p>PAC1 immunoreactivity in the normal-appearing white matter of RRMS, PPMS and SPMS cases. (<b>A</b>) Representative images showing PAC1 immunoreactive cells in the NAWM of MS donors with a clinical history of RRMS, PPMS or SPMS and non-MS control cases. White arrows in each panel point to PAC1<sup>+</sup> cells, which exhibit chromatin-dense and rounded/oval shaped nuclei, consistent with the oligodendrocyte/OPC morphology. (<b>B</b>) The average cell density (total # of cells per region of interest (ROI); ROI area = 1.23 mm<sup>2</sup>) was calculated using 2–4 ROIs from <span class="html-italic">n</span> = 5 (non-MS), <span class="html-italic">n</span> = 4 (PPMS), <span class="html-italic">n</span> = 6 (RRMS) and <span class="html-italic">n = 6</span> (SPMS) cases. (<b>C</b>) The PAC1 immunoreactivity in cells was determined by normalizing the mean PAC1 staining intensity/average # of cells counted within the same ROIs/cases as in (<b>B</b>). *** <span class="html-italic">p</span> &lt; 0.001 or **** <span class="html-italic">p</span> &lt; 0.0001 vs. non-MS cases, as determined by one-way ANOVA followed by Sidak’s post hoc test. Scale bar = 30 µm. OPC = Oligodendrocyte progenitor cell, PAC1 = Pituitary adenylate cyclase-activating polypeptide type I receptor, MS = multiple sclerosis, NAWM = normal-appearing white matter, RRMS = relapsing–remitting MS, PPMS = primary progressive MS, SPMS = secondary progressive MS.</p>
Full article ">Figure 5
<p>PAC1 co-localizes to OLIG2<sup>+</sup> cells in the normal-appearing white matter of RRMS, PPMS and SPMS cases. Representative images showing PAC1 (green)/OLIG2 (red) colocalization in the NAWM of (<b>A</b>) non-MS, (<b>B</b>) RRMS, (<b>C</b>) PPMS or (<b>D</b>) SPMS donors. Nuclei were counterstained with DAPI (blue). Scale bar = 50 µm.</p>
Full article ">Figure 6
<p>PAC1 immunoreactivity in representative white matter lesions from selected MS clinical cases. (<b>A</b>–<b>C</b>, left panels) Low-magnification images showing PAC1 immunoreactivity in a lesion taken from one RRMS, PPMS or SPMS-exemplary case. Lesion borders are demarcated by the black dashed lines. Scale bar = 1000 µm. (Insets in <b>A</b>–<b>C</b>) High-power images of ROIs in the left panels (orange and red squares) demonstrating PAC1<sup>+</sup> staining around the lesion edge (top inset) and within the lesion (bottom inset) of the selected RRMS, PPMS and SPMS cases. Scale bar = 30 µm. WM = white matter.</p>
Full article ">Figure 7
<p>VPAC1 immunoreactivity in the normal-appearing white matter of RRMS, PPMS and SPMS cases. (<b>A</b>–<b>D</b>) Representative images depicting VPAC1 immunoreactive sites in the NAWM of MS donors with a clinical history of RRMS, PPMS or SPMS and non-MS control cases. Scale bar = 1000 µm. (<b>A′</b>–<b>D′</b>) Insets of the NAWM taken at a higher magnification. Black arrowheads point to VPAC1<sup>+</sup> axonal fibers. Scale bar (NAWM) = 30 µm (<b>B″</b>–<b>D″</b>). Insets showing VPAC1<sup>+</sup> in the grey matter of the selected cases. Black arrowheads indicate VPAC1<sup>+</sup> neurons. Scale bar (GM) = 50 µm. (<b>E</b>) Bar graph showing the average VPAC1 immunoreactivity (IR) in the NAWM. The data shown are the mean grey intensity ± SEM and were calculated by averaging the grey intensity of 2–4 ROIs from <span class="html-italic">n</span> = 5 (non-MS), <span class="html-italic">n</span> = 4 (PPMS), <span class="html-italic">n</span> = 6 (RRMS) and <span class="html-italic">n</span> = 6 (SPMS) cases. Each ROI area = 1.23 mm<sup>2</sup>. No statistical significance was found using one-way ANOVA. Ns = Not significant. VPAC1 = Vasoactive Intestinal Peptide/Pituitary Adenylate Cyclase Activating Polypeptide Receptor 1, MS = multiple sclerosis, NAWM = normal-appearing white matter, WM = white matter, GM = grey matter, RRMS = relapsing–remitting MS, PPMS = primary progressive MS, SPMS = secondary progressive MS.</p>
Full article ">Figure 8
<p>VPAC1 immunoreactivity in white matter lesions from selected MS clinical cases. (<b>A</b>–<b>C</b>, left panels) Low-magnification images showing VPAC1 immunoreactivity in a lesion taken from one RRMS, PPMS or SPMS-exemplary case. Lesion borders are demarcated by the black dashed lines. Scale bar = 1000 µm. (Insets in <b>A</b>–<b>C</b>) High-power images of ROIs in left panels (orange and red squares) demonstrating VPAC1<sup>+</sup> staining around the lesion edge (top inset) and within the lesion (bottom inset) of the selected RRMS, PPMS and SPMS cases. Scale bar = 30 µm. WM = white matter.</p>
Full article ">Figure 9
<p>VPAC2 immunoreactivity in the normal-appearing white matter of RRMS, PPMS and SPMS cases. (<b>A</b>–<b>D</b>) Representative images depicting VPAC2 immunoreactive (IR) cells in the NAWM of MS donors with a clinical history of RRMS, PPMS or SPMS and non-MS controls. Scale bar = 1000 µm. (<b>A′</b>–<b>D′</b>) Insets of the NAWM taken at a higher magnification. White arrowheads in C′ show VPAC2<sup>+</sup> vessel walls. Scale bar (NAWM) = 30 µm. (<b>B″</b>–<b>D″</b>) Insets showing VPAC2<sup>+</sup> in the grey matter of the selected cases. Black arrowheads indicate VPAC2<sup>+</sup> neurons, whereas white arrowheads show VPAC2-IR in axons. Scale bar (GM) = 50 µm. (<b>E</b>) Bar graph showing the average VPAC1 immunoreactivity (IR) in the NAWM. The data shown are the mean grey intensity ± SEM and were calculated by averaging the grey intensity of 2–4 ROIs from <span class="html-italic">n = 5</span> (non-MS), <span class="html-italic">n</span> = 4 (PPMS), <span class="html-italic">n</span> = 6 (RRMS) and <span class="html-italic">n</span> = 6 (SPMS) cases. Each ROI area = 1.23 mm<sup>2</sup>. **** <span class="html-italic">p</span> &lt; 0.0001 vs. non-MS (control) cases, as determined by one-way ANOVA and Sidak’s post hoc test. VPAC2 = Vasoactive Intestinal Peptide/Pituitary Adenylate Cyclase Activating Polypeptide Receptor 1, MS = multiple sclerosis, NAWM = normal-appearing white matter, GM = grey matter, RRMS = relapsing–remitting MS, PPMS = primary progressive MS, SPMS = secondary progressive MS.</p>
Full article ">Figure 10
<p>VPAC2 immunoreactivity in white matter lesions from selected MS clinical cases. (<b>A</b>–<b>C</b>, left panels) Low-magnification images showing VPAC2 immunoreactivity in a lesion taken from one RRMS, PPMS or SPMS-exemplary case. Lesion borders are demarcated by the black dashed lines. Scale bar = 1000 µm. (Insets in <b>A</b>–<b>C</b>) High-power images of ROIs in left panels (orange and red squared) demonstrating VPAC2 staining around the lesion edge (top inset) and within the lesion (bottom inset) of the selected RRMS, PPMS and SPMS cases. Scale bar = 30 µm. GM = grey matter; WM = white matter.</p>
Full article ">
17 pages, 3127 KiB  
Article
Exploring the Role of Neuropeptide PACAP in Cytoskeletal Function Using Spectroscopic Methods
by Roland Gábor Vékony, Andrea Tamás, András Lukács, Zoltán Ujfalusi, Dénes Lőrinczy, Veronika Takács-Kollár and Péter Bukovics
Int. J. Mol. Sci. 2024, 25(15), 8063; https://doi.org/10.3390/ijms25158063 - 24 Jul 2024
Viewed by 1101
Abstract
The behavior and presence of actin-regulating proteins are characteristic of various clinical diseases. Changes in these proteins significantly impact the cytoskeletal and regenerative processes underlying pathological changes. Pituitary adenylate cyclase-activating polypeptide (PACAP), a cytoprotective neuropeptide abundant in the nervous system and endocrine organs, [...] Read more.
The behavior and presence of actin-regulating proteins are characteristic of various clinical diseases. Changes in these proteins significantly impact the cytoskeletal and regenerative processes underlying pathological changes. Pituitary adenylate cyclase-activating polypeptide (PACAP), a cytoprotective neuropeptide abundant in the nervous system and endocrine organs, plays a key role in neuron differentiation and migration by influencing actin. This study aims to elucidate the role of PACAP as an actin-regulating polypeptide, its effect on actin filament formation, and the underlying regulatory mechanisms. We examined PACAP27, PACAP38, and PACAP6-38, measuring their binding to actin monomers via fluorescence spectroscopy and steady-state anisotropy. Functional polymerization tests were used to track changes in fluorescent intensity over time. Unlike PACAP27, PACAP38 and PACAP6-38 significantly reduced the fluorescence emission of Alexa488-labeled actin monomers and increased their anisotropy, showing nearly identical dissociation equilibrium constants. PACAP27 showed weak binding to globular actin (G-actin), while PACAP38 and PACAP6-38 exhibited robust interactions. PACAP27 did not affect actin polymerization, but PACAP38 and PACAP6-38 accelerated actin incorporation kinetics. Fluorescence quenching experiments confirmed structural changes upon PACAP binding; however, all studied PACAP fragments exhibited the same effect. Our findings indicate that PACAP38 and PACAP6-38 strongly bind to G-actin and significantly influence actin polymerization. Further studies are needed to fully understand the biological significance of these interactions. Full article
(This article belongs to the Special Issue New Drugs Regulating Cytoskeletons in Human Health and Diseases)
Show Figures

Figure 1

Figure 1
<p>Sequences of PACAP forms (PACAP38, PACAP27, and PACAP6-38). This figure illustrates the amino acid sequences of four different forms of PACAP, highlighting the variations in their N-terminal and C-terminal regions. The numbers in the PACAP names indicate the specific amino acid orders included in each form, with the left number representing the starting amino acid from the N-terminal side and the right number indicating the ending amino acid on the C-terminal side.</p>
Full article ">Figure 2
<p>The 3D structure of PACAP27, which is identical to the first 27 residues of PACAP38. This figure is based on the PDB file 6M1I (from Cryo-EM structures of PACAP38-PAC1R-G<sub>s</sub> [<a href="#B83-ijms-25-08063" class="html-bibr">83</a>]). The yellow coloring represents the N-terminal of the peptide, while the aromatic residues crucial for fluorescence are highlighted (green: tyrosine; red: phenylalanine). This figure was created using Discovery Studio Visualizer (v16.1.0.15350, Biovia).</p>
Full article ">Figure 3
<p>Anisotropy of 0.2 µM A488NHS-labeled G-actin as a function of increasing concentrations of PACAP isoforms (PACAP38, PACAP27, and PACAP6-38). Anisotropy measurements were conducted with PACAP concentrations ranging from 0 µM (control) to 5 µM. Data points represent the mean ± standard deviation of at least three independent experiments.</p>
Full article ">Figure 4
<p>Representative emission spectra of Alexa488-labeled actin (0.2 µM) with increasing concentrations of PACAP isoforms ((<b>A</b>): PACAP38, (<b>B</b>): PACAP27, and (<b>C</b>): PACAP6-38). The spectra were recorded ranging from 0 µM (control) to 5 µM. Each spectrum illustrates changes in fluorescence intensity, with potential shifts in emission peaks, allowing for a comparative analysis of the interaction between PACAP38 and actin monomers.</p>
Full article ">Figure 5
<p>Normalized fluorescence intensity (F/F<sub>0</sub>) of 0.2 µM A488NHS-labeled actin as a function of increasing PACAP concentrations. Fluorescence measurements were conducted with PACAP concentrations ranging from 0 µM (control) to 5 µM. The data points represent the mean ± standard deviation from three independent experiments.</p>
Full article ">Figure 6
<p>Polymerization kinetics of 2.5 µM actin (5% pyrene-labeled) as a function of increasing PACAP concentrations. The polymerization process was monitored by the increase in pyrene fluorescence over time. Polimerization assays were conducted with PACAP concentrations ranging from 0 µM (control) to 12.5 µM. Data points represent the mean ± standard deviation from three independent experiments. Two-way ANOVA did not reject the null hypothesis, indicating no significant difference between the PACAP forms.</p>
Full article ">Figure 7
<p>Tryptophan fluorescence quenching by acrylamide in 1 mg/mL actin as a function of various PACAP forms/fragments. The graph depicts the quenching efficiency (F<sub>0</sub>/F) of tryptophan fluorescence in the presence of PACAP38, PACAP27, and PACAP6-38. The quenching measurements were performed with increasing concentrations of acrylamide, and the data points represent the mean ± standard deviation from at least three independent experiments. Two-way ANOVA also did not reject the null hypothesis, indicating no significant difference between the control and PACAP isoforms.</p>
Full article ">Figure 8
<p>This figure outlines the sequential steps involved in the preparation, analysis, and characterization of PACAP interactions with actin. The key steps include PACAP and actin preparation, steady-state anisotropy and fluorescence emission measurements, polymerization assays, and fluorescence quenching studies.</p>
Full article ">
16 pages, 7400 KiB  
Article
Optimization of an Ischemic Retinopathy Mouse Model and the Consequences of Hypoxia in a Time-Dependent Manner
by Inez Bosnyak, Nelli Farkas, Dorottya Molitor, Balazs Meresz, Evelin Patko, Tamas Atlasz, Alexandra Vaczy and Dora Reglodi
Int. J. Mol. Sci. 2024, 25(15), 8008; https://doi.org/10.3390/ijms25158008 - 23 Jul 2024
Viewed by 1123
Abstract
The retina is one of the highest metabolically active tissues with a high oxygen consumption, so insufficient blood supply leads to visual impairment. The incidence of related conditions is increasing; however, no effective treatment without side effects is available. Furthermore, the pathomechanism of [...] Read more.
The retina is one of the highest metabolically active tissues with a high oxygen consumption, so insufficient blood supply leads to visual impairment. The incidence of related conditions is increasing; however, no effective treatment without side effects is available. Furthermore, the pathomechanism of these diseases is not fully understood. Our aim was to develop an optimal ischemic retinopathy mouse model to investigate the retinal damage in a time-dependent manner. Retinal ischemia was induced by bilateral common carotid artery occlusion (BCCAO) for 10, 13, 15 or 20 min, or by right permanent unilateral common carotid artery occlusion (UCCAO). Optical coherence tomography was used to follow the changes in retinal thickness 3, 7, 14, 21 and 28 days after surgery. The number of ganglion cells was evaluated in the central and peripheral regions on whole-mount retina preparations. Expression of glial fibrillary acidic protein (GFAP) was analyzed with immunohistochemistry and Western blot. Retinal degeneration and ganglion cell loss was observed in multiple groups. Our results suggest that the 20 min BCCAO is a good model to investigate the consequences of ischemia and reperfusion in the retina in a time-dependent manner, while the UCCAO causes more severe damage in a short time, so it can be used for testing new drugs. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

Figure 1
<p>Optical coherence tomography (OCT) results and layers of the retina. The dots show the individual values, while the lines show the averages. Statistical analysis was performed with the linear random effect mixed model, and multiple comparisons were performed with Satterthwaite’s method. ˙ <span class="html-italic">p</span> &lt; 0.1, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. change in the control group. UCCAO: unilateral common carotid artery occlusion. Control: <span class="html-italic">n</span> = 10; 10-min BCCAO: <span class="html-italic">n</span> = 12; 13-min BCCAO: <span class="html-italic">n</span> = 14; 15-min BCCAO: <span class="html-italic">n</span> = 14; 20-min BCCAO: <span class="html-italic">n</span> = 17; UCCAO: <span class="html-italic">n</span> = 25. (<b>A</b>) OCT results of inner retinal layers. The upper bracket shows the inner layers of the retina. (<b>B</b>) OCT results of middle retinal layers. The middle bracket indicates the middle layers of the retina. (<b>C</b>) OCT results of the outer part of the retina. The bottom bracket shows the outer retinal layers. (<b>D</b>) OCT results of total retinal thickness. (<b>E</b>) A representative OCT image. Scale bar: 202 µm. RNFL: retinal nerve fiber layer. IPL: inner plexiform layer. INL: inner nuclear layer. OPL: outer plexiform layer. ONL: outer nuclear layer. IS/OS: inner and outer segments of photoreceptor cells. RPE: retinal pigment epithelium.</p>
Full article ">Figure 2
<p>Results of optical coherence tomography (OCT) measurements of the different retinal layers. The dots show the individual values, while the lines show the averages. Statistical analysis was performed with the linear random effect mixed model, and multiple comparisons were performed with Satterthwaite’s method. ˙ <span class="html-italic">p</span> &lt; 0.1, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. change in control group. Control: <span class="html-italic">n</span> = 10; 10 min BCCAO: <span class="html-italic">n</span> = 12; 13 min BCCAO: <span class="html-italic">n</span> = 14; 15 min BCCAO: <span class="html-italic">n</span> = 14; 20 min BCCAO: <span class="html-italic">n</span> = 17; UCCAO: <span class="html-italic">n</span> = 25. The thickness of (<b>A</b>) nerve fiber layer; (<b>B</b>) retinal pigment epithelium; (<b>C</b>) whole photoreceptor cells; (<b>D</b>) outer segment of photoreceptor cells; (<b>E</b>) inner segment of photoreceptor cells.</p>
Full article ">Figure 3
<p>Brn3a labeling of retinal ganglion cells. Statistical analysis was performed with Kruskal–Wallis ANOVA and uncorrected Dunn’s post hoc test. <span class="html-italic">n</span> = 3/group. (<b>A</b>) Representative images of ganglion cells from the different groups in the central region of the retina under 20× magnification. Scale bar: 50 µm. (<b>B</b>) Representative images of ganglion cells from the different groups in the peripheral region of the retina under 20× magnification. Scale bar: 50 µm. (<b>C</b>) Representative image of a whole mount retina preparation. Rectangle I. indicates the location of the images of the central region, while the rectangle II. shows the location of the images of the peripheral region. Scale bar: 100 µm. (<b>D</b>) Ganglion cell count in the peripheral regions of the retina. * <span class="html-italic">p</span> &lt; 0.05 vs. control. Area = 0.344 mm<sup>2</sup>. (<b>E</b>) Number of ganglion cells in the central regions of the retina. * <span class="html-italic">p</span> &lt; 0.05 vs. control. Area = 0.344 mm<sup>2</sup>.</p>
Full article ">Figure 4
<p>(<b>A</b>) Representative images of GFAP staining on retinal whole mounts from the peripheral regions of the retina. UCCAO: unilateral common carotid artery occlusion. Scale bar: 10 µm (<b>B</b>) Representative images of GFAP labeling with DAPI counterstaining on cross sections from the same groups as above. INL: inner nuclear layer. ONL: outer nuclear layer. Scale bar: 10 µm (<b>C</b>) Results of Western blot measurements. Statistical analysis: one-way ANOVA, Dunnet’s post hoc test. * <span class="html-italic">p</span> &lt; 0.05 vs. control. <span class="html-italic">n</span> = 4/group × 3 runs (<b>D</b>) A representative blot of GFAP expression with the same groups as above. GFAP: glial fibrillary acidic protein. GAPDH: internal control.</p>
Full article ">Figure 5
<p>Illustration of the surgical procedure. Schematic drawing of the isolated right common carotid artery, with the vagus nerve next to it. (<b>A</b>) Representative picture of the bilateral common carotid artery occlusion (BCCAO). Both common carotid arteries are isolated and occluded with surgical microclips. (<b>B</b>) The right common carotid artery is occluded with two sutures and cut between the two sutures to avoid reperfusion.</p>
Full article ">
12 pages, 2699 KiB  
Article
Sex Differences in the Brain Transcriptomes of Adult Blue Gourami Fish (Trichogaster trichopterus)
by Gad Degani and Ari Meerson
Fishes 2024, 9(7), 287; https://doi.org/10.3390/fishes9070287 - 18 Jul 2024
Cited by 1 | Viewed by 1008
Abstract
Blue gourami (gourami, Trichogaster trichopterus) is a model for labyrinth fishes (Anabantoidei) adapted to partial air breathing. Its reproductive endocrinology has been extensively studied, and transcriptomic sex differences in the gonads were described. Nevertheless, sex differences in gene expression in non-gonadal tissues [...] Read more.
Blue gourami (gourami, Trichogaster trichopterus) is a model for labyrinth fishes (Anabantoidei) adapted to partial air breathing. Its reproductive endocrinology has been extensively studied, and transcriptomic sex differences in the gonads were described. Nevertheless, sex differences in gene expression in non-gonadal tissues ostensibly affected by the sex-specific hormonal balance, e.g., the brain, are unknown. To assess such differences, we used bulk RNA-seq to assemble and compare polyA+ transcriptomes between whole brains of four adult male and five adult female gourami, in addition to other tissues (three dorsal fin and five ovary samples) from the same female group. While all nine brain transcriptomes clustered together relative to the other tissues, they showed separation according to sex. A total of 3568 genes were differentially expressed between male and female brains; of these, 1962 and 1606 showed lower and higher expression in males, respectively. Male brains showed stronger down-regulation of specific genes, which included hormone receptors, e.g., pituitary adenylate cyclase-activating polypeptide receptor (pacap-r1). Among the genes with lower expression in male brains, multiple pathways essential to brain function were over-represented, including GABA, acetylcholine and glutamate receptor signaling, calcium and potassium transmembrane transport, and neurogenesis. In contrast, genes with higher expression in male brains showed no significant over-representation of brain-specific functions. To measure the mRNA levels of specific hormone receptors known from prior studies to regulate reproductive function and behavior in gourami and to validate RNA-seq results for these specific genes, we performed RT-qPCR for five receptors, pacap-r1, gonadotropin-releasing hormone 2 receptor (gnrh2r), kisspeptin receptor 1 (gpαr1/kiss1), insulin-like growth factor 1 receptor (igf1r), and membrane progesterone receptor 1 (mpr1), in the brain RNA sample groups. Of these, pacap-r1 showed a significant, three-fold down-regulation, while gpαr1/kiss1 showed a significant two-fold down-regulation in male vs. female gourami brains. Our results are novel in describing the suppression of brain function-related gene expression in male, as compared to female, gourami brains. Further research is needed to assess the behavioral significance of this effect and its prevalence in other vertebrate groups. Full article
(This article belongs to the Section Physiology and Biochemistry)
Show Figures

Figure 1

Figure 1
<p>Scheme of experimental design and analysis.</p>
Full article ">Figure 2
<p>Adult gourami brain transcriptomes tend to cluster by sex. (<b>A</b>) PCA of all tissues; (<b>B</b>) PCA of brain samples; (<b>C</b>) hierarchical clustering (HCL) of brain samples; (<b>D</b>) volcano plot of brain DEseq2 results (male vs. female, significantly up- or down-regulated genes in red and blue, respectively); (<b>E</b>) heatmap of significantly DE genes from DEseq2 results (all tissues).</p>
Full article ">Figure 3
<p>G.profiler graphs and compact tables of selected over-represented terms among genes that were (<b>A</b>) down-regulated and (<b>B</b>) up-regulated in male vs. female gourami brains.</p>
Full article ">Figure 4
<p>A. Relative mean mRNA abundance (based on RT-qPCR), in female and male gourami brains, of hormone receptors as follows: <span class="html-italic">pacap-r1</span> (pituitary adenylate cyclase-activating polypeptide receptor 1), <span class="html-italic">gnrh2r</span> (gonadotropin-releasing hormone receptor), <span class="html-italic">gpar1/kiss1</span> (G protein-coupled receptor 1 (<b>A</b>), <span class="html-italic">igf1r</span> (insulin-like growth factor 1 receptor), and <span class="html-italic">mpr1</span> (membrane progesterone receptor 1). *: <span class="html-italic">p</span> = 0.01; **: <span class="html-italic">p</span> = 0.0038 (<span class="html-italic">t</span>-test). Bars, <span class="html-italic">stdev</span> (N = 5). (<b>B</b>): Relative mean mRNA abundance (based on RNA-seq, see <a href="#app1-fishes-09-00287" class="html-app">Supplementary File S3</a>), in female and male gourami brains, of two of the same hormone receptors (<span class="html-italic">pacap-r1</span> and <span class="html-italic">igf1r</span>). *: <span class="html-italic">p</span> = 0.0153; **: <span class="html-italic">p</span> = 0.0002 (adjusted <span class="html-italic">p</span>-values, DESeq2).</p>
Full article ">Figure 5
<p>Scheme of suggested model—inter-regulation between brain function pathways and sex-specific hormone signaling in gourami. The brain–pituitary–gonad signaling axis in gourami, its principal hormones, and receptors have been extensively described by the first author and colleagues (review, [<a href="#B4-fishes-09-00287" class="html-bibr">4</a>]). The feedback of sex hormones affecting the brain is known from zebrafish and other species; the male-specific down-regulation of genes involved in essential brain functions is reported in the current study.</p>
Full article ">
14 pages, 836 KiB  
Article
Effects of the Neuropeptides Pituitary Adenylate Cyclase Activating Polypeptide and Vasoactive Intestinal Peptide in Male Fertility
by Roba Bdeir, Maha S. Al-Keilani and Khaldoun Khamaiseh
Medicina 2024, 60(4), 652; https://doi.org/10.3390/medicina60040652 - 18 Apr 2024
Viewed by 1986
Abstract
Background and Objectives: The neuroendocrine system plays a crucial role in regulating various bodily functions, including reproduction, with evidence suggesting its significant involvement in male fertility and sperm development. Vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase activating polypeptide (PACAP) are expressed [...] Read more.
Background and Objectives: The neuroendocrine system plays a crucial role in regulating various bodily functions, including reproduction, with evidence suggesting its significant involvement in male fertility and sperm development. Vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase activating polypeptide (PACAP) are expressed in both male and female reproductive tissues, influencing penile erection and regulating steroidogenesis in males. Therefore, our study aimed to compare the protein levels of VIP and PACAP in seminal plasma between healthy controls and sub-fertile patients. Additionally, we sought to correlate the levels of these biomarkers with clinical, functional, and laboratory findings in the participants. Materials and Methods: The study included a total of 163 male participants for analysis. The participants were further stratified into subgroups of fertile and sub-fertile men of four subgroups according to the 2021 WHO guidelines. Seminal plasma concentrations of the neuropeptides VIP and PACAP were measured using human enzyme-linked immunosorbent assay technique. Results: The findings showed statistically significant differences in total sperm count, sperm concentration, total motility, and vitality (p < 0.001) between the fertile group and the sub-fertile group. Specifically, significant differences found between healthy males and oligoasthenospermic patients (p = 0.002), and between asthenospermic and oligoasthenospermic patients (p = 0.039). An ROC analysis showed associated sensitivity and specificity values of 62.2% and 55.6%, respectively, to PACAP seminal levels differentiated between sub-fertile patients from fertile males (p = 0.028). No significant difference in seminal levels of VIP was found between the sub-fertile and fertile groups. Conclusions: Previous research leads to the point of PACAP active involvement in spermatogenesis. In accordance to our study, in human semen samples, we have seen a significance change in PACAP levels amongst patients with low sperm count or with both low sperm count and low motility, hinting at its contribution and acting as a possible factor in this complex process. Thus, alterations in the levels or actions of these neuropeptides have been associated with certain reproductive disorders in males. Full article
Show Figures

Figure 1

Figure 1
<p>PACAP and VIP seminal levels in sub-fertile group versus fertile healthy males. The seminal levels of two neuropeptides among two groups (SF: sub-fertile male; F: fertile male) are shown. (<b>A</b>) PACAP: pituitary adenylate cyclase-activating peptide; * <span class="html-italic">p</span> = 0.011. (<b>B</b>) VIP: vasoactive intestinal peptide; ns = non-significant. <span class="html-italic">p</span>-values were calculated with an independent <span class="html-italic">t</span>-test.</p>
Full article ">Figure 2
<p>Receiver operating characteristic (ROC) curve showing cut-off of PACAP seminal level for the diagnosis of male infertility. PACAP: pituitary adenylate cyclase-activating peptide; AUC: area under the curve.</p>
Full article ">Figure 3
<p><b>Comparison of PACAP and VIP seminal levels between sub-fertile groups and fertile healthy males.</b> One-way ANOVA test followed by Tukey test to find significant differences between groups among (<b>A</b>) PACAP seminal levels, where significant is noted by * <span class="html-italic">p</span> = 0.002 and ** <span class="html-italic">p</span> = 0.039, while no bar was not significant, and (<b>B</b>) VIP seminal levels, where ns (= not significant) differences were found amongst all groups. <span class="html-italic">p</span> ≤ 0.05 was considered statistically significant. PACAP: pituitary adenylate cyclase-activating peptide; VIP: vasoactive intestinal peptide; F: fertile healthy males. Sub-fertile males were further stratified into four groups: O: oligospermic patients; A: asthenospermic patients; OA: oligoasthenospermic patients; Z: azoospermic patients.</p>
Full article ">
Back to TopTop