Next Article in Journal
The Role of ATF3 in Neuronal Differentiation and Development of Neuronal Networks in Opossum Postnatal Cortical Cultures
Next Article in Special Issue
The Role of Glucocorticoid Receptor in the Pathophysiology of Pituitary Corticotroph Adenomas
Previous Article in Journal
New Insights into and Emerging Roles of Animal Models for Neurological Disorders
Previous Article in Special Issue
Difference in miRNA Expression in Functioning and Silent Corticotroph Pituitary Adenomas Indicates the Role of miRNA in the Regulation of Corticosteroid Receptors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Effects of Pituitary Adenylate Cyclase Activating Polypeptide on Cell Death

Department of Anatomy, MTA-PTE PACAP Research Team, University of Pecs Medical School, 7624 Pecs, Hungary
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(9), 4953; https://doi.org/10.3390/ijms23094953
Submission received: 30 March 2022 / Revised: 25 April 2022 / Accepted: 28 April 2022 / Published: 29 April 2022
(This article belongs to the Special Issue Molecular Biology of the Pituitary 2.0)

Abstract

:
Pituitary adenylate cyclase activating polypeptide (PACAP) was first isolated as a hypothalamic peptide based on its efficacy to increase adenylate cyclase (AC) activity. It has a widespread distribution throughout the body including the nervous system and peripheral organs, where PACAP exerts protective effects both in vivo and in vitro through its anti-apoptotic, anti-inflammatory, and antioxidant functions. The aim of the present paper was to review the currently available literature regarding the effects of PACAP on cell death in vitro in neural and non-neural cells. Among others, its effect on apoptosis can be detected in cerebellar granule cells against different toxic stimuli. Different neural cell types from the cerebral cortex are also prevented from cell death. PACAP also shows effects on cell death in cells belonging to the peripheral nervous system and protects both neural and non-neural cells of sensory organs. In addition, cell survival-promoting effect can be observed in different peripheral organ systems including cardiovascular, immune, respiratory, gastrointestinal, urinary, and reproductive systems. The studies summarized here indicate its noteworthy effect on cell death in different in vitro models, suggesting PACAP’s potential therapeutic usage in several pathological conditions.

1. Introduction

Pituitary adenylate cyclase activating polypeptide (PACAP) was first isolated as a hypothalamic peptide based on its efficacy to increase adenylate cyclase (AC) activity [1]. PACAP belongs to the secretin/glucagon/vasoactive intestinal peptide (VIP) family. It exists in two isoforms: PACAP38 with 38 and PACAP27 with 27 amino acid residues [2]. Its primary structure is evolutionarily conserved [2], indicating its possible preserved biological role. The peptide acts through G-protein coupled receptors: PAC1, VPAC1, and 2. The PAC1 receptor is the specific receptor for PACAP binding with high affinity, while VPAC receptors bind PACAP and VIP with similar affinity [2]. The N-terminally truncated peptide form, PACAP6-38, was discovered as an antagonist [3]. It antagonizes both the PACAP specific PAC1 receptor and the VPAC2 receptor [4]. After conformational changes of PACAP receptors caused by PACAP binding, different downstream pathways can be initiated. First, adenylate cyclase can be activated, leading to cAMP production with subsequent activation of protein kinase A (PKA). This might be crucial in many physiological functions of PACAP [2]. Through the activation of the extracellular signal-regulated kinase (ERK) pathway, downstream molecules of apoptotic signaling cascade can be influenced, leading to cell survival. PKA activation is also thought to be responsible for the anti-inflammatory actions of PACAP [5]. Another possible pathway activated by PACAP binding is the phospholipase C (PLC)/calcium (Ca2+) cascade. Increased intracellular Ca2+ participates in exerting PACAP’s biological activities (e.g., cell migration, secretion of neurotransmitters and neurohormones, and glial cell differentiation) [5]. In addition, the transcription factor, cAMP response element-binding protein (CREB), can also be activated through PACAP’s receptors. PKA, ERK, and calmodulin-dependent protein kinase (CamK) are capable of activating CREB [5], which is responsible for cell differentiation processes and neurohormone/neurotransmitter secretion of the hypothalamus [2,6,7]. Moreover, other G protein-independent pathways can be initiated by PAC1 and VPAC receptors [5]. Early studies have already focused on the protective effects of PACAP in both the nervous system and peripheral organs [2,8,9,10,11,12]. Based on these studies, it seems that one of the conserved functions of PACAP is its cytoprotective actions, as anti-apoptotic effects have also been shown in invertebrates and molluscan salivary glands [13]. It exerts these protective functions through its anti-apoptotic, anti-inflammatory, and antioxidant effect [2]. Numerous studies have been performed to investigate the effect of PACAP on cell death processes. The aim of our present paper was to review the effects of PACAP on cell death in different cell types including neural cells and cells of the peripheral tissues and organs. The review focuses on the current knowledge obtained from in vitro studies.

2. Types of Cell Death

Different types of cell death processes can be distinguished. Apoptosis is a highly regulated form of programmed cell death in multicellular organisms. It is associated with characteristic changes of cell morphology (e.g., shrinkage, nuclear fragmentation, chromatin condensation, and DNA fragmentation) [14]. Another type of programmed cell death is autophagy (or autophagocytosis), which plays a crucial role in the homeostasis of cells. This process serves the degeneration and recycling of dysfunctional or unnecessary cellular cytoplasmic components with lysosomes. Insufficient autophagy is widely considered to be associated with human neurodegenerative diseases and cancer [15]. Oncosis can be described as a form of programmed cell death with characteristics such as cellular and organelle swelling, increased membrane permeability, and blebbing. These events may be caused by the failure of ionic pumps of the cell plasma membrane and is usually accompanied by karyolysis and can evolve to necrosis. Ischemia or agents interfering with ATP generation or increasing membrane permeability can lead to oncosis [16]. The other main type of cell death is non-programmed necrotic cell death. Necrosis is an energy independent form of traumatic cell death, which is triggered by external or internal factors (radiation, chemicals, hypoxia, temperature) that lead to upregulation of pro-inflammatory molecules in the cells. These events do not follow the signaling pathways of apoptosis, and they finally cause the rupture of the cell membrane and uncontrolled release of cell components into the surrounding extracellular space. The inflammatory response in the adjacent areas attracts leukocytes and phagocytes to eliminate dead cells, however, contribution of leukocytes can cause collateral tissue damage due to the release of their substances [14]. Necroptosis is a regulated type of necrosis, which can be considered as a special type of programmed cell death. When apoptotic pathways are blocked, necroptosis can be initiated [14]. Most studies investigating the protective mechanism exerted by PACAP have shown that of these above-mentioned types of cell death, PACAP mainly counteracts apoptotic pathways. Recent investigations, however, have also described its effects on autophagy processes [17].

3. Effects of PACAP on Cell Death in the Central Nervous System

PACAP has well-known protective effects in the entire body including the central nervous system. Effects of PACAP on cell death in vitro in the nervous system are summarized in Table 1.

3.1. Cerebellum

Early studies investigated the cytoprotective effect of PACAP in primary cell culture of rat cerebellar granule cells. Kienlan Campard et al. [18] detected its survival-enhancing effect in experiments demonstrating the in vitro environment of serum and potassium deprivation. The number of living cells was elevated by PACAP27 or PACAP38 addition similarly to BDNF (brain-derived neurotrophic factor). Both PACAP isoforms were able to reduce DNA fragmentation [18]. This survival-promoting effect in serum-deprived cells could already be observed at picomolar concentrations [19]. The cAMP/PKA signaling cascade seems to be the major pathway involved in the survival-enhancing effect of PACAP against apoptosis evoked by serum and K+ withdrawal [18,19,20]. The anti-apoptotic effect of PACAP was enhanced by ethanol although PACAP was able to reduce ethanol-induced apoptosis [21]. These results indicate a fine balance between inhibition and enhancement in the action of neuropeptides [20].
Several other research groups have investigated the effects of PACAP on programmed cell death in rat cerebellar granule cells. Chang and colleagues [22] found a dose-dependent survival promoting effect of PACAP. This could be even seen after a 3-day potassium deprivation. Similar results were published by Gonzalez and co-workers [23] since PACAP dose-dependently prevented cell death in rat cerebellar granule cells. Besides the increased number of living cells, improvement in neurite outgrowth could also be observed [23]. Further studies have explored the same protection exerted by PACAP in rat cerebellar granule cells, where it was shown to protect them against serum and potassium deprivation [24,25,26]. PACAP mitigated the extent of DNA fragmentation and increased the ERK activity, likely controlled by the PKA/ERK cascade [24,25]. In addition, other studies have shown PACAP’s anti-apoptotic effect in the granule cells of developing rat cerebellum in ceramide-induced apoptosis. PACAP counteracted the changes evoked by C2-ceramide administration. PACAP was able to decrease the activation of caspase-3, the elevated Bax expression, cytochrome c release, and restore the ceramide-altered mitochondrial activity. Restoration of mitochondrial activity could be abrogated by inhibition of MEK [27,28]. Moreover, Vaudry and colleagues demonstrated the role of both the PKA and PKC signaling mechanism in PACAP-mediated caspase-3 suppression [29]. PACAP’s protection was also demonstrated against H2O2-induced oxidative stress [30] and protected granule cells dose-dependently through the cAMP and MAP kinases. Besides a decrease in caspase-3 activity, reduced DNA fragmentation and restoration of mitochondrial membrane potentials were also described [30]. Vaudry and colleagues [31] described that the anti-apoptotic effect did not change when cells were incubated with PACAP continuously for 48 h or only for 1 h. This finding suggests that a short-term stimulation by PACAP leads to a long-lasting response. Although PACAP could decrease the caspase-3 activation in Abeta25–35 neurotoxicity, it did not prevent membrane potential alteration and cell death [32]. Furthermore, Ito and co-workers [26] also described the anti-apoptotic effect in rat cerebellar granule cells exposed to 4-hydroxynonenal.
Cisplatin is a widely used chemotherapeutic agent having well-known neurological side effects [33]. Interestingly, PACAP could prevent DNA laddering, caspase-3 and -9 activation, and Bax induction evoked by cisplatin exposure in quiescent neurons of rat cerebellar granule cell culture and macaque organotypic cerebellum slices. In contrast, these protective actions could not be detected in CHO proliferating ovarian cells, indicating the possible therapeutic use of PACAP against neurologic side effects. This could prevent cisplatin-induced neurotoxicity by maintaining its chemotherapeutic efficacy [34].
Not only exogenously given PACAP, but also its endogenously present form may exert survival-promoting action in mouse cerebellar granule cells since cells treated with the PACAP antagonist, PACAP6-38, showed decreased cell survival [35]. This was supported by studies performed on cerebellar neurons derived from PACAP-deficient mice since those cells responded with higher sensitivity to both ethanol toxicity and oxidative stress compared to cells obtained from wild type animals [36].

3.2. Cerebrum

The effect of PACAP against cell death was also tested in neuronal cultures in in vitro models of various neuronal injuries. Morio and co-workers [37,38] found a protective effect exerted by PACAP pretreatment against glutamate-induced excitotoxicity in rat cortical neurons. Endogenous PACAP seemed to have similar actions since upregulation of PACAP mRNA expression was detected in cultured rat cortical neurons after glutamate exposure. The protective role of PACAP was supported by cell survival-worsening effect of the PACAP antagonist, PACAP6-38, even if cells were not exposed to excitotoxic glutamate [39]. In addition, PACAP was also able to increase the cell viability of rat cortical neurons exposed to NMDA (N-methyl-D-aspartate) or serum deprivation [40]. Skoglosa and colleagues [41] detected the same action against ionomycin-induced apoptosis. Furthermore, rat cortical neurons could also be protected from Tat (transactivator of transcription) toxicity [42] and oxygen-glucose deprivation-reperfusion [43] by PACAP. PACAP and its transduction led to protection against sodium nitroprusside toxicity in primary cortical neurons through caspase-3 deactivation and Bcl-2 induction [44]. It could also promote cell survival in cortical neurons exposed to thrombin or thrombin receptor activating peptide with its anti-caspase effect, but without influencing Bcl-2 level [45]. BDNF is related to PACAP’s protective mechanism [40].
Investigating rat cortical astrocytes, PACAP was shown to prevent decline in cell viability caused by H2O2-induced oxidative stress with the suppression of caspase-3 activation [46]. In hCMEC/D3 human cerebral endothelial cells, no noteworthy effect on cell death could be observed [47]. Furthermore, protective actions against LPS neurotoxicity were registered in mesencephalic neuron/glia cultures [48] and mixed cortical neuron/glia cultures [49]. PACAP was identified to be effective against 6-OHDA injury, leading to loss of mesencephalic DAergic neurons [50]. MPP+ (1-methyl-4-phenylpyridinium) toxicity, leading to loss of dopaminergic neurons, was mitigated by PACAP in mesencephalic neuron/glia culture [48]. Cell survival-promoting actions could also be detected in dopaminergic HS-SY5Y human neuroblastoma cell line treated with high ethanol or high nicotine concentrations [51], salsolinol [52], or microglia-derived mediators [53]. In the same SH-SY5Y cell line, PACAP downregulated MPP+-induced autophagy [17]. It reduced the cell death, maintained mitochondrial function, and decreased autophagic morphological changes [17].
Hypoglycemia-induced impaired cell viability could be improved with PACAP treatment in neural stem cells in correlation with decreased apoptosis (increased Bcl-2, suppressed caspase-3) and endoplasmic reticulum stress [54]. Similarly, lipoapoptosis evoked by palmitate-induced lipotoxicity and ketamine toxicity were also attenuated by PACAP via PAC1 receptor activation [55,56]. Cell death related to HIV envelope protein gp120 toxicity was attenuated by both PACAP isoforms in hippocampal cell culture [57]. In a recent study, PACAP was demonstrated to act protective in an in vitro model of Huntington’s disease. It attenuated cell death induced by mutant huntingtin with a reduction in caspase-3 activity and increased in pERK and Akt [58]. Broome et al. [59] described PACAP’s cell viability-increasing action in rotenone exposure in BV2 microglial cell culture.

3.3. Spinal Cord

PACAP was shown to be protective against glutamate excitotoxicity in rat motoneurons by acting via the cAMP-protein kinase A signaling pathway [60]. In an in vitro model of familial ALS, PACAP was able to protect motor neurons against serum deprivation [61]. Furthermore, in the same ALS model, it reduced desferrioxamine mesylate salt (DFX)-induced cell death by modulating the autophagy with elevated expression of p62II and suppression of LC3II [62]. In both cases, PACAP led to increased ERK1/2 phosphorylation [61,62].

4. Effects of PACAP on Cell Death in the Peripheral Nervous System

MPAPO, a highly stable PACAP-27-derived mutant peptide, and PACAP were proven to mitigate apoptosis evoked by hypoxic conditions in trigeminal ganglion cells. Both peptides decreased caspase-3 activity and cytochrome c release [63]. PACAP was shown to prevent rat schwannoma cells from apoptosis induced by serum deprivation by influencing components of the apoptotic signaling cascade. Interestingly, it suppressed both the anti-apoptotic Bcl-2 and pro-apoptotic Bax [64].

PC12 Cells

PACAP was shown to act protective in neuron-like rat pheochromocytoma, PC12, cells against prion toxicity. It increased cell viability and deactivated the caspase-3 of PC 12 cells exposed to the prion protein fragment [65]. In addition, it could also prevent neuronal toxicity induced by the beta-amyloid peptide [66]. PACAP does not act only in the in vitro model of Alzheimer’s disease, but its protection could also be detected in rotenone toxicity, which is implicated in the pathogenesis of Parkinson’s disease. This neuroprotective effect is associated with the activation of the MAP kinase pathways by PKA and the inhibition of caspase-3 activity [67]. Anisomycin-induced DNA fragmentation was reduced by PACAP through the PKA pathway [68]. In the molecular background of PACAP’s effects in PC12 cells, stathmin 1 phosphorylation could also be identified [69]. Moreover, it could also exert protective actions against MPP+ toxicity, associated with dopaminergic neuron death, in PC12 [70] and neuro-2a neuroblastoma cells [71].

5. Effects of PACAP on Cell Death in the Sensory Organs

Effects of PACAP on cell death in vitro in the sensory organs are summarized in Table 2.

5.1. Eye

5.1.1. Retina

The protective effect of PACAP on adult retinal pigment epithelial (ARPE-19) cell line has been examined by several research groups. In H2O2-induced oxidative stress, PACAP was found to be anti-apoptotic in a dose dependent manner [72]. A subsequent study also revealed the underlying molecular biological mechanism. PACAP activated the ERK1/2 and CREB pathways, while it attenuated the expression of the pro-apoptotic p38 and JNK in ARPE-19 cells [73]. The activating effect of PACAP on ERK1/2 was also confirmed by another research group [74] who investigated the protective effect of PACAP on the blood–retina barrier by using a diabetic macular edema model. The edema was mimicked by maintaining the ARPE-19 cells in high glucose medium (hyperglycemia) and treating them with desferrioxamine-mesylate salt, a hypoxia-mimetic agent. According to their results, the peptide prevented hypoxia-induced apoptosis through the activation of the PI3K/Akt and MAPK/ERK signaling pathways. Subsequent research further deepened the knowledge about the underlying mechanisms [75]. In a similar diabetic macular edema model, they showed that in order to protect the ARPE cells, PACAP could decrease HIF-1α and increase HIF-3α expression. Furthermore, PACAP was able to attenuate the expression of the pro-apoptotic p38, which was activated by the hyperglycemia induced elevated VEGFR1 and 2 levels.
The other cell line that has been thoroughly investigated is the retinal ganglion cell line (RGC-5). Cells were protected against UV-B radiation induced apoptosis [76] by both PACAP and cyclopeptide C*HSDGIC* (CHC). CHC, which is formed by the cyclization of PACAP (1–5) and has been proven to be a potent agonist of PACAP [77], was used to avoid the fast degradation of PACAP. In a subsequent study, CHC was again proved to protect the RGC cells against ultraviolet B irradiation [78]. CHC could not only inhibit apoptotic cell death measured by the MTT assay, flow cytometry, and fluorescent microscopy, but also decreased the amount of reactive oxygen species (ROS) and attenuated the expression of the pro-apoptotic Bax and Bcl-2. In their next study, they investigated the regulation of mitochondrial function exerted by PACAP [79]. With the use of a cell counting kit and flow cytometry, they showed that PACAP is protective in serum deprivation induced apoptosis. Besides facilitating apoptosis, serum deprivation also increases the level of ROS, leading to the loss of mitochondrial membrane integrity. According to the findings, PACAP could decrease the level of ROS in serum deprivation, helping to maintain mitochondrial function in the retinal ganglion cells.
Silveira and co-workers [80] detected the protective effect of PACAP against anisomycin-induced cell death in rat retina explants. PACAP exerted anti-apoptotic action since it led to a lower number of degenerating profiles observed with the TUNEL assay or measuring caspase-3 activity. Similarly to other studies, the regulatory role of the cAMP/PKA pathway was described. PACAP was also shown to prevent from thapsigargin-induced photoreceptor degeneration [80].

5.1.2. Cornea

In human corneal endothelial cells isolated from donor’s cornea, PACAP exerted a cell survival-enhancing effect in cellular death evoked by growth factor deprivation [81]. In addition, PACAP protected human corneal endothelial cells against ultraviolet B (UV-B) irradiation by influencing apoptotic pathways. It inhibited Bax and caspase-3 activity and upregulated Bcl-2 protein [82].

5.2. Inner Ear

Oxidative stress is involved in the pathogenesis of several ototoxic insults. PACAP was able to protect against apoptosis evoked by H2O2-induced oxidative stress in a mixed culture of sensory hair cells and supporting cells from the sensory epithelium of chicken inner ear. It reduced the number of apoptotic cells and suppressed the oxidative stress-induced caspase-3 activation [83].

5.3. Olfactory System

In mouse olfactory epithelium, PACAP prevented TNFα-induced apoptosis. It protected live slices of olfactory epithelium and it exerted this protection in olfactory placodal cell lines. TNFα-induced activation of initiator caspase, caspase-8, was reversed by PACAP treatment in OP6 olfactory placodal cell lines. Involvement of the PLC pathway in the protection from TNFα was proven, but blocking it led only to an incomplete block in the protection, hence additional pathways should contribute [84].

6. Effects of PACAP on Cell Death in Peripheral Organs

The effects of PACAP on cell death in vitro in the peripheral organs are summarized in Table 3.

6.1. Cardiovascular System

The effects of PACAP in the cardiovascular system have been widely studied. Besides in vivo investigations and human observations [85], PACAP has been described to act in the cardiovascular system in vitro. It was proven to protect EOMA mouse endothelial cells from hemangioendothelioma from apoptosis evoked by H2O2-treatment [86]. In the molecular background, suppression of pro-apoptotic JNK and p38 phosphorylation and enhancement of anti-apoptotic ERK phosphorylation were identified. The PACAP antagonist PACAP6-38 abolished all these effects. Its cytoprotective effect could also be detected in human endothelial colony-forming cells exposed to TNFα, where PACAP was able to decrease the number of apoptotic cells [87]. PACAP was detected to protect rat neonatal cardiac myocytes in culture against H2O2-induced oxidative stress, it increased the number of living cells, and decreased that of the annexin V positive early apoptotic cells [88,89]. PACAP also mitigated caspase-3 activity and increased the expression of anti-apoptotic Bcl-2 and phospho-Bad. PACAP was able to inhibit ASK-1 activation induced by oxidative stress [88,89]. The in vitro cardioprotective action of PACAP was shown against simulated ischemia/reperfusion injury in rat cardiomyocytes. Apoptotic signaling pathways were influenced by PACAP, and induced the phosphorylation of PKA, Akt, and Bad. Reduction in both 14-3-3 and Bcl-xL caused by simulated ischemia/reperfusion were counteracted by PACAP treatment. Similarly to cell injury provoked by H2O2, PACAP was effective in reducing increased caspase-3 activity in ischemia/reperfusion injury [90]. Effects of PACAP in cardiomyocytes can be abolished by the PACAP antagonist, PACAP6-38 [88,90]. Further studies in rat neonatal cardiomyocytes investigating the relation between PACAP and preconditioning against ischemia/reperfusion induced cardiomyocyte injury found that both were cardioprotective and were able to reduce the initiator caspase-8 activity, but their effects were not additive [91]. Li and colleagues [92] studied the effects of PACAP against irradiation in rat embryonic ventricular H9C2 cardiomyoblast cells. PACAP could diminish the survival-worsening effect of irradiation by influencing pro-apoptotic gene-regulated signaling.

6.2. Immune System

Apoptosis plays a crucial role during the development of lymphocytes. In mature peripheral T cells, PACAP inhibited antigen-induced apoptosis by reducing Fas ligand expression [93]. Both isoforms of PACAP were detected to protect CD4+/CD8+ thymocytes from glucocorticoid-induced apoptosis, indicating the possible role of PACAP in T lymphocyte maturation [94].

6.3. Respiratory System

Few data in the literature are available regarding the effects of PACAP against cell death in the respiratory system. Cell survival influencing effect of several neuropeptides was tested in L2 cells, originally derived from pneumocyte type II of adult rat lung. Among others, PACAP27 was able to reduce the toxic effect of cigarette smoke extract with the inhibition of cell death and caspase-3 activity [95,96].

6.4. Gastrointestinal Tract

6.4.1. Intestines

Aside from performing several effects on physiological intestinal processes, for instance, motility [97] and growth factor secretion [98], PACAP possesses an influencing effect on cell viability processes. Our research group investigated INT407 cells, human embryonic jejunal, and ileal cells. PACAP was able to ameliorate the cell survival decreasing effect of H2O2-induced oxidative stress, but only if it was applied simultaneously. In the case of other investigated stressors, CoCl2-induced in vitro hypoxia and gamma irradiation, but failed to protect the INT407 cells from cell death measured by the MTT cytotoxicity assay. Not only was the exogenously given PACAP proven to increase cell survival against oxidative stress, but endogenous PACAP was also demonstrated to be protective since posttranscriptional RNA silencing of PACAP led to the higher vulnerability of H2O2-treated INT407 cells [99]. Le et al. [100] detected the Fas-R expression regulating effect of PACAP, suggesting a possible effect in apoptotic signaling in HCT-8 human colonic tumor cells.

6.4.2. Liver

There is some experimental evidence proving the protective role of PACAP in different hepatological pathologies in vivo [11]. PACAP was shown to ameliorate apoptotic/necrotic effect of TNFα/actinomycin D administration and H2O2-treatment. Both PACAP isoforms were effective, where they could influence not only the number of dead cells, but both lactate dehydrogenase (LDH) and alanin aminotransferase (ALT) releases were alleviated [101]. This survival-promoting effect observed in mouse hepatocytes could not be detected in human hepatocytes. PACAP did not improve the cell survival of H2O2-treated WRL-68 hepatocytes and Hep-G2 hepatocellular carcinoma cells [102].

6.4.3. Pancreas

Apoptosis of β cells is a crucial element in the evolution of diabetes mellitus [103]. Onoue and colleagues [104] performed experiments studying the potential protective effects of various neuropeptides including PACAP using RIN-m5F rat pancreatic β cells. PACAP showed a cell survival enhancing effect since it inhibited streptozotocin-induced LDH release. It was able to influence apoptotic signaling pathways, it decreased mRNA levels of pro-apoptotic Noxa and Bax, and increased the mRNA level of anti-apoptotic Bcl-2. Not only did the exogenously added PACAP influence pancreas β cells, but overexpression of Adcyap1 encoding PACAP isoforms led to increased cell survival against cytokine-mediated apoptosis in NIT-1 mouse insulinoma cells [105]. PACAP failed to be protective against apoptosis evoked by exposure to cytokines in the EndoC-βH1 human β cell line [106].

6.5. Urinary System

PACAP and its receptors have been shown to be present in the kidney and lower urinary tract [107,108]. PACAP exerts several functions in the urinary system including its cell survival enhancing effect [11,107]. Exogenously given PACAP was shown to decrease the toxic effect of hydrogen-peroxide-induced oxidative stress in primary renal cell culture of newborn rats. It was already effective at 100 pM concentration, while it did not influence the cell proliferation rate if added alone [102]. In accordance with the results of experiments studying exogenous PACAP, this cell viability improving effect could also be observed in the primary renal cell culture of PACAP knockout mice. Animals lacking endogenous PACAP displayed higher vulnerability against oxidative stress [109] and cobalt chloride-induced hypoxia [110]. Effects of PACAP on cell survival could also be detected in other renal pathologies [111]. Arimura et al. studied the effect of PACAP in various models of myeloma nephropathy [112,113,114,115]. Investigating the effect of the peptide on purified κ light chain-treated SV40 immortalized human proximal tubule culture, they described the survival-enhancing effect: it could decrease the light chain induced cell death indicated by cellular detachment from the cultured plate [112]. Furthermore, PACAP was shown to exert an anti-apoptotic action in mineral oil induced in vitro hypoxia in primary cultures of proximal tubule epithelial cells of MyD88+/+ and MyD88−/− mice [116]. Although PACAP showed protective effects in in vitro models of different renal pathologies, it could not act against albumin exposure, mimicking proteinuria-related cell damage in HK-2 cells [117].
Possible effects of PACAP have also been widely investigated in different in vitro models of drug-induced nephropathies, generally using the human proximal tubule cell line HK-2. PACAP was able to mitigate the cell survival-worsening effect of gentamicin treatment in HK-2 cells assessed by the MTT assay [118]. In addition, PACAP was also proven to act against chemotherapeutic-induced cellular damage in this cell line. It could counteract the cisplatin-induced apoptosis assessed by the determination of DNA fragmentation. In these series of experiments, PACAP-induced suppression of p53 activation was identified in the molecular background mechanism. It could also counteract the p53-activated TNFα-secretion and transcriptional control of caspase-7 and PARP-1. Furthermore, PACAP treatment led to the restoration of APE-1, which is essential in the DNA repair mechanism. It could also influence p53-independent apoptotic signaling, increased the levels of anti-apoptotic Bcl-2 and Bcl-XL, and decreased that of pro-apoptotic Bax [119]. The anti-apoptotic effect was also demonstrated in primary cultures of mouse renal proximal tubule cells, where PACAP treatment could decrease the ratio of annexin V positive apoptotic cells both before and after the cisplatin exposure [120]. Khan et al. conducted studies exploring the effects of PACAP against cyclosporine A and contrast-induced nephropathies in HK-2 cells [121,122]. It prevented cyclosporine A-induced morphological changes in the human proximal tubule cells observed with phase-contrast microscopy and reduced LDH release and DNA fragmentation [121]. Moreover, PACAP was able to reduce contrast medium induced LDH release and DNA fragmentation in both cases of ionic (Urografin) and non-ionic (iohexol) contrast media [122].

6.6. Reproductive System

Numerous sources in the literature have described the functions and occurrence of PACAP in the reproductive organs [11,123,124,125]. In the female reproductive system, investigations performed on HTR-8/SVneo nontumorous primary trophoblast cells proved the cell survival increasing effect of PACAP, but only if PACAP was used as a pretreatment [126]. Trophoblast-related, JAR human choriocarcinoma cells behaved in a surprising manner. In contrast to most of the cell types investigated, PACAP was shown to enhance the survival-worsening effect of the applied H2O2- and CoCl2-treatment. PACAP performed this survival-decreasing effect via changes of signaling pathways, and decreased the activation of Akt, ERK-1/2, p38, JNK/SAPK, and Bax [127]. Surprisingly, in JAR cells, PACAP6-38, which is usually used as an antagonist, behaved as an agonist exerting similar functions to PACAP on MAPK signaling [128]. Furthermore, it did not take any action if JAR cells were treated with methothrexate [129]. Similarly, PACAP failed to act against methothrexate-induced cellular insult in human invasive proliferative extravillous cytotrophoblast (HIPEC) cells [126]. PACAP’s anti-apoptotic effect was also studied in the male reproductive system. Shan and co-workers found a cytoprotective effect in GC-2 spermatocyte culture against palmitate-induced apoptosis with the attenuation of palmitate-induced activation of caspase-3 and Bax [130]. It could also ameliorate the Bcl-2 downregulation [130]. Gutiérrez-Cañas and co-workers [131] found a cell viability improving effect against serum deprivation induced apoptosis. Anti-apoptotic action of PACAP could not only be detected in nontumorous cells, but also in PC-3 human prostate cancer cells, where PACAP exerted a cell viability improving effect against serum deprivation induced apoptosis with increased Bcl-2 and procaspase-3 levels [131].

6.7. Glands

In contrast to the general cytoprotective actions of PACAP, it promoted apoptosis in MCF-7 breast cancer cells. It increased the expression of pro-apoptotic Bax and decreased the level of anti-apoptotic Bcl-2 [132]. The conserved anti-apoptotic effect of PACAP has been proven in salivary gland extracts of Helix pomatia. It protected against dopamine- and colchicine-induced apoptosis with the suppression of caspase-3 activity [13].
Furthermore, the cell survival promoting effect of PACAP was detected in the effector hormone-producing organ of the circadian biological rhythms. Interestingly, this effect could only be observed in the dark phase, indicating that the time of the day can influence the effect of PACAP on cell viability processes [133]. These and the above results also confirm that the signaling pathways and effects of PACAP in tumorous cells can be different from those of normal cells, and PACAP can have either pro-or anti-apoptotic effects or no effect at all. These variable effects depend on the cell type, circadian rhythm, damaging insult, and the expression of different receptor splice-variant.

7. Conclusions

The present paper summarized the currently available data in the literature regarding the in vitro effects of PACAP on cell death processes. In most cell types, PACAP exerts a cell survival promoting action in in vitro models of different pathologies. This remarkable effect on cell death suggests its potential therapeutic usage in various pathological conditions.

Author Contributions

G.H. and D.R. conceptualized the paper. G.H. constructed the tables. D.R., B.O., E.F. and G.H. wrote parts of the review. All authors have read and agreed to the published version of the manuscript.

Funding

The research was financed by the Thematic Excellence Program 2021 Health Sub-program of the Ministry for Innovation and Technology in Hungary, within the framework of the EGA-16 project of the Pecs of University. This study was supported by the National Research, Development, and Innovation Fund (FK129190, K119759, K135457); National Brain Research Program (NAP2017-1.2.1-NKP-2017-00002); MTA-TKI-14016; GINOP-2.3.2-15-2016-00050 “PEPSYS”; “The role of neuroinflammation in neurodegeneration: from molecules to clinics”; and Higher Education Institutional Excellence Program of the Ministry of Human Capacities in Hungary: 20765/3/2018/FEKUTSTRAT, 2020-4.1.1-TKP2020—FIKP III. Project No. TKP2020-IKA-08 was implemented with the support provided from the National Research, Development, and Innovation Fund of Hungary, financed under the 2020-4.1.1-TKP2020 funding scheme.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. 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]
  2. Vaudry, D.; Falluel-Morel, A.; Bourgault, S.; Basille, M.; Burel, D.; Wurtz, O.; Fournier, A.; Chow, B.K.C.; Hashimoto, H.; Galas, L.; et al. Pituitary adenylate cyclase-activating polypeptide and its receptors: 20 years after the discovery. Pharmacol. Rev. 2009, 61, 283–357. [Google Scholar] [CrossRef] [PubMed]
  3. Robberecht, P.; Gourlet, P.; De Neef, P.; Woussen-Colle, M.C.; Vandermeers-Piret, M.C.; Vandermeers, A.; Christophe, J. Structural requirements for the occupancy of pituitary adenylate cyclase-activating peptide (PACAP) receptors and adenylate cyclase activation in human neuroblastoma NB-OK-1 cell membranes. Discovery of PACAP(6–38) as a potent antagonist. Eur. J. Biochem. 1992, 207, 239–246. [Google Scholar] [CrossRef] [PubMed]
  4. 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] [PubMed]
  5. 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]
  6. Dickson, L.; Finlayson, K. VPAC and PAC receptors: From ligands to function. Pharmacol. Ther. 2009, 121, 294–316. [Google Scholar] [CrossRef] [PubMed]
  7. Blechman, J.; Levkowitz, G. Alternative splicing of the pituitary adenylate cyclase-activating polypeptide receptor PAC1: Mechanisms of fine tuning of brain activity. Front. Endocrinol. 2013, 4, 55. [Google Scholar] [CrossRef] [Green Version]
  8. Somogyvari-Vigh, A.; Reglodi, D. Pituitary adenylate cyclase activating polypeptide: A potential neuroprotective peptide. Curr. Pharm. Des. 2004, 10, 2861–2889. [Google Scholar] [CrossRef]
  9. Reglodi, D.; Tamas, A.; Jungling, A.; Vaczy, A.; Rivnyak, A.; Fulop, B.D.; Szabo, E.; Lubics, A.; Atlasz, T. Protective effects of pituitary adenylate cyclase activating polypeptide against neurotoxic agents. Neurotoxicology 2018, 66, 185–194. [Google Scholar] [CrossRef]
  10. Reglodi, D.; Kiss, P.; Lubics, A.; Tamas, A. Review on the protective effects of PACAP in models of neurodegenerative diseases in vitro and in vivo. Curr. Pharm. Des. 2011, 17, 962–972. [Google Scholar] [CrossRef]
  11. Toth, D.; Szabo, E.; Tamas, A.; Juhasz, T.; Horvath, G.; Fabian, E.; Opper, B.; Szabo, D.; Maugeri, G.; D’Amico, A.G.; et al. Protective effects of PACAP in peripheral organs. Front. Endocrinol. 2020, 11, 377. [Google Scholar] [CrossRef] [PubMed]
  12. 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] [Green Version]
  13. Pirger, Z.; Nemeth, J.; Hiripi, L.; Toth, G.; Kiss, P.; Lubics, A.; Tamas, A.; Hernadi, L.; Kiss, T.; Reglodi, D. PACAP has anti-apoptotic effect in the salivary gland of an invertebrate species, Helix pomatia. J. Mol. Neurosci. 2008, 36, 105–114. [Google Scholar] [CrossRef] [PubMed]
  14. D’Arcy, M.S. Cell death: A review of the major forms of apoptosis, necrosis and autophagy. Cell Biol. Int. 2019, 43, 582–592. [Google Scholar] [CrossRef] [PubMed]
  15. Djajadikerta, A.; Keshri, S.; Pavel, M.; Prestil, R.; Rubinsztein, D.C. Autophagy induction as a therapeutic strategy for neurodegenerative diseases. J. Mol. Biol. 2020, 432, 2799–2821. [Google Scholar] [CrossRef]
  16. Majno, G.; Joris, I. Apoptosis, oncosis, and necrosis. An overview of cell death. Am. J. Pathol. 1995, 146, 3–15. [Google Scholar]
  17. Lamine-Ajili, A.; Fahmy, A.M.; Létourneau, M.; Chatenet, D.; Labonté, P.; Vaudry, D.; Fournier, A. Effect of the pituitary adenylate cyclase-activating polypeptide on the autophagic activation observed in in vitro and in vivo models of Parkinson’s disease. Biochim. Biophys. Acta 2016, 1862, 688–695. [Google Scholar] [CrossRef]
  18. Kienlen Campard, P.; Crochemore, C.; Rene, F.; Monnier, D.; Koch, B.; Loeffler, J.P. PACAP type I receptor activation promotes cerebellar neuron survival through the cAMP/PKA signaling pathway. DNA Cell Biol. 1997, 16, 323–333. [Google Scholar] [CrossRef]
  19. Canonico, P.L.; Copani, A.; D’Agata, V.; Musco, S.; Petralia, S.; Travali, S.; Stivala, F.; Cavallaro, S. Activation of pituitary adenylate cyclase-activating polypeptide receptors prevents apoptotic cell death in cultured cerebellar granule cells. Ann. N. Y. Acad. Sci. 1996, 805, 470–472. [Google Scholar] [CrossRef]
  20. Bhave, S.V.; Hoffmann, P.L. Phosphatidylinositol 3′-OH kinase and protein kinase A pathways mediate the anti-apoptotic effect of pituitary adenylate cyclase activating polypeptide in cultured cerebellar granule neurons: Modulation by ethanol. J. Neurochem. 2004, 88, 359–369. [Google Scholar] [CrossRef]
  21. Vaudry, D.; Rousselle, C.; Basille, M.; Falluel-Morel, A.; Pamantung, T.F.; Fontaine, M.; Fournier, A.; Vaudry, H.; Gonzalez, B.J. Pituitary adenylate cyclase-activating polypeptide protects rat cerebellar granule neurons against ethanol-induced apoptotic cell death. Proc. Natl. Acad. Sci. USA 2002, 99, 6398–6403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Chang, J.Y.; Korolev, V.V.; Wang, Z. Cyclic AMP and pituitary adenylate cyclase-activating polypeptide (PACAP) prevent programmed cell death of cultured rat cerebellar granule cells. Neurosci. Lett. 1996, 206, 181–184. [Google Scholar] [CrossRef]
  23. Gonzalez, B.J.; Basille, M.; Vaudry, D.; Fournier, A.; Vaudry, H. Pituitary adenylate cyclase activating polypeptide promotes cell survival and neurite outgrowth in rat cerebellar neuroblasts. Neuroscience 1997, 78, 419–430. [Google Scholar] [CrossRef]
  24. Villalba, M.; Bockaert, J.; Journot, L. Pituitary adenylate cyclase-activating polypeptide (PACAP-38) protects cerebellar granule neurons from apoptosis by activating the mitogen-activated protein kinase (MAP kinase) pathway. J. Neurosci. 1997, 17, 83–90. [Google Scholar] [CrossRef] [Green Version]
  25. Journot, L.; Villalba, M.; Bockaert, J. PACAP-38 protects cerebellar granule cells from apoptosis. Ann. N. Y. Acad. Sci. 1998, 865, 100–110. [Google Scholar] [CrossRef]
  26. Ito, Y.; Arakawa, M.; Ishige, K.; Fukuda, H. Comparative study of survival signal withdrawal- and 4-hydroxynonenal-induced cell death in cerebellar granule cells. Neurosci. Res. 1999, 35, 321–327. [Google Scholar] [CrossRef]
  27. Vaudry, D.; Falluel-Morel, A.; Basille, M.; Pamantung, T.F.; Fontaine, M.; Fournier, A.; Vaudry, H.; Gonzalez, B.J. Pituitary adenylate cyclase-activating polypeptide prevents C2-ceramide-induced apoptosis of cerebellar granule cells. J. Neurosci. Res. 2003, 72, 303–316. [Google Scholar] [CrossRef]
  28. Falluel-Morel, A.; Aubert, N.; Vaudry, D.; Basille, M.; Fontaine, M.; Fournier, A.; Vaudry, H.; Gonzalez, B.J. Opposite regulation of the mitochondrial apoptotic pathway by C2-ceramide and PACAP through a MAP-kinase-dependent mechanism in cerebellar granule cells. J. Neurochem. 2004, 91, 1231–1243. [Google Scholar] [CrossRef]
  29. Vaudry, D.; Gonzalez, B.J.; Basille, M.; Pamantung, T.F.; Fontaine, M.; Fournier, A.; Vaudry, H. The neuroprotective effect of pituitary adenylate cyclase-activating polypeptide on cerebellar granule cells is mediated through inhibition of the CED3-related cysteine protease caspase-3/CPP32. Proc. Natl. Acad. Sci. USA 2000, 97, 13390–13395. [Google Scholar] [CrossRef] [Green Version]
  30. Vaudry, D.; Pamantung, T.F.; Basille, M.; Rousselle, C.; Fournier, A.; Vaudry, H.; Beauvillain, J.C.; Gonzalez, B.J. PACAP protects cerebellar granule neurons against oxidative stress-induced apoptosis. Eur. J. Neurosci. 2002, 15, 1451–1460. [Google Scholar] [CrossRef] [Green Version]
  31. Vaudry, D.; Basille, M.; Anouar, Y.; Fournier, A.; Vaudry, H.; Gonzalez, B.J. The neurotrophic activity of PACAP on rat cerebellar granule cells is associated with activation of the protein kinase A pathway and c-fos gene expression. Ann. N. Y. Acad. Sci. 1998, 865, 92–99. [Google Scholar] [CrossRef] [PubMed]
  32. Vaudry, D.; Cottet-Rousselle, C.; Basille, M.; Falluel-Morel, A.; Fournier, A.; Vaudry, H.; Gonzalez, B.J. Pituitary adenylate cyclase-activating polypeptide inhibits caspase-3 activity but does not protect cerebellar granule neurons against beta-amyloid (25-35)-induced apoptosis. Regul. Pept. 2004, 123, 43–49. [Google Scholar] [CrossRef] [PubMed]
  33. Brouwers, E.M.; Huitema, A.D.; Boogerd, W.; Beijnen, J.H.; Schellens, J.H. Persistent neuropathy after treatment with cisplatin and oxaliplatin. Acta Oncol. 2009, 48, 832–841. [Google Scholar] [CrossRef] [PubMed]
  34. Aubert, N.; Vaudry, D.; Falluel-Morel, A.; Desfeux, A.; Fisch, C.; Ancian, P.; de Jouffrey, S.; Le Bigot, J.F.; Couvineau, A.; Laburthe, M.; et al. PACAP prevents toxicity induced by cisplatin in rat and primate neurons but not in proliferating ovary cells: Involvement of the mitochondrial apoptotic pathway. Neurobiol. Dis. 2008, 32, 66–80. [Google Scholar] [CrossRef] [PubMed]
  35. Tabuchi, A.; Koizumi, M.; Nakatsubo, J.; Yaguchi, T.; Tsuda, M. Involvement of endogenous PACAP expression in the activity dependent survival of mouse cerebellar granule cells. Neurosci. Res. 2001, 39, 85–93. [Google Scholar] [CrossRef]
  36. Vaudry, D.; Hamelink, C.; Damadzic, R.; Eskay, R.L.; Gonzalez, B.; Eidena, L.E. Endogenous PACAP acts as a stress response peptide to protect cerebellar neurons from ethanol or oxidative insult. Peptides 2005, 26, 2518–2524. [Google Scholar] [CrossRef] [Green Version]
  37. Morio, H.; Tatsuno, I.; Tanaka, T.; Uchida, D.; Hirai, A.; Tamura, Y.; Saito, Y. Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neurotrophic factor for cultured rat cortical neurons. Ann. N. Y. Acad. Sci. 1996, 805, 476–481. [Google Scholar] [CrossRef]
  38. Morio, H.; Tatsuno, I.; Hirai, A.; Tamura, Y.; Saito, Y. Pituitary adenylate cyclase-activating polypeptide protects rat-cultured cortical neurons from glutamate-induced cytotoxicity. Brain Res. 1996, 741, 82–88. [Google Scholar] [CrossRef]
  39. Shintani, N.; Suetake, S.; Hashimoto, H.; Koga, K.; Kasai, A.; Kawaguchi, C.; Morita, Y.; Hirose, M.; Sakai, Y.; Tomimoto, S.; et al. Neuroprotective action of endogenous PACAP in cultured rat cortical neurons. Regul. Pept. 2005, 126, 123–128. [Google Scholar] [CrossRef]
  40. Frechilla, D.; García-Osta, A.; Palacios, S.; Cenarruzabeitia, E.; Del Rio, J. BDNF mediates the neuroprotective effect of PACAP-38 on rat cortical neurons. Neuroreport 2001, 12, 919–923. [Google Scholar] [CrossRef]
  41. Skoglösa, Y.; Lewén, A.; Takei, N.; Hillered, L.; Lindholm, D. Regulation of pituitary adenylate cyclase activating polypeptide and its receptor type 1 after traumatic brain injury: Comparison with brain-derived neurotrophic factor and the induction of neuronal cell death. Neuroscience 1999, 90, 235–247. [Google Scholar] [CrossRef]
  42. Rozzi, S.R.; Borelli, G.; Ryan, K.; Steiner, J.P.; Reglodi, D.; Mocchetti, I.; Avdoshina, V. PACAP27 is protective against tat-induced neurotoxicity. J. Mol. Neurosci. 2014, 54, 485–493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Kaneko, Y.; Tuazon, J.P.; Ji, X.; Borlongan, C.V. Pituitary adenylate cyclase activating polypeptide elicits neuroprotection against acute ischemic neuronal cell death associated with NMDA receptors. Cell. Physiol. Biochem. 2018, 51, 1982–1995. [Google Scholar] [CrossRef]
  44. Sanchez, A.; Internati, M.C.; Grammas, P. Transduction of PACAP38 protects primary cortical neurons from neurotoxic injury. Neurosci. Lett. 2008, 448, 52–55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Sanchez, A.; Rao, H.V.; Grammas, P. PACAP38 protects rat cortical neurons against the neurotoxicity evoked by sodium nitroprusside and thrombin. Regul. Pept. 2009, 152, 33–40. [Google Scholar] [CrossRef] [Green Version]
  46. Masmoudi-Kouki, O.; Douiri, S.; Hamdi, Y.; Kaddour, H.; Bahdoudi, S.; Vaudry, D.; Basille, M.; Leprince, J.; Fournier, A.; Vaudry, H.; et al. Pituitary adenylate cyclase-activating polypeptide protects astroglial cellsagainst oxidative stress-induced apoptosis. J. Neurochem. 2011, 117, 403–411. [Google Scholar] [CrossRef]
  47. Wilhelm, I.; Fazakas, C.; Tamás, A.; Tóth, G.; Reglődi, D.; Krizbai, I.A. PACAP enhances barrier properties of cerebral microvessels. J. Mol. Neurosci. 2014, 54, 469–476. [Google Scholar] [CrossRef] [Green Version]
  48. Yang, S.; Yang, J.; Yang, Z.; Chen, P.; Fraser, A.; Zhang, W.; Pang, H.; Gao, X.; Wilson, B.; Hong, J.S.; et al. Pituitary adenylate cyclaseactivating polypeptide (PACAP) 38 and PACAP4-6 are neuroprotective through inhibition of NADPH oxidase: Potent regulators of microglia-mediated oxidative stress. J. Pharmacol. Exp. Ther. 2006, 319, 595–603. [Google Scholar] [CrossRef] [Green Version]
  49. Kong, L.Y.; Maderdrut, J.L.; Jeohn, G.H.; Hong, J.S. Reduction of lipopolysaccharide-induced neurotoxicity in mixed cortical neuron/glia cultures by femtomolar concentrations of pituitary adenylate cyclase-activating polypeptide. Neuroscience 1999, 91, 493–500. [Google Scholar] [CrossRef]
  50. Takei, N.; Skoglösa, Y.; Lindholm, D. Neurotrophic and neuroprotective effects of pituitary adenylate cyclase-activating polypeptide (PACAP) on mesencephalic dopaminergic neurons. J. Neurosci. Res. 1998, 54, 698–706. [Google Scholar] [CrossRef]
  51. Manavalan, S.; Getachew, B.; Manaye, K.F.; Khundmiri, S.J.; Csoka, A.B.; McKinley, R.; Tamas, A.; Reglodi, D.; Tizabi, Y. PACAP protects against ethanol and nicotine toxicity in SH-SY5Y cells: Implications for drinking-smoking co-morbidity. Neurotox. Res. 2017, 32, 8–13. [Google Scholar] [CrossRef] [PubMed]
  52. Brown, D.; Tamas, A.; Reglodi, D.; Tizabi, Y. PACAP protects against salsolinol-induced toxicity in dopaminergic SH-SY5Y cells: Implication for Parkinson’s disease. J. Mol. Neurosci. 2013, 50, 600–607. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Brown, D.; Tamas, A.; Reglodi, D.; Tizabi, Y. PACAP protects against inflammatory-mediated toxicity in dopaminergic SH-SY5Y cells: Implication for Parkinson’s disease. Neurotox. Res. 2014, 26, 230–239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Mansouri, S.; Lietzau, G.; Lundberg, M.; Nathanson, D.; Nyström, T.; Patrone, C. Pituitary adenlylate cyclase activating peptide protects adult neural stem cells from a hypoglycaemic milieu. PLoS ONE 2016, 11, e0156867. [Google Scholar] [CrossRef]
  55. Mansouri, S.; Ortsäter, H.; Gallego, O.P.; Darsalia, V.; Sjöholm, A.; Patrone, C. Pituitary adenylate cyclase-activating polypeptide counteracts the impaired adult neural stem cell viability induced by palmitate. J. Neurosci. Res. 2012, 90, 759–768. [Google Scholar] [CrossRef]
  56. Mansouri, S.; Agartz, I.; Ögren, S.O.; Patrone, C.; Lundberg, M. PACAP protects adult neural stem cells from the neurotoxic effect of ketamine associated with decreased apoptosis, ER stress and mTOR pathway activation. PLoS ONE 2017, 12, e0170496. [Google Scholar] [CrossRef]
  57. Arimura, A.; Somogyvari-Vigh, A.; Weill, C.; Fiore, R.C.; Tatsuno, I.; Bay, V.; Brenneman, D.E. PACAP functions as a neu- rotrophic factor. Ann. N. Y. Acad. Sci. 1994, 739, 228–243. [Google Scholar] [CrossRef]
  58. Solés-Tarrés, I.; Cabezas-Llobet, N.; Lefranc, B.; Leprince, J.; Alberch, J.; Vaudry, D.; Xifró, X. Pituitary adenylate cyclase-activating polypeptide (PACAP) protects striatal cells and improves motor function in Huntington’s disease models: Role of PAC1 receptor. Front. Pharmacol. 2022, 12, 797541. [Google Scholar] [CrossRef]
  59. Broome, S.T.; Musumeci, G.; Castorina, A. PACAP and VIP mitigate rotenone-induced inflammation in BV-2 microglial cells. J. Mol. Neurosci. 2022, in press. [Google Scholar] [CrossRef]
  60. Tomimatsu, N.; Arakawa, Y. Survival-promoting activity of pituitary adenylate cyclase-activating polypeptide in the presence of phosphodiesterase inhibitors on rat motoneurons in culture: cAMP-protein kinase A-mediated survival. J. Neurochem. 2008, 107, 628–635. [Google Scholar] [CrossRef]
  61. Maugeri, G.; D’Amico, A.D.; Rasà, D.M.; Federico, C.; Saccone, S.; Morello, G.; La Cognata, V.; Cavallaro, S.; D’Agata, V. Molecular mechanisms involved in the protective effect of pituitary adenylate cyclase-activating polypeptide in an in vitro model of amyotrophic lateral sclerosis. J. Cell. Physiol. 2019, 234, 5203–5214. [Google Scholar] [CrossRef] [PubMed]
  62. D’Amico, A.G.; Maugeri, G.; Saccone, S.; Federico, C.; Cavallaro, S.; Reglodi, D.; D’Agata, V. PACAP modulates the autophagy process in an in vitro model of amyotrophic lateral sclerosis. Int. J. Mol. Sci. 2020, 21, 2943. [Google Scholar] [CrossRef] [PubMed]
  63. Li, H.X.; Feng, J.; Liu, Q.; Ou, B.Q.; Lu, S.Y.; Ma, Y. PACAP-derived mutant peptide MPAPO protects trigeminal ganglion cell and the retina from hypoxic injury through anti-oxidative stress, anti-apoptosis, and promoting axon regeneration. Biochim. Biophys. Acta Gen. Subj. 2021, 1865, 130018. [Google Scholar] [CrossRef] [PubMed]
  64. Castorina, A.; Tiralongo, A.; Giunta, S.; Carnazza, M.L.; Rasi, G.; D’Agata, V. PACAP and VIP prevent apoptosis in schwannoma cells. Brain Res. 2008, 1241, 29–35. [Google Scholar] [CrossRef] [PubMed]
  65. Onoue, S.; Ohshima, K.; Endo, K.; Yajima, T.; Kashimoto, K. PACAP protects neuronal PC12 cells from the cytotoxicity of human prion protein fragment 106–126. FEBS Lett. 2002, 522, 65–70. [Google Scholar] [CrossRef] [Green Version]
  66. Onoue, S.; Endo, K.; Ohshima, K.; Yajima, T.; Kashimoto, K. The neuropeptide PACAP attenuates beta-amyloid (1-42)-induced toxicity in PC12 cells. Peptides 2002, 23, 1471–1478. [Google Scholar] [CrossRef]
  67. Wang, G.; Qi, C.; Fan, G.H.; Zhou, H.Y.; Chen, S.D. PACAP protects neuronal differentiated PC12 cells against the neurotoxicity induced by a mitochondrial complex I inhibitor, rotenone. FEBS Lett. 2005, 579, 4005–4011. [Google Scholar] [CrossRef] [Green Version]
  68. Reglodi, D.; Fabian, Z.; Tamas, A.; Lubics, A.; Szeberenyi, J.; Alexy, T.; Toth, K.; Marton, Z.; Borsiczky, B.; Roth, E.; et al. Effects of PACAP on in vitro and in vivo neuronal cell death, platelet aggregation, and production of reactive oxygen radicals. Regul. Pept. 2004, 123, 51–59. [Google Scholar] [CrossRef]
  69. Dejda, A.; Chan, P.; Seaborn, T.; Coquet, L.; Jouenne, T.; Fournier, A.; Vaudry, H.; Vaudry, D. Involvement of stathmin 1 in the neurotrophic effects of PACAP in PC12 cells. J. Neurochem. 2010, 114, 1498–1510. [Google Scholar] [CrossRef]
  70. Chung, C.Y.; Seo, H.; Sonntag, K.C.; Brooks, A.; Lin, L.; Isacson, O. Cell type-specific gene expression of midbrain dopaminergic neurons reveals molecules involved in their vulnerability and protection. Hum. Mol. Genet. 2005, 14, 1709–1725. [Google Scholar] [CrossRef] [Green Version]
  71. Deguil, J.; Jailloux, D.; Page, G.; Fauconneau, B.; Houeto, J.L.; Philippe, M.; Muller, J.M.; Pain, S. Neuroprotective effects of pituitary adenylate cyclase-activating polypeptide (PACAP) in MPP+-induced alteration of translational control in Neuro-2a neuroblastoma cells. J. Neurosci. Res. 2007, 85, 2017–2025. [Google Scholar] [CrossRef] [PubMed]
  72. Mester, L.; Kovacs, K.; Racz, B.; Solti, I.; Atlasz, T.; Szabadfi, K.; Tamas, A.; Reglodi, D. Pituitary adenylate cyclase-activating polypeptide is protective against oxidative stress in human retinal pigment epithelial cells. J. Mol. Neurosci. 2011, 43, 35–43. [Google Scholar] [CrossRef] [PubMed]
  73. Fabian, E.; Reglodi, D.; Mester, L.; Szabo, A.; Szabadfi, K.; Tamas, A.; Toth, G.; Kovacs, K. Effects of PACAP on intracellular signaling pathways in human retinal pigment epithelial cells exposed to oxidative stress. J. Mol. Neurosci. 2012, 48, 493–500. [Google Scholar] [CrossRef] [PubMed]
  74. Maugeri, G.; D’Amico, A.G.; Gagliano, C.; Saccone, S.; Federico, C.; Cavallaro, S.; D’Agata, V. VIP family members prevent outer blood retinal barrier damage in a model of diabetic macular edema. J. Cell. Physiol. 2017, 232, 1079–1085. [Google Scholar] [CrossRef]
  75. Maugeri, G.; D’Amico, A.G.; Saccone, S.; Federico, C.; Cavallaro, S.; D’Agata, V. PACAP and VIP inhibit HIF-1α-mediated VEGF expression in a model of diabetic macular edema. J. Cell. Physiol. 2017, 232, 1209–1215. [Google Scholar] [CrossRef]
  76. Ding, Y.; Cheng, H.; Yu, R.; Tang, C.; Liu, X.; Chen, J. Effects of cyclopeptide C*HSDGIC* from the cyclization of PACAP (1-5) on the proliferation and UVB-induced apoptosis of the retinal ganglion cell line RGC-5. Peptides 2012, 36, 280–285. [Google Scholar] [CrossRef]
  77. Yu, R.; Wang, J.; Li, J.; Wang, Y.; Zhang, H.; Chen, J.; Huang, L.; Liu, X. A novel cyclopeptide from the cyclization of PACAP(1-5) with potent activity towards PAC1 attenuates STZ-induced diabetes. Peptides 2010, 31, 1062–1067. [Google Scholar] [CrossRef]
  78. Cheng, H.; Ding, Y.; Yu, R.; Chen, J.; Wu, C. Neuroprotection of a novel cyclopeptide C*HSDGIC* from the cyclization of PACAP (1-5) in cellular and rodent models of retinal ganglion cell apoptosis. PLoS ONE 2014, 9, e108090. [Google Scholar] [CrossRef]
  79. Cheng, H.; Ye, H.; Peng, R.P.; Deng, J.; Ding, Y. Inhibition of retinal ganglion cell apoptosis: Regulation of mitochondrial function by PACAP. Neural. Regen. Res. 2018, 13, 923–929. [Google Scholar] [CrossRef]
  80. Silveira, M.S.; Costa, M.R.; Bozza, M.; Linden, R. Pituitary adenylyl cyclase-activating polypeptide prevents induced cell death in retinal tissue through activation of cyclic AMP-dependent protein kinase. J. Biol. Chem. 2002, 277, 16075–16080. [Google Scholar] [CrossRef] [Green Version]
  81. Maugeri, G.; Longo, A.; D’Amico, A.G.; Rasà, D.M.; Reibaldi, M.; Russo, A.; Bonfiglio, V.; Avitabile, T.; D’Agata, V. Trophic effect of PACAP on human corneal endothelium. Peptides 2018, 99, 20–26. [Google Scholar] [CrossRef] [PubMed]
  82. Maugeri, G.; D’Amico, A.G.; Amenta, A.; Saccone, S.; Federico, C.; Reibaldi, M.; Russo, A.; Bonfiglio, V.; Avitabile, T.; Longo, A.; et al. Protective effect of PACAP against ultraviolet B radiation-induced human corneal endothelial cell injury. Neuropeptides 2020, 79, 101978. [Google Scholar] [CrossRef] [PubMed]
  83. Racz, B.; Horvath, G.; Reglodi, D.; Gasz, B.; Kiss, P.; Gallyas, F., Jr.; Sumegi, B.; Toth, G.; Nemeth, A.; Lubics, A.; et al. PACAP ameliorates oxidative stress in the chicken inner ear: An in vitro study. Regul. Pept. 2010, 160, 91–98. [Google Scholar] [CrossRef] [PubMed]
  84. Kanekar, S.; Gandham, M.; Lucero, M.T. PACAP protects against TNFα-induced cell death in olfactory epithelium and olfactory placodal cell lines. Mol. Cell. Neurosci. 2010, 45, 345–354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Szabo, D.; Sarszegi, Z.; Polgar, B.; Saghy, E.; Nemeth, A.; Reglodi, D.; Makkos, A.; Gorbe, A.; Helyes, Z.; Ferdinandy, P.; et al. PACAP-38 in acute ST-segment elevation myocardial infarction in humans and pigs: A translational study. Int. J. Mol. Sci. 2021, 22, 2883. [Google Scholar] [CrossRef]
  86. Racz, B.; Gasz, B.; Borsiczky, B.; Gallyas, F.; Tamas, A.; Jozsa, R.; Lubics, A.; Kiss, P.; Roth, E.; Ferencz, A.; et al. Protective effects of pituitary adenylate cyclase activating polypeptide in endothelial cells against oxidative stress-induced apoptosis. Gen. Comp. Endocrinol. 2007, 153, 115–123. [Google Scholar] [CrossRef]
  87. Bian, N.; Du, G.; Ip, M.F.; Ding, J.; Chang, Q.; Li, Z. Pituitary adenylate cyclase-activating polypeptide attenuates tumor necrosis factor-α-induced apoptosis in endothelial colony-forming cells. Biomed. Rep. 2017, 7, 11–16. [Google Scholar] [CrossRef] [Green Version]
  88. Gasz, B.; Racz, B.; Roth, E.; Borsiczky, B.; Ferencz, A.; Tamas, A.; Cserepes, B.; Lubics, A.; Gallyas, F., Jr.; Toth, G.; et al. Pituitary adenylate cyclase activating polypeptide protects cardiomyocytes against oxidative stress-induced apoptosis. Peptides 2006, 27, 87–94. [Google Scholar] [CrossRef]
  89. Gasz, B.; Racz, B.; Roth, E.; Borsiczky, B.; Tamas, A.; Boronkai, A.; Gallyas, F., Jr.; Toth, G.; Reglodi, D. PACAP inhibits oxidative stress-induced activation of MAP kinase-dependent apoptotic pathway in cultured cardiomyocytes. Ann. N. Y. Acad. Sci. 2006, 1070, 293–297. [Google Scholar] [CrossRef]
  90. Racz, B.; Gasz, B.; Gallyas, F.; Kiss, P.; Tamas, A.; Szanto, Z.; Lubics, A.; Lengvari, I.; Toth, G.; Hegyi, O.; et al. PKA-Bad-14-3-3 and Akt-Bad-14-3-3 signaling pathways are involved in the protective effects of PACAP against ischemia/reperfusion-induced cardiomyocyte apoptosis. Regul. Pept. 2008, 145, 105–115. [Google Scholar] [CrossRef]
  91. Roth, E.; Weber, G.; Kiss, P.; Horvath, G.; Toth, G.; Gasz, B.; Ferencz, A.; Gallyas, F., Jr.; Reglodi, D.; Racz, B. Effects of PACAP and preconditioning against ischemia/reperfusion-induced cardiomyocyte apoptosis in vitro. Ann. N. Y. Acad. Sci. 2009, 1163, 512–516. [Google Scholar] [CrossRef] [PubMed]
  92. Li, H.; Cao, L.; Yi, P.Q.; Xu, C.; Su, J.; Chen, P.Z.; Li, M.; Chen, J.Y. Pituitary adenylate cyclase-activating polypeptide ameliorates radiation-induced cardiac injury. Am. J. Transl. Res. 2019, 11, 6585–6599. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Delgado, M.; Ganea, D. Vasoactive intestinal peptide and pituitary adenylate cyclase-activating polypeptide inhibit antigen-induced apoptosis of mature T lymphocytes by inhibiting Fas ligand expression. J. Immunol. 2000, 164, 1200–1210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Delgado, M.; Garrido, E.; Martinez, C.; Leceta, L.; Gomariz, R.P. Vasoactive intestinal peptide and pituitary adenylate cyclase-activating polypeptides (PACAP27) and PACAP38) protect CD4+CD8+ thymocytes from glucocorticoid-induced apoptosis. Blood 1996, 87, 5152–5161. [Google Scholar] [CrossRef]
  95. Onoue, S.; Endo, K.; Ohmori, Y.; Yamada, S.; Kimura, R.; Yajima, T.; Kashimoto, K. Long-acting analogue of vasoactive intestinal peptide, [R15, 20, 21, L17]-VIP-GRR (IK312532), protects rat alveolar L2 cells from the cytotoxicity of cigarette smoke. Regul. Pept. 2004, 123, 193–199. [Google Scholar] [CrossRef]
  96. Onoue, S.; Ohmori, Y.; Endo, K.; Yamada, S.; Kimura, R.; Yajima, T. Vasoactive intestinal peptide and pituitary adenylate cyclase-activating polypeptide attenuate the cigarette smoke extract-induced apoptotic death of rat alveolar L2 cells. Eur. J. Biochem. 2004, 271, 1757–1767. [Google Scholar] [CrossRef]
  97. Fujimiya, M.; Inui, A. Peptidergic regulation of gastrointestinal motility in rodents. Peptides 2000, 20, 1565–1582. [Google Scholar] [CrossRef]
  98. Al-Qudah, M.; Alkahtani, R.; Akbarali, H.I.; Murthy, K.S.; Grider, J.R. Stimulation of synthesis and release of brain-derived neurotropic factor from intestinal smooth muscle cells by substance P and pituitary adenylate cyclase-activating peptide. Neurogastroenterol. Motil. 2015, 27, 1162–1174. [Google Scholar] [CrossRef] [Green Version]
  99. Illes, A.; Opper, B.; Reglodi, D.; Kerenyi, M.; Czetany, P.; Boronkai, A.; Schafer, E.; Toth, G.; Fabian, E.; Horvath, G. Effects of pituitary adenylate cyclase activating polypeptide on small intestinal INT 407 cells. Neuropeptides 2017, 65, 106–113. [Google Scholar] [CrossRef]
  100. Le, S.V.; Yamaguchi, D.J.; McArdle, C.A.; Tachiki, K.; Pisegna, J.R.; Germano, P. PAC1 and PACAP expression, signaling, and effect on the growth of HCT8, human colonic tumor cells. Regul. Pept. 2002, 109, 115–125. [Google Scholar] [CrossRef]
  101. Ji, H.; Zhang, Y.; Shen, X.; Gao, F.; Huang, C.Y.; Abad, C.; Busuttil, R.W.; Waschek, J.A.; Kupiec-Weglinski, J.W. Neuropeptide PACAP in mouse liver ischemia and reperfusion injury: Immunomodulation by the cAMP-PKA pathway. Hepatology 2013, 57, 1225–1237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Horvath, G.; Brubel, R.; Kovacs, K.; Reglodi, D.; Opper, B.; Ferencz, A.; Szakaly, P.; Laszlo, E.; Hau, L.; Kiss, P.; et al. Effects of PACAP on oxidative stress-induced cell death in rat kidney and human hepatocyte cells. J. Mol. Neurosci. 2011, 43, 67–75. [Google Scholar] [CrossRef] [PubMed]
  103. Robertson, R.P.; Harmon, J.S. Diabetes, glucosetoxicity, and oxidative stress: A case of double jeopardyfor the pancreatic islet beta cell. Free Radic. Biol. Med. 2006, 41, 177–184. [Google Scholar] [CrossRef] [PubMed]
  104. Onoue, S.; Hanato, J.; Yamada, S. Pituitary adenylate cyclase-activating polypeptide attenuates streptozotocin-induced apoptotic death of RIN-m5F cells through regulation of Bcl-2 family protein mRNA expression. FEBS J. 2008, 275, 5542–5551. [Google Scholar] [CrossRef]
  105. Han, B.; Wu, J. DcR3 protects islet β cells from apoptosis through modulating Adcyap1 and bank1 expression. J. Immunol. 2009, 183, 8157–8166. [Google Scholar] [CrossRef] [Green Version]
  106. Tsonkova, V.G.; Sand, F.W.; Wolf, X.A.; Grunnet, L.G.; Ringgaard, A.K.; Ingvorsen, C.; Winkel, L.; Kalisz, M.; Dalgaard, K.; Bruun, C.; et al. The EndoC-βH1 cell line is a valid model of human beta cells and applicable for screenings to identify novel drug target candidates. Mol. Metab. 2018, 8, 144–157. [Google Scholar] [CrossRef]
  107. Reglodi, D.; Kiss, P.; Horvath, G.; Lubics, A.; Laszlo, E.; Tamas, A.; Racz, B.; Szakaly, P. Effects of pituitary adenylate cyclase activating polypeptide in the urinary system, with special emphasis on its protective effects in the kidney. Neuropeptides 2012, 46, 61–70. [Google Scholar] [CrossRef]
  108. Arms, L.; Vizzard, M.A. Neuropeptides in lower urinary tract function. In Urinary Tract. Handbook of Experimental Pharmacology; Andersson, K.E., Michel, M., Eds.; Springer: Berlin, Germany, 2011; pp. 395–423. [Google Scholar] [CrossRef] [Green Version]
  109. Horvath, G.; Mark, L.; Brubel, R.; Szakaly, P.; Racz, B.; Kiss, P.; Tamas, A.; Helyes, Z.; Lubics, A.; Hashimoto, H.; et al. Mice deficient in pituitary adenylate cyclase activating polypeptide display increased sensitivity to renal oxidative stress in vitro. Neurosci. Lett. 2010, 469, 70–74. [Google Scholar] [CrossRef]
  110. Horvath, G.; Racz, B.; Szakaly, P.; Kiss, P.; Laszlo, E.; Hau, L.; Tamas, A.; Helyes, Z.; Lubics, A.; Hashimoto, H.; et al. Mice deficient in neuropeptide PACAP demonstrate increased sensitivity to in vitro kidney hypoxia. Transplant. Proc. 2010, 42, 2293–2295. [Google Scholar] [CrossRef]
  111. Horvath, G.; Opper, B.; Reglodi, D. The neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) is protective in inflammation and oxidative stress-induced damage in the kidney. Int. J. Mol. Sci. 2019, 20, 4944. [Google Scholar] [CrossRef] [Green Version]
  112. Arimura, A.; Li, M.; Batuman, V. Potential protective action of pituitary adenylate cyclase-activating polypeptide (PACAP38) on in vitro and in vivo models of myeloma kidney injury. Blood 2006, 107, 661–668. [Google Scholar] [CrossRef] [PubMed]
  113. Li, M.; Cortez, S.; Nakamachi, T.; Batuman, V.; Arimura, A. Pituitary adenylate cyclase activating polypeptide is a potent inhibitor of the growth of light chain secreting human multiple myeloma cells. Cancer Res. 2006, 66, 8796–8803. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Li, M.; Maderdrut, J.L.; Lertora, J.J.L.; Batuman, V. Intravenous infusion of pituitary adenylate cyclase activating polypeptide (PACAP) in a patient with multiple myeloma and myeloma kidney: A case study. Peptides 2007, 28, 1891–1895. [Google Scholar] [CrossRef]
  115. Li, M.; Maderdrut, J.L.; Lertora, J.J.L.; Arimura, A.; Batuman, V. Renoprotection by pituitary adenylate cyclase activating polypeptide in multiple myeloma and other kidney diseases. Regul. Pept. 2008, 145, 24–32. [Google Scholar] [CrossRef] [PubMed]
  116. Li, M.; Khan, A.M.; Maderdrut, J.L.; Simon, E.E.; Batuman, V. The effect of PACAP38 on MyD88-mediated signal transduction in ischemia-/hypoxia-induced acute kidney injury. Am. J. Nephrol. 2010, 32, 522–532. [Google Scholar] [CrossRef]
  117. Eneman, B.; van den Heuvel, L.; Freson, K.; Van Geet, C.; Willemsen, B.; Dijkman, H.; Levtchenko, E. Distribution and function of PACAP and its receptors in the healthy and nephrotic kidney. Nephron 2016, 132, 301–311. [Google Scholar] [CrossRef]
  118. Horvath, G.; Reglodi, D.; Czetany, P.; Illes, A.; Reman, G.; Fekete, A.; Toth, G.; Laszlo, E.; Opper, B. Effects of pituitary adenylate cyclase activating polypeptide in human proximal tubule cells against gentamicin toxicity. Int. J. Pept. Res. Ther. 2019, 25, 257–264. [Google Scholar] [CrossRef]
  119. Li, M.; Balamuthusamy, S.; Khan, A.M.; Maderdrut, J.L.; Simon, E.E.; Batuman, V. Pituitary adenylate cyclase-activating polypeptide ameliorates cisplatin-induced acute kidney injury. Peptides 2010, 31, 592–602. [Google Scholar] [CrossRef]
  120. Li, M.; Balamuthusamy, S.; Khan, A.M.; Maderdrut, J.L.; Simon, E.E.; Batuman, V. Pituitary adenylate cyclase-activating polypeptide prevents cisplatin-induced renal failure. J. Mol. Neurosci. 2011, 43, 58–66. [Google Scholar] [CrossRef]
  121. Khan, A.M.; Li, M.; Brant, E.; Maderdrut, J.L.; Majid, D.S.A.; Simon, E.E.; Batuman, V. Renoprotection with pituitary adenylate cyclase-activating polypeptide in cyclosporine A-induced nephrotoxicity. J. Investig. Med. 2011, 59, 793–802. [Google Scholar] [CrossRef]
  122. Khan, A.M.; Maderdrut, J.L.; Li, M.; Toliver, H.L.; Coy, D.H.; Simon, E.E.; Batuman, V. Pituitary adenylate cyclase-activating polypeptide prevents contrast-induced nephropathy in a novel mouse model. Physiol. Rep. 2013, 1, e00163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Reglodi, D.; Tamas, A.; Koppan, M.; Szogyi, D.; Welke, L. Role of PACAP in female fertility and reproduction at gonadal level—Recent advances. Front. Endocrinol. 2012, 3, 155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Winters, S.J.; Moore, J.P., Jr. PACAP: A regulator of mammalian reproductive function. Mol. Cell. Endocrinol. 2020, 518, 110912. [Google Scholar] [CrossRef] [PubMed]
  125. Lacombe, A.; Lelievre, V.; Roselli, C.E.; Salameh, W.; Lue, Y.H.; Lawson, G.; Muller, J.M.; Waschek, J.A.; Vilain, E. Delayed testicular aging in pituitary adenylate cyclase-acivating peptide (PACAP) null mice. Proc. Natl. Acad. Sci. USA 2006, 103, 3793–3798. [Google Scholar] [CrossRef] [Green Version]
  126. Horvath, G.; Reglodi, D.; Brubel, R.; Halasz, M.; Barakonyi, A.; Tamas, A.; Fabian, E.; Opper, B.; Toth, G.; Cohen, M.; et al. Investigation of the possible functions of PACAP in human trophoblast cells. J. Mol. Neurosci. 2014, 54, 320–330. [Google Scholar] [CrossRef] [Green Version]
  127. Boronkai, A.; Brubel, R.; Racz, B.; Tamas, A.; Kiss, P.; Horvath, G.; Lubics, A.; Szigeti, A.; Bellyei, S.; Toth, G.; et al. Effects of pituitary adenylate cyclase activating polypeptide on the survival and signal transduction pathways in human choriocarcinoma cells. Ann. N. Y. Acad. Sci. 2009, 1163, 353–357. [Google Scholar] [CrossRef]
  128. Reglodi, D.; Borzsei, R.; Bagoly, T.; Boronkai, A.; Racz, B.; Tamas, A.; Kiss, P.; Horvath, G.; Brubel, R.; Nemeth, J.; et al. Agonistic behavior of PACAP6-38 on sensory nerve terminals and cytotrophoblast cells. J. Mol. Neurosci. 2008, 36, 270–278. [Google Scholar] [CrossRef]
  129. Brubel, R.; Boronkai, A.; Reglodi, D.; Racz, B.; Nemeth, J.; Kiss, P.; Lubics, A.; Toth, G.; Horvath, G.; Varga, T.; et al. Changes in the expression of pituitary adenylate cyclase-activating polypeptide in the human placenta during pregnancy and its effects on the survival of JAR choriocarcinoma cells. J. Mol. Neurosci. 2010, 42, 450–458. [Google Scholar] [CrossRef]
  130. Shan, W.; Lu, S.; Ou, B.; Feng, J.; Wang, Z.; Li, H.; Lu, X.; Ma, Y. PACAP ameliorates the fertility of obese mice through PAC1/PKA/ERK/Nrf2 signal axis. J. Endocrinol. 2021, 248, 337–354. [Google Scholar] [CrossRef]
  131. Gutiérrez-Cañas, I.; Rodríguez-Henche, N.; Bolaños, O.; Carmena, M.J.; Prieto, J.C.; Juarranz, M.G. VIP and PACAP are autocrine factors that protect the androgen-independent prostate cancer cell line PC-3 from apoptosis induced by serum withdrawal. Br. J. Pharmacol. 2003, 139, 1050–1058. [Google Scholar] [CrossRef] [Green Version]
  132. Zibara, K.; Zeidan, A.; Mallah, K.; Kassem, N.; Awad, A.; Mazurier, F.; Badran, B.; El-Zein, N. Signaling pathways activated by PACAP in MCF-7 breast cancer cells. Cell. Signal. 2018, 50, 37–47. [Google Scholar] [CrossRef] [PubMed]
  133. Horvath, G.; Reglodi, D.; Opper, B.; Brubel, R.; Tamas, A.; Kiss, P.; Toth, G.; Csernus, V.; Matkovits, A.; Racz, B. Effects of PACAP on the oxidative stress-induced cell death in chicken pinealocytes is influenced by the phase of the circadian clock. Neurosci. Lett. 2010, 484, 148–152. [Google Scholar] [CrossRef] [PubMed]
Table 1. Effects of exogenous and endogenous PACAP in vitro on cell death (CD) in the nervous system. In case the type of cell death was not specified in the cited study, the term “cell death” was used. * shows effects of endogenous PACAP.
Table 1. Effects of exogenous and endogenous PACAP in vitro on cell death (CD) in the nervous system. In case the type of cell death was not specified in the cited study, the term “cell death” was used. * shows effects of endogenous PACAP.
Cell TypeSpeciesStressorEffect on CDMechanismReferences
CENTRAL NERVOUS SYSTEM
Cerebellum
Cerebellar granule cellRatSerum and K+ deprivationAnti-apoptoticcAMP/PKA pathway[18,19]
[24,25,26]
Cerebellar granule cellRatK+ deprivationAnti-apoptoticcAMP/PKA pathway[20,22]
Cerebellar granule cellRatEthanolAnti-apoptoticCaspase-3↓[21]
Caspase-6↓
Cerebellar granule cellRatCeramideAnti-apoptoticCaspase-3↓[27,28]
Restoration of mitochondrial activity
Cerebellar granule cellRatH2O2-induced oxidative stressAnti-apoptoticcAMP/PKA pathway
caspase-3↓
[30]
Cerebellar granule cellRatAbeta25–35No effectcAMP/PKA pathway[32]
Caspase-3↓
Cerebellar granule cellRat4-HydroxynonenalAnti-apoptotic [26]
Cerebellar granule cellRatCisplatinAnti-apoptoticCaspase-3↓[34]
Caspase-9↓
Bax↓
Cerebellar granule cell *PACAP knockout mouseEthanolHigher sensitivity [36]
Cerebellar granule cell *PACAP knockout mouseOxidative stresshigher sensitivity [36]
Cerebrum
Cortical neuronRatGluCell death↓cAMP/PKA pathway[37,38,39]
PACAP mRNA↑
Primary culture of cerebral cortexRatNMDACell death↓Involvement of BDNF[40]
Primary culture of cerebral cortexRatSerum deprivationCell death↓ [40]
Primary culture of cerebral cortexRatIonomycinCell death↓ [41]
Primary culture of cerebral cortexRatTatCell death↓ [42]
Primary culture of cerebral cortexRatOxygen-glucose deprivation-reperfusionAnti-apoptoticCaspase-3↓[43]
Cytochrome-c↓
Primary cortical neuron cultureRatSodium nitroprussideCell death↓Bcl-2↑[44]
Caspase-3↓
Primary cortical neuron cultureRatThrombin, thrombin receptor activating peptideCell death↓Caspase-3↓[45]
SH-SY5Y neuroblastoma cellHumanHigh ethanolCell death↓ [51]
SH-SY5Y neuroblastoma cellHumanHigh nicotineCell death↓ [51]
SH-SY5Y neuroblastoma cellHumanSalsolinolCell death↓Caspase-3↓[52]
SH-SY5Y neuroblastoma cellHumanLPS + IFNγ-stimulated microglia-derived mediatorsAnti-apoptoticcaspase-3↓[53]
pCREB↑
BDNF↑
SH-SY5Y neuroblastoma cellHumanLPS-stimulated microglia-derived mediatorsAnti-apoptoticCaspase-3↓[53]
pCREB↑
BDNF↑
SH-SY5Y neuroblastoma cellHumanMPP+Autophagy↓ [17]
Cortical astrocytesRatH2O2-induced oxidative stressCell death↓Caspase-3↓[46]
hCMEC/D3 cerebral endothelial cellsHumanGlucose deprivationNo effect [47]
hCMEC/D3 cerebral endothelial cellsHumanDMNQ-induced oxidative stressNo effect [47]
Mesencephalic neuron/glia cultureRatLPSCell death↓ [48]
Mesencephalic neuron/glia cultureRatMPP+Cell death↓ [48]
Cortical neuron/glia cultureMouseLPSCell death↓ [49]
Mesencephalic neuron cultureRat6-OHDACell death↓ [50]
Neural stem cellsMouseHypoglycemiaCell death↓Caspase-3↓[54]
Bcl-2↑
Neural stem cellsMousePalmitate-induced lipotoxicityCell death↓Bcl-2↑[55]
Neural stem cellsMouseKetamineCell death↓Caspase-3↓[56]
Bcl-2↑
Neuro-2a neuroblastoma cellMouseMPP+Anti-apoptoticPhospho-eIF2α↓[71]
mTOR↑
Hippocampal cultureMouseHIV envelope protein gp120Cell death↓ [57]
STHdhQ111/Q111 striatal cellsMouseMutant huntingtin expressionAnti-apoptoticpERK↑[58]
pAkt↑
Caspase-3↓
BV-2 microgliaMouseRotenoneCell death↓ [59]
Spinal cord
Primary culture of motoneuronsRatGluCell death↓cAMP/PKA pathway[60]
NSC-34 motor neuronMouseSerum deprivationAnti-apoptoticpERK1/2↑[61]
NSC-34 motor neuronMouseDesferrioxamine mesylate saltAnti-apoptoticLC3II↓[62]
Modulation of autophagyp62↑
pERK1/2↑
PERIPHERAL NERVOUS SYSTEM
Trigeminal ganglion cellMouseHypoxiaAnti-apoptoticCaspase-3↓[63]
Cytochrome c↓
CRL-2768 schwannoma cellRatSerum deprivationAnti-apoptoticBcl-2 mRNA↓[64]
Bax mRNA↓
PC12RatPrion protein fragmentCell death↓Caspase-3↓[65]
pheochromocytoma
cell
PC12RatBeta-amyloid peptideCell death↓Caspase-3↓[66]
pheochromocytoma
cell
PC12
pheochromocytoma
cell


RatRotenoneAnti-apoptoticCaspase-3↓
PKA
ERK,
p38 MAPK
[67]
PC12
pheochromocytoma
cell
RatAnisomycinAnti-apoptoticPKA[68]
PC12RatMPP+Cell death↓ [70]
pheochromocytoma
cell
Table 2. Effects of exogenous PACAP in vitro on cell death (CD) in the sensory organs. In case the type of cell death was not specified in the cited study, the term “cell death” was used.
Table 2. Effects of exogenous PACAP in vitro on cell death (CD) in the sensory organs. In case the type of cell death was not specified in the cited study, the term “cell death” was used.
Cell TypeSpeciesStressorEffect on CDMechanismReference(s)
Eye
ARPE-19
pigment epithelial cell
HumanH2O2-induced oxidative stressAnti-apoptotic [72]
ARPE-19
pigment epithelial cell
HumanH2O2-induced oxidative stressAnti-apoptoticpAkt↑
pERK1/2↑
pp38MAPK ↓
pJNK↓
p53↓
pp53↓
Bad↓
Bax↓
FADD↓
SMAC/Diablo↓
Fas/TNFSF6↓
[73]
ARPE-19
pigment epithelial cell
HumanHyperglycemia/hypoxia insultAnti-apoptoticpAkt↑
pERK1/2↑
pp38 MAPK↓
[74,75]
RGC-5
ganglion cell
RatUV-B irradiationCell death↓ [76]
Retina explantRatAnisomycinAnti-apoptoticcAMP/PKA pathway[80]
Retina explantratThapsigarginCell death↓cAMP/PKA pathway[80]
Corneal endothelial cellHumanGrowth factor deprivationCell death↓ [81]
Corneal endothelial cellHumanUV-B irradiationAnti-apoptoticCaspase-3↓
Bax↓
Bcl-2↑
[82]
Inner Ear
Primary cochlear
cell culture
ChickenH2O2-induced oxidative stressAnti-apoptoticcaspase-3↓[83]
Table 3. Effects of exogenous and endogenous PACAP in vitro on cell death (CD) in the peripheral organs. If type of cell death was not specified in the cited study, the term “cell death” was used. * shows effects of endogenous PACAP.
Table 3. Effects of exogenous and endogenous PACAP in vitro on cell death (CD) in the peripheral organs. If type of cell death was not specified in the cited study, the term “cell death” was used. * shows effects of endogenous PACAP.
Cell TypeSpeciesStressorEffect on CDMechanismReference(s)
Cardiovascular System
EOMAMouseH2O2-induced oxidative stressAnti-apoptoticERK↑[86]
hemangioendotheliomap38 MAPK↓
JNK↓
Endothelial colony-forming cellsHumanTNF-αAnti-apoptotic [87]
Primary cardiomyocyte cultureRatH2O2-induced oxidative stressAnti-apoptoticCaspase-3↓[88,89]
Bcl-2↑
Bad↑
ASK-1↓
Primary cardiomyocyte cultureRatSimulated ischemia/reperfusionAnti-apoptoticPhospho-PKA↑[90,91]
Phospho-Akt↑
Phospho-Bad↑
14-3-3↑
Bcl-xL↑
H9C2 cardiomyoblastRatIrradiationAnti-apoptoticBcl-2↑[92]
Bax↓
Immune System
T cellMouseAnti-CD3Anti-apoptoticFasL↓[93]
Respiratory System
L2 alveolar cellRatCigarette smoke extractCell death ↓Caspase-3↓[95,96]
Gastrointestinal Tract
INT 407 jejunal and ileal cellHumanH2O2-induced oxidative stressCell death ↓ [99]
INT 407 jejunal and ileal cellHumanCoCl2-induced in vitro hypoxiaNo effect [99]
INT 407 jejunal and ileal cellHumangamma radiationNo effect [99]
INT 407 jejunal and ileal cell *HumanH2O2-inducedHigher vulnerability [99]
oxidative stress
Primary mouse hepatocyte cultureMouseH2O2Anti-apoptotic [101]
Primary mouse hepatocyte cultureMouseTNF-αAnti-apoptoticCaspase-3↓[101]
WRL-68 hepatocyte H2O2No effect [102]
Hep-G2 hepatocellular carcinoma cell H2O2No effect [102]
RIN-m5F pancreatic cellRatStreptozotocinCell death ↓Bcl-2 mRNA↑[104]
Noxa mRNA↓
Bax mrNA↓
NIT-1 insulinoma cell *MouseMixture of cytokines (IL-1β, IFNγ)Cell survival↑ [105]
EndoC-βH1 pancreatic cellHumanMixture of cytokines (IL-1β, IFNγ, TNFα)No effect [106]
Urinary Tract
Primary renal cell cultureRatH2O2-induced oxidative stressCell death ↓ [102]
Primary renal cell cultureMouseH2O2-induced oxidative stressCell death ↓ [109]
Primary renal cell culture *MouseH2O2-induced oxidative stressHigher vulnerability [109]
SV 40 proximal tubule epithelial cellHumanMyeloma κ light chainCell death ↓ [112]
proximal tubule epithelial cellMouseMineral oil induced in vitro hypoxiaAnti-apoptotic [116]
HK-2 proximal tubule cellHumanAlbuminNo effect [117]
HK-2 proximal tubule cellHumanGentamicinCell death ↓ [118]
HK-2 proximal tubule cellHumanCisplatinAnti-apoptoticDNA fragmentation↓[119]
p53↓
Caspase-7↑, PARP-1↑
APE-1↑
Bcl-2↑,
Bcl-xL↑
Bax↓
Proximal tubule epithelial cellMouseCisplatinAnti-apoptotic [120]
HK-2 proximal tubule cellHumanCyclosporin AAnti-apoptotic [121]
HK-2 proximal tubule cellHumanContrast mediumAnti-apoptotic [122]
Reproductive System
HIPEC65 trophoblastHumanMethothrexateNo effect [126]
HTR-8/Svneo trophoblastHumanH2O2-induced oxidative stressCell death↓ [126]
JAR choriocarcinoma cellHumanH2O2-induced oxidative stressCell death↑p-AKT↓[127]
p-ERK-1/2↓
p-p38MAPK↓
p-JNK/SAPK↓
Bax↓
JAR choriocarcinoma cellHumanCoCl2-induced in vitro hypoxiaCell death↑ [127]
JAR choriocarcinoma cellHumanMethothrexateNo effect [129]
CHO ovaryHamsterCisplatinNo effect [34]
GC-2 spermatocyteMousePalmitateAnti-apoptoticCaspase-3↓[130]
Bax↓
Bcl-2↑
PC-3 prostateHumanSerum deprivationCell death↓Bcl-2↑[131]
Procaspase-3↑
Glands
MCF-7 breast adenocarcinomaHuman-Pro-apoptoticBax↑[132]
Bcl-2↓
Salivary gland extractSnailDopamineAnti-apoptoticCaspase-3↓[13]
Salivary gland extractSnailColchicineAnti-apoptotic [13]
PinealocyteChickenH2O2-induced oxidative stressAnti-apoptotic in the dark phase, [133]
No effect in the light phase
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Horvath, G.; Reglodi, D.; Fabian, E.; Opper, B. Effects of Pituitary Adenylate Cyclase Activating Polypeptide on Cell Death. Int. J. Mol. Sci. 2022, 23, 4953. https://doi.org/10.3390/ijms23094953

AMA Style

Horvath G, Reglodi D, Fabian E, Opper B. Effects of Pituitary Adenylate Cyclase Activating Polypeptide on Cell Death. International Journal of Molecular Sciences. 2022; 23(9):4953. https://doi.org/10.3390/ijms23094953

Chicago/Turabian Style

Horvath, Gabriella, Dora Reglodi, Eszter Fabian, and Balazs Opper. 2022. "Effects of Pituitary Adenylate Cyclase Activating Polypeptide on Cell Death" International Journal of Molecular Sciences 23, no. 9: 4953. https://doi.org/10.3390/ijms23094953

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