1
|
Horvath G, Reglodi D, Fabian E, Opper B. Effects of Pituitary Adenylate Cyclase Activating Polypeptide on Cell Death. Int J Mol Sci 2022; 23:ijms23094953. [PMID: 35563353 PMCID: PMC9100246 DOI: 10.3390/ijms23094953] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/25/2022] [Accepted: 04/28/2022] [Indexed: 12/18/2022] Open
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.
Collapse
|
2
|
Guo X, Tian Y, Yang Y, Li S, Guo L, Shi J. Pituitary Adenylate Cyclase-Activating Polypeptide Protects Against Cognitive Impairment Caused by Chronic Cerebral Hypoperfusion. Mol Neurobiol 2021; 58:4309-4322. [PMID: 33999349 DOI: 10.1007/s12035-021-02381-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/31/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Pituitary adenylate cyclase-activating polypeptide (PACAP) has beneficial effects in learning and memory. However, the mechanism by which PACAP improves cognitive impairment of vascular dementia (VaD) is not clear. METHODS We established a VaD model by bilateral common carotid stenosis (BCAS) to investigate the molecular mechanism of cognitive impairment. Protein levels of PACAP, Sirtuin 3 (Sirt3), brain-derived neurotrophic factor (BDNF), and postsynaptic density 95 (PSD-95) were assessed by Western blot. In vitro, oxygen glucose deprivation (OGD) was used to simulate the ischemia/hypoxia state. HT22 cells were transfected with Sirt3 knockdown and overexpression to study the relationship between PACAP, Sirt3, and BDNF. In vivo, PACAP was administered intranasally to assess its protective effects on BCAS. RESULTS The study showed that the levels of PACAP, Sirt3, BDNF, and PSD-95 were decreased in the BCAS model of VaD. PACAP increased the protein levels of Sirt3, BDNF, PSD-95, Bcl-2, and Bax under OGD condition in vitro. Sirt3 regulated BDNF and synaptic plasticity. Intranasal PACAP increased the protein levels of PAC1, Sirt3, BDNF, and PSD-95 in vivo. CONCLUSIONS This study provides evidence that PACAP regulates synaptic plasticity and plays an antiapoptotic role through Sirt3.
Collapse
Affiliation(s)
- Xiaosu Guo
- The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ye Tian
- The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yaping Yang
- The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shiping Li
- The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Li Guo
- The Second Hospital of Hebei Medical University, Shijiazhuang, China.
| | - Jiong Shi
- National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China. .,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China. .,Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
3
|
Fang Y, Ren R, Shi H, Huang L, Lenahan C, Lu Q, Tang L, Huang Y, Tang J, Zhang J, Zhang JH. Pituitary Adenylate Cyclase-Activating Polypeptide: A Promising Neuroprotective Peptide in Stroke. Aging Dis 2020; 11:1496-1512. [PMID: 33269103 PMCID: PMC7673855 DOI: 10.14336/ad.2020.0626] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 06/26/2020] [Indexed: 12/11/2022] Open
Abstract
The search for viable, effective treatments for acute stroke continues to be a global priority due to the high mortality and morbidity. Current therapeutic treatments have limited effects, making the search for new treatments imperative. Pituitary adenylate cyclase-activating polypeptide (PACAP) is a well-established cytoprotective neuropeptide that participates in diverse neural physiological and pathological activities, such as neuronal proliferation, differentiation, and migration, as well as neuroprotection. It is considered a promising treatment in numerous neurological diseases. Thus, PACAP bears potential as a new therapeutic strategy for stroke treatment. Herein, we provide an overview pertaining to the current knowledge of PACAP, its receptors, and its potential neuroprotective role in the setting of stroke, as well as various mechanisms of neuroprotection involving ionic homeostasis, excitotoxicity, cell edema, oxidative stress, inflammation, and cell death, as well as the route of PACAP administration.
Collapse
Affiliation(s)
- Yuanjian Fang
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Reng Ren
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hui Shi
- 2Department of Neurosurgery, Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Lei Huang
- 3Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA.,4Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Cameron Lenahan
- 3Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA.,4Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA.,5Burrell College of Osteopathic Medicine, Las Cruces, NM, USA
| | - Qin Lu
- 6Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University, School of Medicine, Hangzhou, Zhejiang, China
| | - Lihui Tang
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yi Huang
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jiping Tang
- 3Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA.,4Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA.,7Department of Anesthesiology, Loma Linda University, Loma Linda, CA, USA
| | - Jianmin Zhang
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - John H Zhang
- 3Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA.,4Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA.,7Department of Anesthesiology, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
4
|
Abstract
Dysregulation of neuropeptides may play an important role in aging-induced impairments. In the long list of neuropeptides, pituitary adenylate cyclase-activating polypeptide (PACAP) represents a highly effective cytoprotective peptide that provides an endogenous control against a variety of tissue-damaging stimuli. PACAP has neuro- and general cytoprotective effects due to anti-apoptotic, anti-inflammatory, and antioxidant actions. As PACAP is also a part of the endogenous protective machinery, it can be hypothesized that the decreased protective effects in lack of endogenous PACAP would accelerate age-related degeneration and PACAP knockout mice would display age-related degenerative signs earlier. Recent results support this hypothesis showing that PACAP deficiency mimics aspects of age-related pathophysiological changes including increased neuronal vulnerability and systemic degeneration accompanied by increased apoptosis, oxidative stress, and inflammation. Decrease in PACAP expression has been shown in different species from invertebrates to humans. PACAP-deficient mice display numerous pathological alterations mimicking early aging, such as retinal changes, corneal keratinization and blurring, and systemic amyloidosis. In the present review, we summarize these findings and propose that PACAP deficiency could be a good model of premature aging.
Collapse
|
5
|
Solymar M, Ivic I, Balasko M, Fulop BD, Toth G, Tamas A, Reman G, Koller A, Reglodi D. Pituitary adenylate cyclase-activating polypeptide ameliorates vascular dysfunction induced by hyperglycaemia. Diab Vasc Dis Res 2018; 15:277-285. [PMID: 29466879 DOI: 10.1177/1479164118757922] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Short-lasting hyperglycaemia occurs frequently in prediabetes and poorly controlled diabetes mellitus leading to vascular damage. Pituitary adenylate cyclase-activating polypeptide (PACAP) has been shown to play a protective role in vascular complications of diabetes; moreover, antioxidant effects of PACAP were also described. Therefore, we hypothesized that PACAP exerts protective effects in short-term hyperglycaemia-induced vascular dysfunctions. METHODS After short-term hyperglycaemia, acetylcholine-induced and sodium nitroprusside-induced vascular relaxation of mouse carotid arteries were tested with a myograph with or without the presence of PACAP or superoxide dismutase. Potential direct antioxidant superoxide-scavenging action of pituitary adenylate cyclase-activating peptide was tested with pyrogallol autoxidation assay; furthermore, the effect of pituitary adenylate cyclase-activating peptide or superoxide dismutase was investigated on hyperglycaemia-associated vascular markers. RESULTS PACAP administration resulted in reduced endothelial dysfunction after a 1-h hyperglycaemic episode. PACAP was able to restore acetylcholine-induced relaxation of the vessels and improved sodium nitroprusside-induced relaxation. This effect was comparable to the protective effect of superoxide dismutase, but PACAP was unable to directly scavenge superoxide produced by autoxidation of pyrogallol. Endothelial dysfunction was associated with elevated levels of fibroblast growth factor basic, matrix metalloproteinase 9 and nephroblastoma overexpressed gene proteins. Their release was reduced by PACAP administration. CONCLUSION These results suggest a strong protective role of PACAP in the vascular complications of diabetes.
Collapse
Affiliation(s)
- Margit Solymar
- 1 Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Ivan Ivic
- 2 Department of Anatomy, MTA-PTE PACAP Research Team, Centre for Neuroscience, Medical School, University of Pécs, Pécs, Hungary
| | - Marta Balasko
- 1 Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Balazs D Fulop
- 2 Department of Anatomy, MTA-PTE PACAP Research Team, Centre for Neuroscience, Medical School, University of Pécs, Pécs, Hungary
| | - Gabor Toth
- 3 Department of Medical Chemistry, University of Szeged, Szeged, Hungary
| | - Andrea Tamas
- 2 Department of Anatomy, MTA-PTE PACAP Research Team, Centre for Neuroscience, Medical School, University of Pécs, Pécs, Hungary
| | - Gyongyver Reman
- 2 Department of Anatomy, MTA-PTE PACAP Research Team, Centre for Neuroscience, Medical School, University of Pécs, Pécs, Hungary
| | - Akos Koller
- 4 Department of Physiology, New York Medical College, Valhalla, NY, USA
- 5 Department of Neurosurgery, Medical School, University of Pécs, Pécs, Hungary
- 6 Institute of Natural Sciences, University of Physical Education, Budapest, Hungary
| | - Dora Reglodi
- 2 Department of Anatomy, MTA-PTE PACAP Research Team, Centre for Neuroscience, Medical School, University of Pécs, Pécs, Hungary
| |
Collapse
|
6
|
Protective effects of pituitary adenylate cyclase activating polypeptide against neurotoxic agents. Neurotoxicology 2018; 66:185-194. [DOI: 10.1016/j.neuro.2018.03.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 03/09/2018] [Accepted: 03/27/2018] [Indexed: 01/28/2023]
|
7
|
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) is an ubiquitous peptide involved, among others, in neurodevelopment, neuromodulation, neuroprotection, neurogenic inflammation and nociception. Presence of PACAP and its specific receptor, PAC1, in the trigeminocervical complex, changes of PACAP levels in migraine patients and the migraine-inducing effect of PACAP injection strongly support the involvement of PACAP/PAC1 receptor in migraine pathogenesis. While antagonizing PAC1 receptor is a promising therapeutic target in migraine, the diverse array of PACAP's functions, including protection in ischemic events, requires that the cost-benefit of such an intervention is well investigated by taking all the beneficial effects of PACAP into account. In the present review we summarize the protective effects of PACAP in ischemia, especially in neuronal ischemic injuries, and discuss possible points to consider when developing strategies in migraine therapy interfering with the PACAP/PAC1 receptor system.
Collapse
|
8
|
Huang J, Waters K, Machaalani R. Hypoxia and nicotine effects on Pituitary adenylate cyclase activating polypeptide (PACAP) and its receptor 1 (PAC1) in the developing piglet brainstem. Neurotoxicology 2017; 62:30-38. [DOI: 10.1016/j.neuro.2017.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 05/11/2017] [Accepted: 05/11/2017] [Indexed: 11/27/2022]
|
9
|
Lamine A, Létourneau M, Doan ND, Maucotel J, Couvineau A, Vaudry H, Chatenet D, Vaudry D, Fournier A. Characterizations of a synthetic pituitary adenylate cyclase-activating polypeptide analog displaying potent neuroprotective activity and reduced in vivo cardiovascular side effects in a Parkinson's disease model. Neuropharmacology 2016; 108:440-50. [PMID: 26006268 DOI: 10.1016/j.neuropharm.2015.05.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 05/07/2015] [Accepted: 05/12/2015] [Indexed: 02/08/2023]
Abstract
Parkinson's disease (PD) is characterized by a steady loss of dopamine neurons through apoptotic, inflammatory and oxidative stress processes. In that line of view, the pituitary adenylate cyclase-activating polypeptide (PACAP), with its ability to cross the blood-brain barrier and its anti-apoptotic, anti-inflammatory and anti-oxidative properties, has proven to offer potent neuroprotection in various PD models. Nonetheless, its peripheral actions, paired with low metabolic stability, hampered its clinical use. We have developed Ac-[Phe(pI)(6), Nle(17)]PACAP(1-27) as an improved PACAP-derived neuroprotective compound. In vitro, this analog stimulated cAMP production, maintained mitochondrial potential and protected SH-SY5Y neuroblastoma cells from 1-methyl-4-phenylpyridinium (MPP(+)) toxicity, as potently as PACAP. Furthermore, contrasting with PACAP, it is stable in human plasma and against dipeptidyl peptidase IV activity. When injected intravenously to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice, PACAP and Ac-[Phe(pI)(6), Nle(17)]PACAP(1-27) restored tyrosine hydoxylase expression into the substantia nigra and modulated the inflammatory response. Albeit falls of mean arterial pressure (MAP) were observed with both PACAP- and Ac-[Phe(pI)(6), Nle(17)]PACAP(1-27)-treated mice, the intensity of the decrease as well as its duration were significantly less marked after iv injections of the analog than after those of the native polypeptide. Moreover, no significant changes in heart rate were measured with the animals for both compounds. Thus, Ac-[Phe(pI)(6), Nle(17)]PACAP(1-27) appears as a promising lead molecule for the development of PACAP-derived drugs potentially useful for the treatment of PD or other neurodegenerative diseases.
Collapse
Affiliation(s)
- Asma Lamine
- INRS - Institut Armand-Frappier, 531 boul. des Prairies, Laval, QC H7V 1B7, Canada; Laboratoire International Associé Samuel de Champlain, Université de Rouen, France; INSERM-U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, IRIB, Université de Rouen, 76821 Mont-Saint-Aignan, France
| | - Myriam Létourneau
- INRS - Institut Armand-Frappier, 531 boul. des Prairies, Laval, QC H7V 1B7, Canada; Laboratoire International Associé Samuel de Champlain, Université de Rouen, France
| | - Ngoc Duc Doan
- INRS - Institut Armand-Frappier, 531 boul. des Prairies, Laval, QC H7V 1B7, Canada; Laboratoire International Associé Samuel de Champlain, Université de Rouen, France
| | - Julie Maucotel
- Laboratoire International Associé Samuel de Champlain, Université de Rouen, France; INSERM-U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, IRIB, Université de Rouen, 76821 Mont-Saint-Aignan, France
| | - Alain Couvineau
- INSERM U1149/Inflammation Research Center (CRI), Université Paris-Diderot, Faculté de Médecine Site Bichat, 16, rue H. Huchard, 75018 Paris, France
| | - Hubert Vaudry
- Laboratoire International Associé Samuel de Champlain, Université de Rouen, France; INSERM-U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, IRIB, Université de Rouen, 76821 Mont-Saint-Aignan, France
| | - David Chatenet
- INRS - Institut Armand-Frappier, 531 boul. des Prairies, Laval, QC H7V 1B7, Canada
| | - David Vaudry
- Laboratoire International Associé Samuel de Champlain, Université de Rouen, France; INSERM-U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, IRIB, Université de Rouen, 76821 Mont-Saint-Aignan, France
| | - Alain Fournier
- INRS - Institut Armand-Frappier, 531 boul. des Prairies, Laval, QC H7V 1B7, Canada; Laboratoire International Associé Samuel de Champlain, Université de Rouen, France.
| |
Collapse
|
10
|
Seaborn T, Ravni A, Au R, Chow BKC, Fournier A, Wurtz O, Vaudry H, Eiden LE, Vaudry D. Induction of serpinb1a by PACAP or NGF is required for PC12 cells survival after serum withdrawal. J Neurochem 2014; 131:21-32. [PMID: 24899316 DOI: 10.1111/jnc.12780] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 04/30/2014] [Accepted: 05/13/2014] [Indexed: 01/11/2023]
Abstract
PC12 cells are used to study the signaling mechanisms underlying the neurotrophic and neuroprotective activities of pituitary adenylate cyclase-activating polypeptide (PACAP) and nerve growth factor (NGF). Previous microarray experiments indicated that serpinb1a was the most induced gene after 6 h of treatment with PACAP or NGF. This study confirmed that serpinb1a is strongly activated by PACAP and NGF in a time-dependent manner with a maximum induction (~ 50-fold over control) observed after 6 h of treatment. Co-incubation with PACAP and NGF resulted in a synergistic up-regulation of serpinb1a expression (200-fold over control), suggesting that PACAP and NGF act through complementary mechanisms. Consistently, PACAP-induced serpinb1a expression was not blocked by TrkA receptor inhibition. Nevertheless, the stimulation of serpinb1a expression by PACAP and NGF was significantly reduced in the presence of extracellular signal-regulated kinase, calcineurin, protein kinase A, p38, and PI3K inhibitors, indicating that the two trophic factors share some common pathways in the regulation of serpinb1a. Finally, functional investigations conducted with siRNA revealed that serpinb1a is not involved in the effects of PACAP and NGF on PC12 cell neuritogenesis, proliferation or body cell volume but mediates their ability to block caspases 3/7 activity and to promote PC12 cell survival.
Collapse
Affiliation(s)
- Tommy Seaborn
- Neurotrophic Factor and Neuronal Differentiation Team, Inserm U982, DC2N, Mont-Saint-Aignan, France; International Associated Laboratory Samuel de Champlain, Mont-Saint-Aignan, France; Department of Pediatrics, Hôpital St-François d'Assise, Centre de Recherche du Centre Hospitalier Universitaire de Québec (CRCHUQ), Laval University, Québec, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Greenberg A, Ward-Flanagan R, Dickson CT, Treit D. ANI inactivation: Unconditioned anxiolytic effects of anisomycin in the ventral hippocampus. Hippocampus 2014; 24:1308-16. [DOI: 10.1002/hipo.22312] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/29/2014] [Indexed: 12/22/2022]
Affiliation(s)
| | | | - Clayton T. Dickson
- Centre for Neuroscience; University of Alberta; Edmonton Alberta
- Department of Psychology; University of Alberta; Edmonton Alberta
- Department of Physiology; University of Alberta; Edmonton Alberta
| | - Dallas Treit
- Centre for Neuroscience; University of Alberta; Edmonton Alberta
- Department of Psychology; University of Alberta; Edmonton Alberta
| |
Collapse
|
12
|
Lazarovici P, Cohen G, Arien-Zakay H, Chen J, Zhang C, Chopp M, Jiang H. Multimodal neuroprotection induced by PACAP38 in oxygen-glucose deprivation and middle cerebral artery occlusion stroke models. J Mol Neurosci 2012; 48:526-40. [PMID: 22678884 DOI: 10.1007/s12031-012-9818-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Accepted: 05/21/2012] [Indexed: 01/30/2023]
Abstract
Pituitary adenylate cyclase activating peptide (PACAP), a potent neuropeptide which crosses the blood-brain barrier, is known to provide neuroprotection in rat stroke models of middle cerebral artery occlusion (MCAO) by mechanism(s) which deserve clarification. We confirmed that following i.v. injection of 30 ng/kg of PACAP38 in rats exposed to 2 h of MCAO focal cerebral ischemia and 48 h reoxygenation, 50 % neuroprotection was measured by reduced caspase-3 activity and volume of cerebral infarction. Similar neuroprotective effects were measured upon PACAP38 treatment of oxygen-glucose deprivation and reoxygenation of brain cortical neurons. The neuroprotection was temporally associated with increased expression of brain-derived neurotrophic factor, phosphorylation of its receptor-tropomyosin-related kinase receptor type B (trkB), activation of phosphoinositide 3-kinase and Akt, and reduction of extracellular signal-regulated kinases 1/2 phosphorylation. PACAP38 increased expression of neuronal markers beta-tubulin III, microtubule-associated protein-2, and growth-associated protein-43. PACAP38 induced stimulation of Rac and suppression of Rho GTPase activities. PACAP38 downregulated the nerve growth factor receptor (p75(NTR)) and associated Nogo-(Neurite outgrowth-A) receptor. Collectively, these in vitro and in vivo results propose that PACAP exhibits neuroprotective effects in cerebral ischemia by three mechanisms: a direct one, mediated by PACAP receptors, and two indirect, induced by neurotrophin release, activation of the trkB receptors and attenuation of neuronal growth inhibitory signaling molecules p75(NTR) and Nogo receptor.
Collapse
Affiliation(s)
- Philip Lazarovici
- School of Pharmacy Institute for Drug Research, The Hebrew University of Jerusalem, POB 12065, Jerusalem 91120, Israel.
| | | | | | | | | | | | | |
Collapse
|
13
|
Szabadfi K, Atlasz T, Kiss P, Danyadi B, Tamas A, Helyes Z, Hashimoto H, Shintani N, Baba A, Toth G, Gabriel R, Reglodi D. Mice deficient in pituitary adenylate cyclase activating polypeptide (PACAP) are more susceptible to retinal ischemic injury in vivo. Neurotox Res 2011; 21:41-8. [PMID: 21717232 DOI: 10.1007/s12640-011-9254-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Revised: 06/14/2011] [Accepted: 06/16/2011] [Indexed: 12/19/2022]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) is a neuroprotective peptide exerting protective effects in neuronal injuries. We have provided evidence that PACAP is neuroprotective in several models of retinal degeneration in vivo. Our previous studies showed that PACAP treatment ameliorated the damaging effects of chronic hypoperfusion modeled by permanent bilateral carotid artery occlusion. We have also demonstrated in earlier studies that treatment with PACAP antagonists further aggravates retinal lesions. It has been shown that PACAP deficient mice have larger infarct size in cerebral ischemia. The aim of this study was to compare the degree of retinal damage in wild type and PACAP deficient mice in ischemic retinal insult. Mice underwent 10 min of bilateral carotid artery occlusion followed by 2-week reperfusion period. Retinas were then processed for histological analysis. It was found that PACAP deficient mice had significantly greater retinal damage, as shown by the thickness of the whole retina, the morphometric analysis of the individual retinal layers, and the cell numbers in the inner nuclear and ganglion cell layers. Exogenous PACAP administration could partially protect against retinal degeneration in PACAP deficient mice. These results clearly show that endogenous PACAP reacts as a stress-response peptide that is necessary for endogenous protection against different retinal insults.
Collapse
Affiliation(s)
- K Szabadfi
- Department of Experimental Zoology, University of Pecs, Pecs, Hungary
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Ferencz A, Nedvig K, Fekecs T, Rácz B, Wéber G, Hashimoto H, Baba A, Helyes Z, Reglödi D. Comparison of intestinal cold preservation injury on pituitary adenylate cyclase-activating polypeptide in knockout and wild-type mice. Transplant Proc 2011; 42:2290-2. [PMID: 20692465 DOI: 10.1016/j.transproceed.2010.05.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Tissue injury caused by cold preservation remains a problem in small intestinal transplantation. Pituitary adenylate cyclase-activating polypeptide (PACAP) has a central role in intestinal physiology. The objective of the present study was to compare the effects of cold ischemia injury in PACAP-38 knockout and wild-type mice after cold storage of small bowel. Cold ischemia was produced using small bowel preservation in University of Wisconsin solution at 4 degrees C in 20 PACAP-38 wild-type mice for 1, 3, and 6 hours (groups 1, 2, and 3, respectively) and 20 PACAP-38 knockout mice for 1, 3, and 6 hours (groups 4, 5, and 6, respectively). Biopsy samples of small bowel were obtained after laparotomy (control) and at the end of preservation periods. To determine oxidative stress, malondialdehyde, reduced glutathione, and superoxide dismutase concentrations were measured. Tissue damage was assessed using a quantitative method on sections stained with hematoxylin-eosin. After 6 hours, tissue lipid peroxidation was increased significantly in PACAP-38 knockout mice (mean +/- SD, 153.04 +/- 7.2 micromol/g) compared with sham-operated mice (110.44 +/- 5.5 micromol/g) and wild-type mice (120.0 +/- 1.1 micromol/g) (P < .05). The capacity and activity of the endogenous antioxidant system decreased significantly after 3 and 6 hours of preservation (reduced glutathione, 808.7 +/- 5.2 micromol/g and 720.4 +/- 8.7 micromol/g; and superoxide dismutase, 125.1 +/- 1.4 IU/g and 103.3 +/- 1.9 IU/g vs 212.11 +/- 5.8 IU/g; P < .05). Quantitative histologic analysis demonstrated destruction of mucosal and submucosal layers and crypts in knockout mice compared with wild-type mice. These processes depended on duration of cold preservation. These findings demonstrate that PACAP-38 has a key role in protection against intestinal cold preservation injury.
Collapse
Affiliation(s)
- A Ferencz
- Department of Surgical Research and Techniques, University of Pécs Medical School, Pécs, Hungary.
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Horvath G, Brubel R, Kovacs K, Reglodi D, Opper B, Ferencz A, Szakaly P, Laszlo E, Hau L, Kiss P, Tamas A, Racz B. Effects of PACAP on oxidative stress-induced cell death in rat kidney and human hepatocyte cells. J Mol Neurosci 2010; 43:67-75. [PMID: 20676802 DOI: 10.1007/s12031-010-9428-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Accepted: 07/07/2010] [Indexed: 11/30/2022]
Abstract
Oxidative stress plays an important role in various renal and hepatic pathologies, and reduction of oxidative stress-induced processes is an important protective strategy in tissues of diverse origins against harmful stimuli. Pituitary adenylate cyclase activating polypeptide (PACAP) is a well-known cytotrophic and cytoprotective peptide. PACAP promotes cell survival in numerous cells and tissues exposed to various stimuli. Protective effects of PACAP have been shown in the kidney, but it is not known whether PACAP is protective against oxidative stress in renal cells. Little is known about the effects of PACAP in the liver. The aim of the present study was to investigate whether PACAP is protective against oxidative stress in primary rat kidney cell culture and whether PACAP has any effect on cell survival in human WRL-68 hepatocytes and HEP-G2 hepatocellular carcinoma cells subjected to oxidative stress. Cells were exposed to various concentrations of H(2)O(2) with or without PACAP co-treatment and cell viability was evaluated with the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide test (MTT). We found that oxidative stress induced a significant decrease in cell viability in both cell lines. PACAP could dose-dependently increase the percentage of living cells in kidney cells, but it failed to do so in hepatocytes. Given the survival-promoting effects of PACAP against oxidative stress in rat kidney, we conducted a further experiment to determine whether PACAP influences the markers of oxidative stress in vivo. We have proven earlier that PACAP was effective in kidney ischemia/reperfusion injury in vivo. In the present study, we determined the levels of the oxidative stress marker malondialdehyde and the activity of the scavenger molecules glutathione (GSH) and superoxide dismutase (SOD) following kidney ischemia/reperfusion in rats. We found that PACAP significantly increased the level of GSH and counteracted the marked reduction of SOD activity after ischemia/reperfusion in vivo. In summary, the present study showed that while PACAP was able to significantly increase the cell survival in primary kidney cell cultures exposed to oxidative stress, possibly involving interaction with the endogenous scavenger system, it failed to influence the viability of normal or cancerous hepatocytes.
Collapse
Affiliation(s)
- Gabriella Horvath
- Department of Anatomy, University of Pécs, Szigeti u 12, 7624, Pécs, Hungary.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Comparison of intestinal warm ischemic injury in PACAP knockout and wild-type mice. J Mol Neurosci 2010; 42:435-42. [PMID: 20387008 DOI: 10.1007/s12031-010-9357-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2010] [Accepted: 03/19/2010] [Indexed: 10/19/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is present in the gastrointestinal tract and plays a central role in the intestinal physiology, mainly in the secretion and motility. The aim of our study was to compare the ischemic injury in wild-type and PACAP-38 knockout mice following warm mesenteric small bowel ischemia. Warm ischemia groups were designed with occlusion of superior mesenteric artery for 1, 3, and 6 h in wild-type (n = 10 in each group) and PACAP-38 knockout (n = 10 in each group) mice. Small bowel biopsies were collected after laparotomy (control) and at the end of the ischemia periods. To determine oxidative stress parameters, malondialdehyde (MDA), reduced glutathione (GSH), and superoxide dismutase (SOD) were measured. Tissue damage was analyzed by qualitative and quantitative methods on hematoxylin/eosin-stained sections. In PACAP-38 knockout animals, tissue MDA increased significantly after 3 and 6 h ischemia (133.97 ± 6,2; 141.86 ± 5,8) compared to sham-operated (100.92 ± 3,6) and compared to wild-type results (112.8 ± 2,1; 118.4 ± 1.03 μmol/g, p < 0.05). Meanwhile, tissue concentration of GSH and activity of SOD decreased significantly in knockout mice compared to wild-type form (GSH, 795.97 ± 10.4; 665.1 ± 8,8 vs. 893.23 ± μmol/g; SOD, 94.4 ± 1.4; 81.2 ± 3.9 vs. 208.09 ± 3,7 IU/g). Qualitative and quantitative histological results showed destruction of the mucous, submucous layers, and crypts in knockout mice compared to wild-type tissues. These processes correlated with the warm ischemia periods. Our present results propose an important protective effect of endogenous PACAP-38 against intestinal warm ischemia, which provides basis for further investigation to elucidate the mechanism of this protective effect.
Collapse
|
17
|
Ohtaki H, Satoh A, Nakamachi T, Yofu S, Dohi K, Mori H, Ohara K, Miyamoto K, Hashimoto H, Shintani N, Baba A, Matsunaga M, Shioda S. Regulation of Oxidative Stress by Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) Mediated by PACAP Receptor. J Mol Neurosci 2010; 42:397-403. [DOI: 10.1007/s12031-010-9350-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2010] [Accepted: 03/09/2010] [Indexed: 11/28/2022]
|
18
|
Ferencz A, Weber G, Helyes Z, Hashimoto H, Baba A, Reglodi D. Presence of Endogenous PACAP-38 Ameliorated Intestinal Cold Preservation Tissue Injury. J Mol Neurosci 2010; 42:428-34. [DOI: 10.1007/s12031-010-9352-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2010] [Accepted: 03/12/2010] [Indexed: 10/19/2022]
|
19
|
Racz B, Horvath G, Reglodi D, Gasz B, Kiss P, Gallyas F, Sumegi B, Toth G, Nemeth A, Lubics A, Tamas A. PACAP ameliorates oxidative stress in the chicken inner ear: an in vitro study. ACTA ACUST UNITED AC 2009; 160:91-8. [PMID: 19969027 DOI: 10.1016/j.regpep.2009.12.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Revised: 11/09/2009] [Accepted: 12/02/2009] [Indexed: 11/30/2022]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) is a pleiotropic and multifunctional neuropeptide. Numerous studies prove that PACAP has neuroprotective effects in diverse neuronal systems in vitro and in vivo. The involvement of PACAP in visual and olfactory sensory processing has also been documented, but little is known about its effects in the auditory system. The presence of PACAP and its receptor, the specific PAC1 receptor, has been shown in the cochlea and in brain structures involved in auditory pathways. The aim of the present study was to investigate whether PACAP is protective in cochlear oxidative stress-induced cell death, which is known to play a role in several ototoxic insults. Chicken cochlear cells were exposed to 1mM H(2)O(2), which resulted in a marked reduction of cell viability and a parallel increase of apoptotic and necrotic cells assessed by MTT test, annexin V/propidium iodide flow cytometry and JC-1 apoptosis assay. Co-incubation with 100nM PACAP increased cell viability and reduced the percentage of apoptotic cells. Furthermore, oxidative stress increased the activation of caspase-3, while simultaneous PACAP treatment reduced it. In summary, our present results demonstrate that PACAP effectively protects cochlear cells against oxidative stress-induced apoptotic cell death.
Collapse
Affiliation(s)
- Boglarka Racz
- Department of Biochemistry and Medical Chemistry, University of Pecs, Hungary
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
PACAP and VIP differentially preserve neurovascular reactivity after global cerebral ischemia in newborn pigs. Brain Res 2009; 1283:50-7. [PMID: 19538945 DOI: 10.1016/j.brainres.2009.06.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Revised: 06/08/2009] [Accepted: 06/09/2009] [Indexed: 01/01/2023]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) and vasoactive intestinal peptide (VIP) are neuroprotective in numerous models. Impairment of cerebrovascular reactivity (CR) contributes to ischemia/reperfusion (I/R)-induced neuronal damage. We tested whether PACAP and/or VIP preserve CR to I/R-sensitive dilator responses dependent on endothelial and/or neuronal function. Accordingly, changes in pial arteriolar diameters in response to hypercapnia (5-10% CO(2) ventilation) or topical N-methyl-d-aspartate (NMDA, 10(-4) M) were determined before and after I/R via intravital microscopy in anesthetized/ventilated piglets. Local pretreatment with non-vasoactive doses of PACAP (10(-8) M) and VIP (10(-9) M) prevented the attenuation of postischemic CR to hypercapnia; to 10% CO(2), the CR values were 27+/-8% vs 92+/-5% vs 88+/-13% (vehicle vs PACAP38 vs VIP, CR expressed as a percentage of the response before I/R, mean+/-SEM, n=8-8, p<0.05). PACAP, but not VIP, preserved CR to NMDA after I/R, with CR values of 31+/-10% vs 87+/-8% vs 35+/-12% (vehicle vs PACAP38 vs VIP, n=6-6). Unlike PACAP, VIP-induced vasodilation has not yet been investigated in the piglet. We tested whether VIP-induced arteriolar dilation was sensitive to inhibitors of cyclooxygenase (COX)-1 (SC-560, 1 mg/kg), COX-2 (NS-398, 1 mg/kg), indomethacin (5 mg/kg), and nitric oxide synthase (L-NAME, 15 mg/kg). VIP (10(-8)-10(-7)-10(-6) M, n=8) induced reproducible, dose-dependent vasodilation of 16+/-3%, 33+/-6%, and 70+/-8%. The response was unaffected by all drugs, except that the vasodilation to 10(-8) M VIP was abolished by SC-560 and indomethacin. In conclusion, PACAP and VIP differentially preserve postischemic CR; independent of their vasodilatory effect.
Collapse
|
21
|
Guillot TS, Richardson JR, Wang MZ, Li YJ, Taylor TN, Ciliax BJ, Zachrisson O, Mercer A, Miller GW. PACAP38 increases vesicular monoamine transporter 2 (VMAT2) expression and attenuates methamphetamine toxicity. Neuropeptides 2008; 42:423-34. [PMID: 18533255 PMCID: PMC2569970 DOI: 10.1016/j.npep.2008.04.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2007] [Revised: 03/05/2008] [Accepted: 04/10/2008] [Indexed: 12/01/2022]
Abstract
Pituitary adenylyl cyclase activating polypeptide, 38 amino acids (PACAP38) is a brain-gut peptide with diverse physiological functions and is neuroprotective in several models of neurological disease. In this study, we show that systemic administration of PACAP38, which is transported across the blood-brain barrier, greatly reduces the neurotoxicity of methamphetamine (METH). Mice treated with PACAP38 exhibited an attenuation of striatal dopamine loss after METH exposure as well as greatly reduced markers of oxidative stress. PACAP38 treatment also prevented striatal neuroinflammation after METH administration as measured by overexpression of glial fibrillary acidic protein (GFAP), an indicator of astrogliosis, and glucose transporter 5 (GLUT5), a marker of microgliosis. In PACAP38 treated mice, the observed protective effects were not due to an altered thermal response to METH. Since the mice were not challenged with METH until 28 days after PACAP38 treatment, this suggests the neuroprotective effects are mediated by regulation of gene expression. At the time of METH administration, PACAP38 treated animals exhibited a preferential increase in the expression and function of the vesicular monoamine transporter (VMAT2). Genetic reduction of VMAT2 has been shown to increase the neurotoxicity of METH, thus we propose that the increased expression of VMAT2 may underlie the protective actions of PACAP38 against METH. The ability of PACAP38 to increase VMAT2 expression suggests that PACAP38 signaling pathways may constitute a novel therapeutic approach to treat and prevent disorders of dopamine storage.
Collapse
Affiliation(s)
- T S Guillot
- Center for Neurodegenerative Disease, Emory University, Atlanta, GA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Ferencz A, Racz B, Tamas A, Reglodi D, Lubics A, Nemeth J, Nedvig K, Kalmar-Nagy K, Horvath OP, Weber G, Roth E. Influence of PACAP on oxidative stress and tissue injury following small-bowel autotransplantation. J Mol Neurosci 2008; 37:168-76. [PMID: 18651248 DOI: 10.1007/s12031-008-9132-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2008] [Accepted: 03/31/2008] [Indexed: 02/06/2023]
Abstract
Tissue injury caused by cold preservation and reperfusion remains an unsolved problem during small-bowel transplantation. Pituitary adenylate cyclase-activating polypeptide (PACAP) is present and plays a central role in the intestinal physiology. This study investigated effect of PACAP-38 on the oxidative stress and tissue damage in autotransplanted intestine. Sham-operated, ischemia/reperfusion, and autotransplanted groups were established in Wistar rats. In ischemia/reperfusion groups, 1 h (group A), 2 h (group B), and 3 h (group C) ischemia followed by 3 h of reperfusion was applied. In autotransplanted groups, total orthotopic intestinal autotransplantation was performed. Grafts were preserved in University of Wisconsin (UW) solution and in UW containing 30 microg PACAP-38 for 1, 2, 3, and 6 h. Reperfusion lasted 3 h in all groups. Endogenous PACAP-38 concentration was measured by radioimmunoassay. To determine oxidative stress parameters, malondialdehyde, reduced glutathione, and superoxide dismutase were measured in tissue samples. Tissue damage was analyzed by qualitative and quantitative methods on hematoxylin/eosin-stained sections. Concentration of endogenous PACAP-38 significantly decreased in groups B and C compared to sham-operated group. Preservation solution containing PACAP-38 ameliorated bowel tissue oxidative injury induced by cold ischemia and reperfusion. Histological results showed that preservation caused destruction of the mucous, submucous, and muscular layers, which were further deteriorated by the end of reperfusion. In contrast, PACAP-38 significantly protected the intestinal structure. Ischemia/reperfusion decreased the endogenous PACAP-38 concentration in the intestinal tissue. Administration of PACAP-38 mitigated the oxidative injury and histological lesions in small-bowel autotransplantation model.
Collapse
Affiliation(s)
- Andrea Ferencz
- Department of Surgical Research and Techniques, University of Pécs, Medical Faculty, Kodály Zoltán Street 20, 7624, Pecs, Hungary.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide that was first isolated from an ovine hypothalamus in 1989. Since its discovery, more than 2,000 papers have reported on the tissue and cellular distribution and functional significance of PACAP. A number of papers have reported that PACAP but not the vasoactive intestinal peptide suppressed neuronal cell death or decreased infarct volume after global and focal ischemia in rodents, even if PACAP was administered several hours after ischemia induction. In addition, recent studies using PACAP gene-deficient mice demonstrated that endogenous PACAP also contributes greatly to neuroprotection similarly to exogenously administered PACAP. The studies suggest that neuroprotection by PACAP might extend the therapeutic time window for treatment of ischemia-related conditions, such as stroke. This review summarizes the effects of PACAP on ischemic neuronal cell death, and the mechanism clarified in vivo ischemic studies. In addition, the prospective mechanism of PACAP on ischemic neuroprotection from in vitro neuronal and neuronal-like cell cultures with injured stress model is reviewed. Finally, the development of PACAP and/or receptor agonists for human therapy is discussed.
Collapse
|
24
|
Rácz B, Gasz B, Borsiczky B, Gallyas F, Tamás A, Józsa R, Lubics A, Kiss P, Roth E, Ferencz A, Tóth G, Hegyi O, Wittmann I, Lengvári I, Somogyvári-Vigh A, Reglodi D. Protective effects of pituitary adenylate cyclase activating polypeptide in endothelial cells against oxidative stress-induced apoptosis. Gen Comp Endocrinol 2007; 153:115-23. [PMID: 17270184 DOI: 10.1016/j.ygcen.2006.12.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2006] [Revised: 11/15/2006] [Accepted: 12/18/2006] [Indexed: 11/16/2022]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) is a widely distributed neuropeptide that has various different functions in the nervous system and in non-neural tissues. Little is known about the effects of PACAP in endothelial cells. The aim of the present study was to investigate the effects of PACAP on endothelial cell survival and apoptotic signaling pathways under oxidative stress. Mouse hemangioendothelioma (EOMA) cells were exposed to 0.5mM H(2)O(2) which resulted in a marked reduction of cell viability and a parallel increase of apoptotic cells assessed by MTT test and flow cytometry. Co-incubation with 20nM PACAP1-38 increased cell viability and reduced the percentage of apoptotic cells. Flow cytometry analysis showed that oxidative stress reduced the phosphorylation of the anti-apoptotic ERK and increased the phosphorylation of the pro-apoptotic JNK and p38 MAP kinases. PACAP1-38 treatment ameliorated these changes: levels of phospho-ERK were elevated and those of phospho-JNK and p38 were decreased. All these effects were abolished by simultaneous treatment with the PACAP antagonist PACAP6-38. In summary, our results show that PACAP effectively protects endothelial cells against the apoptosis-inducing effects of oxidative stress.
Collapse
Affiliation(s)
- B Rácz
- Department of Surgical Research and Techniques, University of Pécs, Pécs, Hungary
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Atlasz T, Babai N, Kiss P, Reglodi D, Tamás A, Szabadfi K, Tóth G, Hegyi O, Lubics A, Gábriel R. Pituitary adenylate cyclase activating polypeptide is protective in bilateral carotid occlusion-induced retinal lesion in rats. Gen Comp Endocrinol 2007; 153:108-14. [PMID: 17289046 DOI: 10.1016/j.ygcen.2006.12.022] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2006] [Revised: 12/21/2006] [Accepted: 12/25/2006] [Indexed: 10/23/2022]
Abstract
Pituitary adenylate cyclase activating popypeptide (PACAP) is a pleiotropic neuropeptide, exerting neurotrophic and neuroprotective effects in numerous models of in vitro and in vivo nervous injuries. The aim of the present study was to investigate whether PACAP is neuroprotective in ischemic retinal damage. Adult male Wistar rats underwent bilateral carotid occlusion and PACAP was administered unilaterally into the vitreous body immediately following carotid occlusion. Retinas were analyzed three weeks after the injury. It was found that bilateral carotid occlusion led to a severe degeneration of all retinal layers. PACAP treatment significantly ameliorated the carotid occlusion-induced retinal damage: the overall thickness of the retina was significantly more than in control carotid occluded animals and the morphological characteristics of the photoreceptors showed nearly normal appearance. The outer plexiform layer remained discernible and the inner and outer nuclear layers were significantly thicker than in control animals. In summary, our present study provides evidence, for the first time, that PACAP attenuates ischemic retinal degeneration.
Collapse
Affiliation(s)
- Tamás Atlasz
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Gasz B, Rácz B, Roth E, Borsiczky B, Ferencz A, Tamás A, Cserepes B, Lubics A, Gallyas F, Tóth G, Lengvári I, Reglodi D. Pituitary adenylate cyclase activating polypeptide protects cardiomyocytes against oxidative stress-induced apoptosis. Peptides 2006; 27:87-94. [PMID: 16095757 DOI: 10.1016/j.peptides.2005.06.022] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2005] [Revised: 06/27/2005] [Accepted: 06/30/2005] [Indexed: 11/17/2022]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) has well-known neuroprotective effects, and one of the main factors leading to neuroprotection seems to be its anti-apoptotic effects. The peptide and its receptors are present also in the heart, but whether PACAP can be protective in cardiomyocytes, is not known. Therefore, the aim of the present study was to investigate the effects of PACAP on oxidative stress-induced apoptosis in cardiomyocytes. Our results show that PACAP increased cell viability by attenuating H2O2-induced apoptosis in a cardiac myocyte culture. PACAP also decreased caspase-3 activity and increased the expression of the anti-apoptotic markers Bcl-2 and phospho-Bad. These effects of PACAP were counteracted by the PACAP antagonist PACAP6-38. In summary, our results show that PACAP is able to attenuate oxidative stress-induced cardiomyocyte apoptosis.
Collapse
Affiliation(s)
- B Gasz
- Department of Surgical Research and Techniques, Medical Faculty, Pécs University, Hungary
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|