1
|
Vékony RG, Tamás A, Lukács A, Ujfalusi Z, Lőrinczy D, Takács-Kollár V, Bukovics P. Exploring the Role of Neuropeptide PACAP in Cytoskeletal Function Using Spectroscopic Methods. Int J Mol Sci 2024; 25:8063. [PMID: 39125632 PMCID: PMC11311697 DOI: 10.3390/ijms25158063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
The behavior and presence of actin-regulating proteins are characteristic of various clinical diseases. Changes in these proteins significantly impact the cytoskeletal and regenerative processes underlying pathological changes. Pituitary adenylate cyclase-activating polypeptide (PACAP), a cytoprotective neuropeptide abundant in the nervous system and endocrine organs, plays a key role in neuron differentiation and migration by influencing actin. This study aims to elucidate the role of PACAP as an actin-regulating polypeptide, its effect on actin filament formation, and the underlying regulatory mechanisms. We examined PACAP27, PACAP38, and PACAP6-38, measuring their binding to actin monomers via fluorescence spectroscopy and steady-state anisotropy. Functional polymerization tests were used to track changes in fluorescent intensity over time. Unlike PACAP27, PACAP38 and PACAP6-38 significantly reduced the fluorescence emission of Alexa488-labeled actin monomers and increased their anisotropy, showing nearly identical dissociation equilibrium constants. PACAP27 showed weak binding to globular actin (G-actin), while PACAP38 and PACAP6-38 exhibited robust interactions. PACAP27 did not affect actin polymerization, but PACAP38 and PACAP6-38 accelerated actin incorporation kinetics. Fluorescence quenching experiments confirmed structural changes upon PACAP binding; however, all studied PACAP fragments exhibited the same effect. Our findings indicate that PACAP38 and PACAP6-38 strongly bind to G-actin and significantly influence actin polymerization. Further studies are needed to fully understand the biological significance of these interactions.
Collapse
Affiliation(s)
- Roland Gábor Vékony
- Department of Biophysics, Medical School, University of Pécs, 7624 Pécs, Hungary; (R.G.V.); (A.L.); (Z.U.); (D.L.); (V.T.-K.)
| | - Andrea Tamás
- Department of Anatomy, Medical School, University of Pécs, 7624 Pécs, Hungary;
| | - András Lukács
- Department of Biophysics, Medical School, University of Pécs, 7624 Pécs, Hungary; (R.G.V.); (A.L.); (Z.U.); (D.L.); (V.T.-K.)
| | - Zoltán Ujfalusi
- Department of Biophysics, Medical School, University of Pécs, 7624 Pécs, Hungary; (R.G.V.); (A.L.); (Z.U.); (D.L.); (V.T.-K.)
| | - Dénes Lőrinczy
- Department of Biophysics, Medical School, University of Pécs, 7624 Pécs, Hungary; (R.G.V.); (A.L.); (Z.U.); (D.L.); (V.T.-K.)
| | - Veronika Takács-Kollár
- Department of Biophysics, Medical School, University of Pécs, 7624 Pécs, Hungary; (R.G.V.); (A.L.); (Z.U.); (D.L.); (V.T.-K.)
| | - Péter Bukovics
- Department of Biophysics, Medical School, University of Pécs, 7624 Pécs, Hungary; (R.G.V.); (A.L.); (Z.U.); (D.L.); (V.T.-K.)
| |
Collapse
|
2
|
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
|
3
|
Pathomechanisms and therapeutic opportunities in radiation-induced heart disease: from bench to bedside. Clin Res Cardiol 2021; 110:507-531. [PMID: 33591377 PMCID: PMC8055626 DOI: 10.1007/s00392-021-01809-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/16/2021] [Indexed: 12/14/2022]
Abstract
Cancer management has undergone significant improvements, which led to increased long-term survival rates among cancer patients. Radiotherapy (RT) has an important role in the treatment of thoracic tumors, including breast, lung, and esophageal cancer, or Hodgkin's lymphoma. RT aims to kill tumor cells; however, it may have deleterious side effects on the surrounding normal tissues. The syndrome of unwanted cardiovascular adverse effects of thoracic RT is termed radiation-induced heart disease (RIHD), and the risk of developing RIHD is a critical concern in current oncology practice. Premature ischemic heart disease, cardiomyopathy, heart failure, valve abnormalities, and electrical conduct defects are common forms of RIHD. The underlying mechanisms of RIHD are still not entirely clear, and specific therapeutic interventions are missing. In this review, we focus on the molecular pathomechanisms of acute and chronic RIHD and propose preventive measures and possible pharmacological strategies to minimize the burden of RIHD.
Collapse
|
4
|
Jungling A, Reglodi D, Maasz G, Zrinyi Z, Schmidt J, Rivnyak A, Horvath G, Pirger Z, Tamas A. Alterations of Nigral Dopamine Levels in Parkinson's Disease after Environmental Enrichment and PACAP Treatment in Aging Rats. Life (Basel) 2021; 11:life11010035. [PMID: 33429934 PMCID: PMC7827131 DOI: 10.3390/life11010035] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 12/29/2020] [Accepted: 01/04/2021] [Indexed: 11/16/2022] Open
Abstract
The neuroprotective effects of environmental enrichment and PACAP (pituitary adenylate cyclase-activating polypeptide) are well-described in Parkinson’s disease. The aim of our study is to investigate the beneficial effects of these factors in aging parkinsonian rats. Newborn Wistar rats were divided into standard and enriched groups according to their environmental conditions. Standard animals were raised under regular conditions. During the first five postnatal weeks, enriched pups were placed in larger cages with different objects. Aging animals received (1) saline, (2) 6-hydroxidopamine (6-OHDA), or (3) 6-OHDA + PACAP injections into the left substantia nigra (s.n.). On the seventh postoperative day, the left and right s.n. were collected. The s.n. of young and aging unoperated animals were also examined in our experiment. We determined the dopamine (DA) levels by the HPLC-MS technique, while the sandwich ELISA method was used to measure the Parkinson disease protein 7 (PARK7) protein levels. In healthy animals, we found an age-related decrease of DA levels. In aging parkinsonian-enriched rats, the operation did not result in a significant DA loss. PACAP treatment could prevent the DA loss in both the standard and enriched groups. All injured PACAP-treated rats showed remarkably higher protective PARK7 levels. The protective effect of PACAP correlated with the increase of the DA and PARK7 levels.
Collapse
Affiliation(s)
- Adel Jungling
- MTA-PTE PACAP Research Team, Department of Anatomy, Medical School, University of Pecs, 7624 Pecs, Hungary; (A.J.); (D.R.); (A.R.); (G.H.)
| | - Dora Reglodi
- MTA-PTE PACAP Research Team, Department of Anatomy, Medical School, University of Pecs, 7624 Pecs, Hungary; (A.J.); (D.R.); (A.R.); (G.H.)
| | - Gabor Maasz
- MTA-OK BLI NAP_B Adaptive Neuroethology, Department of Experimental Zoology, Balaton Limnological Institute, MTA-CER, 8237 Tihany, Hungary; (G.M.); (Z.Z.); (Z.P.)
| | - Zita Zrinyi
- MTA-OK BLI NAP_B Adaptive Neuroethology, Department of Experimental Zoology, Balaton Limnological Institute, MTA-CER, 8237 Tihany, Hungary; (G.M.); (Z.Z.); (Z.P.)
| | - Janos Schmidt
- Institute of Biochemistry and Medical Chemistry, Medical School, University of Pecs, 7624 Pecs, Hungary;
| | - Adam Rivnyak
- MTA-PTE PACAP Research Team, Department of Anatomy, Medical School, University of Pecs, 7624 Pecs, Hungary; (A.J.); (D.R.); (A.R.); (G.H.)
| | - Gabor Horvath
- MTA-PTE PACAP Research Team, Department of Anatomy, Medical School, University of Pecs, 7624 Pecs, Hungary; (A.J.); (D.R.); (A.R.); (G.H.)
| | - Zsolt Pirger
- MTA-OK BLI NAP_B Adaptive Neuroethology, Department of Experimental Zoology, Balaton Limnological Institute, MTA-CER, 8237 Tihany, Hungary; (G.M.); (Z.Z.); (Z.P.)
| | - Andrea Tamas
- MTA-PTE PACAP Research Team, Department of Anatomy, Medical School, University of Pecs, 7624 Pecs, Hungary; (A.J.); (D.R.); (A.R.); (G.H.)
- Correspondence: or ; Tel.: +36-72-536-001 (ext. 36421)
| |
Collapse
|
5
|
Edvinsson JCA, Grell AS, Warfvinge K, Sheykhzade M, Edvinsson L, Haanes KA. Differences in pituitary adenylate cyclase-activating peptide and calcitonin gene-related peptide release in the trigeminovascular system. Cephalalgia 2020; 40:1296-1309. [DOI: 10.1177/0333102420929026] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background Several neurotransmitters are expressed in the neurons of the trigeminal ganglion. One such signalling molecule is the pituitary adenylate cyclase-activating peptide (PACAP). PACAP signalling has been suggested to have a possible role in the pathophysiology of primary headaches. Objective The present study was designed to investigate the relationship between PACAP and calcitonin gene-related peptide, currently the two most relevant migraine peptides. Methods In the current study, we used ELISA to investigate PACAP and calcitonin gene-related peptide release in response to 60 mM K+ or capsaicin using a rat hemi-skull model. We combined this analysis with qPCR and immunohistochemistry to study the expression of PACAP and calcitonin gene-related peptide receptors and ligands. Results Calcitonin gene-related peptide (CGRP) is released from the trigeminal ganglion and dura mater. In contrast, PACAP is only released from the trigeminal ganglion. We observed a weak correlation between the stimulated release of the two neuropeptides. PACAP-38 immunoreactivity was expressed alone and in a subpopulation of neurons in the trigeminal ganglion that also store calcitonin gene-related peptide. The receptor subtype PAC1 was mainly expressed in the satellite glial cells (SGCs), which envelop the neurons in the trigeminal ganglion, in some neuronal processes, inside the Aδ-fibres and in the outermost layer of the myelin sheath that envelopes the Aδ-fibres. Conclusion Unlike CGRP, PACAP is only released within the trigeminal ganglion. This raises the question of whether a migraine therapy aimed at preventing peripheral PACAP signalling would be as successful as the CGRP signalling targeted treatments.
Collapse
Affiliation(s)
- Jacob Carl Alexander Edvinsson
- Department of Clinical Experimental Research, Glostrup Research Institute, Rigshospitalet Glostrup, Glostrup, Denmark
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne-Sofie Grell
- Department of Clinical Experimental Research, Glostrup Research Institute, Rigshospitalet Glostrup, Glostrup, Denmark
| | - Karin Warfvinge
- Department of Clinical Experimental Research, Glostrup Research Institute, Rigshospitalet Glostrup, Glostrup, Denmark
- Department of Medicine, Institute of Clinical Sciences, Lund University, Lund, Sweden
| | - Majid Sheykhzade
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lars Edvinsson
- Department of Clinical Experimental Research, Glostrup Research Institute, Rigshospitalet Glostrup, Glostrup, Denmark
- Department of Medicine, Institute of Clinical Sciences, Lund University, Lund, Sweden
| | - Kristian Agmund Haanes
- Department of Clinical Experimental Research, Glostrup Research Institute, Rigshospitalet Glostrup, Glostrup, Denmark
| |
Collapse
|
6
|
Splitthoff P, Rasbach E, Neudert P, Bonaterra GA, Schwarz A, Mey L, Schwarzbach H, Eiden LE, Weihe E, Kinscherf R. PAC1 deficiency attenuates progression of atherosclerosis in ApoE deficient mice under cholesterol-enriched diet. Immunobiology 2020; 225:151930. [PMID: 32173151 PMCID: PMC9741700 DOI: 10.1016/j.imbio.2020.151930] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/27/2020] [Accepted: 02/28/2020] [Indexed: 12/14/2022]
Abstract
The neuropeptide, pituitary adenylate cyclase-activating polypeptide (PACAP) is vasoactive and cytoprotective and exerts immunoregulatory functions throughout the nervous, neuroendocrine cardiovascular and immune systems in health and disease. PACAP mainly acts through PAC1 receptor signaling in neuronal communication, but the role of PAC1 in immune regulation of atherosclerosis is not known. Here, we generated PAC1-/-/ApoE-/- mice to test, whether PAC1-/- influences plasma cholesterol-/triglyceride levels and/or atherogenesis in the brachiocephalic trunk (BT) seen in ApoE-/- mice, under standard chow (SC) or cholesterol-enriched diet (CED). Furthermore, the effect of PAC1-/-, on inflammatory, autophagy-, apoptosis- and necroptosis-relevant proteins in atherosclerotic plaques was determined. In plaques of PAC1-/-/ApoE-/- mice fed a SC, the immunoreactivity for apoptotic, autophagic, necroptotic and proinflammatory proteins was increased, however, proliferation was unaffected. Interestingly, without affecting hyperlipidemia, PAC1-/- in ApoE-/- mice remarkably reduced CED-induced lumen stenosis seen in ApoE-/- mice. Thus, PAC1-/- allows unchecked inflammation, necroptosis and decreased proliferation during SC, apparently priming the BT to develop reduced atheroma under subsequent CED. Remarkably, no differences in inflammation/necroptosis signatures in the atheroma under CED between PAC1-/-/ApoE-/- and ApoE-/- mice were observed. These data indicate that selective PAC1 antagonists should offer potential as a novel class of atheroprotective therapeutics, especially during hypercholesterolemia.
Collapse
Affiliation(s)
- Paul Splitthoff
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Medical Faculty, Philipps-University of Marburg, Robert-Koch-Str. 8, 35037, Marburg, Germany
| | - Erik Rasbach
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Medical Faculty, Philipps-University of Marburg, Robert-Koch-Str. 8, 35037, Marburg, Germany
| | - Philip Neudert
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Medical Faculty, Philipps-University of Marburg, Robert-Koch-Str. 8, 35037, Marburg, Germany
| | - Gabriel A. Bonaterra
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Medical Faculty, Philipps-University of Marburg, Robert-Koch-Str. 8, 35037, Marburg, Germany,Corresponding author at: Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Medical Faculty, Philipps-University of Marburg, Robert-Koch-Str. 8, 35032, Marburg, Germany., (G.A. Bonaterra)
| | - Anja Schwarz
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Medical Faculty, Philipps-University of Marburg, Robert-Koch-Str. 8, 35037, Marburg, Germany
| | - Lilli Mey
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Medical Faculty, Philipps-University of Marburg, Robert-Koch-Str. 8, 35037, Marburg, Germany
| | - Hans Schwarzbach
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Medical Faculty, Philipps-University of Marburg, Robert-Koch-Str. 8, 35037, Marburg, Germany
| | - Lee E. Eiden
- Section on Molecular Neuroscience, Laboratory of Cellular and Molecular Regulation, National Institute of Mental Health Intramural Research Program, Bethesda, 20814, Maryland, USA
| | - Eberhard Weihe
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Medical Faculty, Philipps-University of Marburg, Robert-Koch-Str. 8, 35037, Marburg, Germany
| | - Ralf Kinscherf
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Medical Faculty, Philipps-University of Marburg, Robert-Koch-Str. 8, 35037, Marburg, Germany
| |
Collapse
|
7
|
Toth D, Szabo E, Tamas A, Juhasz T, Horvath G, Fabian E, Opper B, Szabo D, Maugeri G, D'Amico AG, D'Agata V, Vicena V, Reglodi D. Protective Effects of PACAP in Peripheral Organs. Front Endocrinol (Lausanne) 2020; 11:377. [PMID: 32765418 PMCID: PMC7381171 DOI: 10.3389/fendo.2020.00377] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 05/12/2020] [Indexed: 12/21/2022] Open
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) is a neuropeptide widely distributed in the nervous system, where it exerts strong neuroprotective effects. PACAP is also expressed in peripheral organs but its peripheral protective effects have not been summarized so far. Therefore, the aim of the present paper is to review the existing literature regarding the cytoprotective effects of PACAP in non-neuronal cell types, peripheral tissues, and organs. Among others, PACAP has widespread expression in the digestive system, where it shows protective effects in various intestinal pathologies, such as duodenal ulcer, small bowel ischemia, and intestinal inflammation. PACAP is present in both the exocrine and endocrine pancreas as well as liver where it reduces inflammation and steatosis by interfering with hepatic pathology related to obesity. It is found in several exocrine glands and also in urinary organs, where, with its protective effects being mainly published regarding renal pathologies, PACAP is protective in numerous conditions. PACAP displays anti-inflammatory effects in upper and lower airways of the respiratory system. In the skin, it is involved in the development of inflammatory pathology such as psoriasis and also has anti-allergic effects in a model of contact dermatitis. In the non-neuronal part of the visual system, PACAP showed protective effects in pathological conditions of the cornea and retinal pigment epithelial cells. The positive role of PACAP has been demonstrated on the formation and healing processes of cartilage and bone where it also prevents osteoarthritis and rheumatoid arthritis development. The protective role of PACAP was also demonstrated in the cardiovascular system in different pathological processes including hyperglycaemia-induced endothelial dysfunction and age-related vascular changes. In the heart, PACAP protects against ischemia, oxidative stress, and cardiomyopathies. PACAP is also involved in the protection against the development of pre-senile systemic amyloidosis, which is presented in various peripheral organs in PACAP-deficient mice. The studies summarized here provide strong evidence for the cytoprotective effects of the peptide. The survival-promoting effects of PACAP depend on a number of factors which are also shortly discussed in the present review.
Collapse
Affiliation(s)
- Denes Toth
- Department of Forensic Medicine, MTA-PTE PACAP Research Team, University of Pécs Medical School, Pécs, Hungary
| | - Edina Szabo
- Department of Anatomy, MTA-PTE PACAP Research Team, University of Pécs Medical School, Pécs, Hungary
| | - Andrea Tamas
- Department of Anatomy, MTA-PTE PACAP Research Team, University of Pécs Medical School, Pécs, Hungary
| | - Tamas Juhasz
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Department of Biomedical and Biotechnological Sciences, Section of Human Anatomy and Histology, University of Catania, Catania, Italy
| | - Gabriella Horvath
- Department of Anatomy, MTA-PTE PACAP Research Team, University of Pécs Medical School, Pécs, Hungary
| | - Eszter Fabian
- Department of Anatomy, MTA-PTE PACAP Research Team, University of Pécs Medical School, Pécs, Hungary
| | - Balazs Opper
- Department of Anatomy, MTA-PTE PACAP Research Team, University of Pécs Medical School, Pécs, Hungary
| | - Dora Szabo
- Heart Institute, Medical School, University of Pécs, Pécs, Hungary
| | - Grazia Maugeri
- Department of Biomedical and Biotechnological Sciences, Section of Human Anatomy and Histology, University of Catania, Catania, Italy
| | - Agata G. D'Amico
- Department of Drug Sciences, University of Catania, Catania, Italy
| | - Velia D'Agata
- Department of Biomedical and Biotechnological Sciences, Section of Human Anatomy and Histology, University of Catania, Catania, Italy
| | - Viktoria Vicena
- Department of Anatomy, MTA-PTE PACAP Research Team, University of Pécs Medical School, Pécs, Hungary
| | - Dora Reglodi
- Department of Anatomy, MTA-PTE PACAP Research Team, University of Pécs Medical School, Pécs, Hungary
- *Correspondence: Dora Reglodi
| |
Collapse
|
8
|
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:ijms20194944. [PMID: 31591326 PMCID: PMC6801442 DOI: 10.3390/ijms20194944] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 09/20/2019] [Accepted: 10/01/2019] [Indexed: 12/11/2022] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a pleiotropic neuropeptide with a widespread distribution throughout the entire body including the urinary system. PACAP exerts protective actions in different injury models related to several organ systems. Its protective effect is mainly based on its antiapoptotic, anti-inflammatory and antioxidant effects. The present review aims to summarize the effects of PACAP in pathologies associated with inflammation and oxidative stress-induced damage in the kidney. Both in vitro and in vivo data are available proving its protective actions against oxidative stress, hypoxia, renal ischemia/reperfusion, diabetic nephropathy, myeloma kidney injury, amyloidosis and different types of drug-induced nephropathies. Data showing the nephroprotection by PACAP emphasize the potential of PACAP’s therapeutic use in various renal pathologies.
Collapse
|
9
|
Denes V, Geck P, Mester A, Gabriel R. Pituitary Adenylate Cyclase-Activating Polypeptide: 30 Years in Research Spotlight and 600 Million Years in Service. J Clin Med 2019; 8:jcm8091488. [PMID: 31540472 PMCID: PMC6780647 DOI: 10.3390/jcm8091488] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 09/02/2019] [Accepted: 09/10/2019] [Indexed: 12/12/2022] Open
Abstract
Emerging from the depths of evolution, pituitary adenylate cyclase-activating polypeptide (PACAP) and its receptors (i.e., PAC1, VPAC1, VPAC2) are present in multicellular organisms from Tunicates to humans and govern a remarkable number of physiological processes. Consequently, the clinical relevance of PACAP systems spans a multifaceted palette that includes more than 40 disorders. We aimed to present the versatility of PACAP1-38 actions with a focus on three aspects: (1) when PACAP1-38 could be a cause of a malfunction, (2) when PACAP1-38 could be the cure for a malfunction, and (3) when PACAP1-38 could either improve or impair biology. PACAP1-38 is implicated in the pathophysiology of migraine and post-traumatic stress disorder whereas an outstanding protective potential has been established in ischemia and in Alzheimer’s disease. Lastly, PACAP receptors could mediate opposing effects both in cancers and in inflammation. In the light of the above, the duration and concentrations of PACAP agents must be carefully set at any application to avoid unwanted consequences. An enormous amount of data accumulated since its discovery (1989) and the first clinical trials are dated in 2017. Thus in the field of PACAP research: “this is not the end, not even the beginning of the end, but maybe the end of the beginning.”
Collapse
Affiliation(s)
- Viktoria Denes
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary.
| | - Peter Geck
- Department of Immunology, School of Medicine, Tufts University, Boston, MA 02111, USA.
| | - Adrienn Mester
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary.
| | - Robert Gabriel
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary.
| |
Collapse
|
10
|
Protective Effect of PACAP on Ischemia/Reperfusion-Induced Kidney Injury of Male and Female Rats: Gender Differences. J Mol Neurosci 2018; 68:408-419. [PMID: 30443839 DOI: 10.1007/s12031-018-1207-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 11/08/2018] [Indexed: 02/07/2023]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide that exerts general cytoprotective effects, including protection in different kidney disorders. The aim of our study was to investigate the ischemia/reperfusion-induced kidney injury of male and female rats to confirm the protective effects of PACAP in the kidney and to reveal possible gender differences.Male and female Wistar rats underwent unilateral renal artery clamping followed by 24-h, 48-h, or 14-day reperfusion. PACAP was administered intravenously before arterial clamping in half of the rats. Tubular damage, cytokine expression pattern, oxidative stress marker, antioxidative status and signaling pathways were evaluated using histology, immunohistology, cytokine array, PCR, and Western blot. Tubular damage was significantly less severe in the PACAP-treated male and female rats compared to controls. Results of female animals were significantly better in both treated and untreated groups. Cytokine expression, oxidative stress marker and antioxidative status confirmed the histological results. We also revealed that PACAP counteracted the decreased PKA phosphorylation, influenced the expression of BMP2 and BMP4, and increased the expression of the protein Smad1.We conclude that PACAP is protective in ischemia/reperfusion-induced kidney injury in both sexes, but females had markedly less pronounced injury after ischemia/reperfusion, possibly also involving further protective factors, the investigation of which could have future therapeutic value in treating ischemic kidney injuries.
Collapse
|
11
|
Rasbach E, Splitthoff P, Bonaterra GA, Schwarz A, Mey L, Schwarzbach H, Eiden LE, Weihe E, Kinscherf R. PACAP deficiency aggravates atherosclerosis in ApoE deficient mice. Immunobiology 2018; 224:124-132. [PMID: 30447883 DOI: 10.1016/j.imbio.2018.09.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 09/28/2018] [Indexed: 12/31/2022]
Abstract
Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) plays an important role in cytoprotection, inflammation and cardiovascular regulation. Thus, we studied the involvement of PACAP in atherogenesis. Differentiated human THP-1 macrophages (MΦ) were stimulated with oxidized low-density lipoproteins (oxLDL) and the influence of PACAP38 treatment on lipid content and TNF release was determined. To test the effect of PACAP deficiency (PACAP-/-) on the development of atherosclerosis under standard chow (SC) or cholesterol-enriched diet (CED) in vivo, PACAP-/- mice were crossbred with ApoE-/- to generate PACAP-/-/ApoE-/- mice. Blood cholesterol and triglyceride levels were quantified. Lumen stenosis in the brachiocephalic trunk, cellularity and amounts of pro-inflammatory as well as autophagy-, apoptosis- and necroptosis-relevant proteins were analysed in atherosclerotic plaques by quantitative immunohistochemistry. In vitro, PACAP38 inhibited oxLDL-induced intracellular lipid storage as well as TNF release in MФ. In vivo, after SC, but not under CED, PACAP-/-/ApoE-/- mice showed an increased lumen stenosis compared to ApoE-/- mice. In atherosclerotic plaques of PACAP-/-/ApoE-/- mice, the immunoreactive areas of TNF+, IL-1β+, autophagic, apoptotic and necroptotic cells were increased. In contrast, the overall cell density was decreased compared to ApoE-/- under SC, while no differences were seen under CED. Similar plasma cholesterol levels were observed in PACAP-/-/ApoE-/- and ApoE-/- mice under the respective feeding regime. Thus, PACAP-/-/ApoE-/- mice represent a novel mouse model of accelerated atherosclerosis where CED is not required. Our data indicate that PACAP acts as an endogenous atheroprotective neuropeptide. Thus, stable PACAP agonists may have potential as anti-atherosclerotic therapeutics. The specific PACAP receptor(s) mediating atheroprotection remain(s) to be identified.
Collapse
Affiliation(s)
- Erik Rasbach
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Medical Faculty, Philipps-University of Marburg, Robert-Koch-Str. 8, 35037 Marburg, Germany
| | - Paul Splitthoff
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Medical Faculty, Philipps-University of Marburg, Robert-Koch-Str. 8, 35037 Marburg, Germany
| | - Gabriel A Bonaterra
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Medical Faculty, Philipps-University of Marburg, Robert-Koch-Str. 8, 35037 Marburg, Germany.
| | - Anja Schwarz
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Medical Faculty, Philipps-University of Marburg, Robert-Koch-Str. 8, 35037 Marburg, Germany
| | - Lilli Mey
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Medical Faculty, Philipps-University of Marburg, Robert-Koch-Str. 8, 35037 Marburg, Germany
| | - Hans Schwarzbach
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Medical Faculty, Philipps-University of Marburg, Robert-Koch-Str. 8, 35037 Marburg, Germany
| | - Lee E Eiden
- Section on Molecular Neuroscience, Laboratory of Cellular and Molecular Regulation, National Institute of Mental Health Intramural Research Program, Bethesda, 20814 MD, USA
| | - Eberhard Weihe
- Department of Molecular and Cellular Neuroscience, Institute for Anatomy and Cell Biology, Medical Faculty, Philipps-University of Marburg Robert-Koch-Str. 8, 35037 Marburg, Germany
| | - Ralf Kinscherf
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Medical Faculty, Philipps-University of Marburg, Robert-Koch-Str. 8, 35037 Marburg, Germany
| |
Collapse
|
12
|
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
|
13
|
Kasica-Jarosz N, Podlasz P, Kaleczyc J. Pituitary adenylate cyclase-activating polypeptide (PACAP-38) plays an inhibitory role against inflammation induced by chemical damage to zebrafish hair cells. PLoS One 2018; 13:e0198180. [PMID: 29856797 PMCID: PMC5983416 DOI: 10.1371/journal.pone.0198180] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Accepted: 05/15/2018] [Indexed: 12/20/2022] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP-38) is a common neuropeptide exerting a wide spectrum of functions in many fields, including immunology. In the present study, 5-day post-fertilization (dpf) zebrafish larvae of three diverse genetic lines [transgenic lines Tg(MPX:GFP) with GFP-labelled neutrophils and Tg(pou4f3:GAP-GFP) with GFP-labelled hair cells and the wild-type Tuebingen] were used to investigate an inhibitory role of PACAP-38 in inflammation associated with damaged hair cells of the lateral line. Individuals of each genetic line were assigned to four groups: (1) control, and those consisting of larvae exposed to (2) 10 µM CuSO4, (3) 10 µM CuSO4+100 nM PACAP-38 and (4) 100 nM PACAP-38, respectively. Forty-minute exposure to CuSO4 solution was applied to evoke necrosis of hair cells and consequent inflammation. The inhibitory role of PACAP-38 was investigated in vivo under a confocal microscope by counting neutrophils migrating towards damaged hair cells in Tg(MPX:GFP) larvae. In CuSO4-treated individuals, the number of neutrophils associated with hair cells was dramatically increased, while PACAP-38 co-treatment resulted in its over 2-fold decrease. However, co-treatment with PACAP-38 did not prevent hair cells from extensive necrosis, which was found in Tg(pou4f3:GAP-GFP) individuals. Real-Time PCR analysis performed in wild-type larvae demonstrated differential expression pattern of stress and inflammation inducible markers. The most significant findings showed that CuSO4 exposure up-regulated the expression of IL-8, IL-1β, IL-6 and ATF3, while after PACAP-38 co-treatment expression levels of these genes were significantly decreased. The presence of transcripts for all PACAP receptors in neutrophils was also revealed. Adcyap1r1a and vipr1b appeared to be predominant forms. The present results suggest that PACAP-38 should be considered as a factor playing an important regulatory role in inflammatory response associated with pathological processes affecting zebrafish hair cells and it cannot be excluded that this interesting property has more universal significance.
Collapse
Affiliation(s)
- Natalia Kasica-Jarosz
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
- * E-mail: (NK); (PP)
| | - Piotr Podlasz
- Department of Pathophysiology, Forensic Veterinary and Administration, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
- * E-mail: (NK); (PP)
| | - Jerzy Kaleczyc
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| |
Collapse
|
14
|
Examination of Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) as a Potential Biomarker in Heart Failure Patients. J Mol Neurosci 2018; 68:368-376. [PMID: 29353438 DOI: 10.1007/s12031-017-1025-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 12/25/2017] [Indexed: 01/06/2023]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a pleiotropic and multifunctional neuropeptide having neurotrophic, neuroprotective, and general cytoprotective actions in a variety of tissues based on its anti-apoptotic, anti-inflammatory, and antioxidant effects. Several studies have demonstrated its cardioprotective effects in vitro and in various animal models. However, few data are available on the presence of PACAP in human cardiac tissues and its role in the pathomechanism and progression of different cardiac disorders, particularly heart failure. Earlier, our research group has shown PAC1 receptor immunoreactivity in human heart tissue samples and we have found significantly elevated PACAP27- and PACAP38-like immunoreactivity in ischemic cardiac samples compared to valvular abnormalities with radioimmunoassay. In the last few years, numerous studies examined the presence and the changes of PACAP levels in different human tissue samples and biological fluids to show alterations in different physiological and pathological conditions. Therefore, the aim of the present study was to measure the alterations of blood PACAP levels in chronic heart failure caused by primary dilated cardiomyopathy or ischemic cardiomyopathy and to examine the possible relationship between serum levels of PACAP, N-terminal prohormone of brain natriuretic peptide (NT-proBNP), and systolic left ventricular function, the most reliable biomarkers of heart failure. In the group of mild heart failure patients, a significant strong negative correlation was detected. Furthermore, in moderate heart failure, we found a significant moderate negative correlation between PACAP and NT-proBNP levels only in ischemic subgroup. Positive correlation was found between serum PACAP level and ejection fraction only in patients with heart failure due to ischemic cardiomyopathy but not in patients with primary dilated cardiomyopathy. In summary, remarkable differences were observed between the ischemic and non-ischemic heart failure suggesting that PACAP might play an important role in the pathomechanism and progression of ischemic heart failure and it might be a potential biomarker of cardiac diseases in the future.
Collapse
|
15
|
Subramaniam V, Chuang G, Xia H, Burn B, Bradley J, Maderdrut JL, Coy DH, Varner KJ. Pituitary adenylate cyclase-activating polypeptide (PACAP) protects against mitoxantrone-induced cardiac injury in mice. Peptides 2017; 95:25-32. [PMID: 28720396 PMCID: PMC5568240 DOI: 10.1016/j.peptides.2017.07.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 07/06/2017] [Accepted: 07/11/2017] [Indexed: 12/30/2022]
Abstract
Mitoxantrone (MXT) is an androstenedione that is used to treat cancers and progressive forms of multiple sclerosis; however, its use is limited by its cardiotoxicity. Pituitary adenylate cyclase activating polypeptide (PACAP) is a member of the secretin/growth hormone-releasing hormone/vasoactive intestinal peptide family and has many functions, including cytoprotection and immunosuppression. We tested the hypothesis that PACAP can protect against MXT-induced cardiotoxicity in mice. Female BALB/c mice were treated once weekly for 4 weeks with saline (n=14) or MXT (3mg/kg, i.p.; n=14). Half of the mice in each group received PACAP (10μg, i.p.) 1h before and 24 and 48h after MXT, while the remaining mice received injections of saline on the same schedule. Echocardiography was used to assess cardiac structure and function. In mice treated with MXT and saline, body weight was significantly reduced after the third dose of MXT. PACAP significantly attenuated the reduction in body weight; however, the weights did not return to control level. Compared to controls, MXT-treated mice had significantly increased left ventricular (LV) diameter and LV volume and decreased LV posterior wall thickness. Fractional shortening (FS) and ejection fraction (EF) were also significantly decreased. Treatment with PACAP prevented MXT-induced LV dilation and significantly attenuated the reductions in FS and EF, although FS and EF did not return to control level. PACAP38 did not prevent MXT-induced decreases in LV posterior wall thickness. MXT dose-dependently decreased the viability of cultured U937 (human leukemia) cells; PACAP did not protect cultured U937 cells from MXT-mediated cell death. In conclusion, PACAP can attenuate MXT-mediated LV dilation and dysfunction in mice.
Collapse
Affiliation(s)
- Venkat Subramaniam
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University, Health Sciences Center, New Orleans, LA 70112-1393, United States
| | - Gin Chuang
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University, Health Sciences Center, New Orleans, LA 70112-1393, United States
| | - Huijing Xia
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University, Health Sciences Center, New Orleans, LA 70112-1393, United States; Cardiovascular Center of Excellence, Louisiana State University, Health Sciences Center, New Orleans, LA 70112-1393, United States
| | - Brendan Burn
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University, Health Sciences Center, New Orleans, LA 70112-1393, United States
| | - Jessica Bradley
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University, Health Sciences Center, New Orleans, LA 70112-1393, United States; Cardiovascular Center of Excellence, Louisiana State University, Health Sciences Center, New Orleans, LA 70112-1393, United States
| | - Jerome L Maderdrut
- Peptide Research Laboratory, Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112-2699, United States
| | - David H Coy
- Peptide Research Laboratory, Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112-2699, United States
| | - Kurt J Varner
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University, Health Sciences Center, New Orleans, LA 70112-1393, United States; Cardiovascular Center of Excellence, Louisiana State University, Health Sciences Center, New Orleans, LA 70112-1393, United States.
| |
Collapse
|
16
|
Kasica N, Podlasz P, Sundvik M, Tamas A, Reglodi D, Kaleczyc J. Protective Effects of Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) Against Oxidative Stress in Zebrafish Hair Cells. Neurotox Res 2016; 30:633-647. [PMID: 27557978 PMCID: PMC5047952 DOI: 10.1007/s12640-016-9659-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 07/26/2016] [Accepted: 08/09/2016] [Indexed: 12/30/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a pleiotropic neuropeptide, with known antiapoptotic functions. Our previous in vitro study has demonstrated the ameliorative role of PACAP-38 in chicken hair cells under oxidative stress conditions, but its effects on living hair cells is now yet known. Therefore, the aim of the present study was to investigate in vivo the protective role of PACAP-38 in hair cells found in zebrafish (Danio rerio) sense organs-neuromasts. To induce oxidative stress the 5-day postfertilization (dpf) zebrafish larvae were exposed to 1.5 mM H2O2 for 15 min or 1 h. This resulted in an increase in caspase-3 and p-38 MAPK level in the hair cells as well as in an impairment of the larvae basic behavior. To investigate the ameliorative role of PACAP-38, the larvae were incubated with a mixture of 1.5 mM H2O2 and 100 nM PACAP-38 following 1 h preincubation with 100 nM PACAP-38 only. PACAP-38 abilities to prevent hair cells from apoptosis were investigated. Whole-mount immunohistochemistry and confocal microscopy analyses revealed that PACAP-38 treatment decreased the cleaved caspase-3 level in the hair cells, but had no influence on p-38 MAPK. The analyses of basic locomotor activity supported the protective role of PACAP-38 by demonstrating the improvement of the fish behavior after PACAP-38 treatment. In summary, our in vivo findings demonstrate that PACAP-38 protects zebrafish hair cells from oxidative stress by attenuating oxidative stress-induced apoptosis.
Collapse
Affiliation(s)
- Natalia Kasica
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury, Oczapowskiego 13, box 105J, 10-719, Olsztyn, Poland.
| | - Piotr Podlasz
- Department of Pathophysiology, Forensic Veterinary and Administration, Faculty of Veterinary Medicine, University of Warmia and Mazury, Oczapowskiego 13, 10-719, Olsztyn, Poland
| | - Maria Sundvik
- Department of Anatomy, Neuroscience Center, University of Helsinki, Haartmaninkatu 8 (Biomedicum Helsinki), 00290, Helsinki, Finland
| | - Andrea Tamas
- Department of Anatomy, University of Pecs, Szigeti 12, 7624, Pecs, Hungary
| | - Dora Reglodi
- Department of Anatomy, University of Pecs, Szigeti 12, 7624, Pecs, Hungary
| | - Jerzy Kaleczyc
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury, Oczapowskiego 13, box 105J, 10-719, Olsztyn, Poland
| |
Collapse
|
17
|
Egri P, Fekete C, Dénes Á, Reglődi D, Hashimoto H, Fülöp BD, Gereben B. Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) Regulates the Hypothalamo-Pituitary-Thyroid (HPT) Axis via Type 2 Deiodinase in Male Mice. Endocrinology 2016; 157:2356-66. [PMID: 27046436 DOI: 10.1210/en.2016-1043] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The hypothalamic activation of thyroid hormones by type 2 deiodinase (D2), catalyzing the conversion of thyroxine to T3, is critical for the proper function of the hypothalamo-pituitary-thyroid (HPT) axis. Regulation of D2 expression in tanycytes alters the activity of the HPT axis. However, signals that regulate D2 expression in tanycytes are poorly understood. The pituitary adenylate cyclase-activating polypeptide (PACAP) increases intracellular cAMP level, a second messenger known to stimulate the DIO2 gene; however, its importance in tanycytes is not completely characterized. Therefore, we tested whether this ubiquitously expressed neuropeptide regulates the HPT axis through stimulation of D2 in tanycytes. PACAP increased the activity of human DIO2 promoter in luciferase reporter assay that was abolished by mutation of cAMP-response element. Furthermore, PAC1R receptor immunoreactivity was identified in hypothalamic tanycytes, suggesting that these D2-expressing cells could be regulated by PACAP. Intracerebroventricular PACAP administration resulted in increased D2 activity in the mediobasal hypothalamus, suppressed Trh expression in the hypothalamic paraventricular nucleus, and decreased Tshb expression in the pituitary demonstrating that PACAP affects the D2-mediated control of the HPT axis. To understand the role of endogenous PACAP in the regulation of HPT axis, the effect of decreased PACAP expression was studied in heterozygous Adcyap1 (PACAP) knockout mice. These animals were hypothyroid that may be the consequence of altered hypothalamic T3 degradation during set-point formation of the HPT axis. In conclusion, PACAP is an endogenous regulator of the HPT axis by affecting T3-mediated negative feedback via cAMP-induced D2 expression of tanycytes.
Collapse
Affiliation(s)
- P Egri
- Department of Endocrine Neurobiology (P.E., C.F., B.G.), Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest H-1083, Hungary; János Szentágothai PhD School of Neurosciences (P.E.), Semmelweis University, Budapest H-1085, Hungary; Department of Medicine (C.F.), Division of Endocrinology, Diabetes and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts 02111; Department of Gene Technology and Developmental Neurobiology (Á.D.), Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest H-1083, Hungary; Department of Anatomy (D.R., B.D.F.), University of Pécs Medical School, Pécs H-7624, Hungary; and Laboratory of Molecular Neuropharmacology (H.H.) and iPS Cell-Based Research Project on Brain Neuropharmacology and Toxicology (H.H.), Graduate School of Pharmaceutical Sciences, Osaka University, and Molecular Research Center for Children's Mental Development H.H.), United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Osaka 565-0871, Japan
| | - C Fekete
- Department of Endocrine Neurobiology (P.E., C.F., B.G.), Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest H-1083, Hungary; János Szentágothai PhD School of Neurosciences (P.E.), Semmelweis University, Budapest H-1085, Hungary; Department of Medicine (C.F.), Division of Endocrinology, Diabetes and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts 02111; Department of Gene Technology and Developmental Neurobiology (Á.D.), Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest H-1083, Hungary; Department of Anatomy (D.R., B.D.F.), University of Pécs Medical School, Pécs H-7624, Hungary; and Laboratory of Molecular Neuropharmacology (H.H.) and iPS Cell-Based Research Project on Brain Neuropharmacology and Toxicology (H.H.), Graduate School of Pharmaceutical Sciences, Osaka University, and Molecular Research Center for Children's Mental Development H.H.), United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Osaka 565-0871, Japan
| | - Á Dénes
- Department of Endocrine Neurobiology (P.E., C.F., B.G.), Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest H-1083, Hungary; János Szentágothai PhD School of Neurosciences (P.E.), Semmelweis University, Budapest H-1085, Hungary; Department of Medicine (C.F.), Division of Endocrinology, Diabetes and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts 02111; Department of Gene Technology and Developmental Neurobiology (Á.D.), Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest H-1083, Hungary; Department of Anatomy (D.R., B.D.F.), University of Pécs Medical School, Pécs H-7624, Hungary; and Laboratory of Molecular Neuropharmacology (H.H.) and iPS Cell-Based Research Project on Brain Neuropharmacology and Toxicology (H.H.), Graduate School of Pharmaceutical Sciences, Osaka University, and Molecular Research Center for Children's Mental Development H.H.), United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Osaka 565-0871, Japan
| | - D Reglődi
- Department of Endocrine Neurobiology (P.E., C.F., B.G.), Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest H-1083, Hungary; János Szentágothai PhD School of Neurosciences (P.E.), Semmelweis University, Budapest H-1085, Hungary; Department of Medicine (C.F.), Division of Endocrinology, Diabetes and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts 02111; Department of Gene Technology and Developmental Neurobiology (Á.D.), Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest H-1083, Hungary; Department of Anatomy (D.R., B.D.F.), University of Pécs Medical School, Pécs H-7624, Hungary; and Laboratory of Molecular Neuropharmacology (H.H.) and iPS Cell-Based Research Project on Brain Neuropharmacology and Toxicology (H.H.), Graduate School of Pharmaceutical Sciences, Osaka University, and Molecular Research Center for Children's Mental Development H.H.), United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Osaka 565-0871, Japan
| | - H Hashimoto
- Department of Endocrine Neurobiology (P.E., C.F., B.G.), Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest H-1083, Hungary; János Szentágothai PhD School of Neurosciences (P.E.), Semmelweis University, Budapest H-1085, Hungary; Department of Medicine (C.F.), Division of Endocrinology, Diabetes and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts 02111; Department of Gene Technology and Developmental Neurobiology (Á.D.), Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest H-1083, Hungary; Department of Anatomy (D.R., B.D.F.), University of Pécs Medical School, Pécs H-7624, Hungary; and Laboratory of Molecular Neuropharmacology (H.H.) and iPS Cell-Based Research Project on Brain Neuropharmacology and Toxicology (H.H.), Graduate School of Pharmaceutical Sciences, Osaka University, and Molecular Research Center for Children's Mental Development H.H.), United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Osaka 565-0871, Japan
| | - B D Fülöp
- Department of Endocrine Neurobiology (P.E., C.F., B.G.), Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest H-1083, Hungary; János Szentágothai PhD School of Neurosciences (P.E.), Semmelweis University, Budapest H-1085, Hungary; Department of Medicine (C.F.), Division of Endocrinology, Diabetes and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts 02111; Department of Gene Technology and Developmental Neurobiology (Á.D.), Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest H-1083, Hungary; Department of Anatomy (D.R., B.D.F.), University of Pécs Medical School, Pécs H-7624, Hungary; and Laboratory of Molecular Neuropharmacology (H.H.) and iPS Cell-Based Research Project on Brain Neuropharmacology and Toxicology (H.H.), Graduate School of Pharmaceutical Sciences, Osaka University, and Molecular Research Center for Children's Mental Development H.H.), United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Osaka 565-0871, Japan
| | - Balázs Gereben
- Department of Endocrine Neurobiology (P.E., C.F., B.G.), Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest H-1083, Hungary; János Szentágothai PhD School of Neurosciences (P.E.), Semmelweis University, Budapest H-1085, Hungary; Department of Medicine (C.F.), Division of Endocrinology, Diabetes and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts 02111; Department of Gene Technology and Developmental Neurobiology (Á.D.), Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest H-1083, Hungary; Department of Anatomy (D.R., B.D.F.), University of Pécs Medical School, Pécs H-7624, Hungary; and Laboratory of Molecular Neuropharmacology (H.H.) and iPS Cell-Based Research Project on Brain Neuropharmacology and Toxicology (H.H.), Graduate School of Pharmaceutical Sciences, Osaka University, and Molecular Research Center for Children's Mental Development H.H.), United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Osaka 565-0871, Japan
| |
Collapse
|
18
|
Guo X, Yu R, Xu Y, Lian R, Yu Y, Cui Z, Ji Q, Chen J, Li Z, Liu H, Chen J. PAC1R agonist maxadilan enhances hADSC viability and neural differentiation potential. J Cell Mol Med 2016; 20:874-90. [PMID: 26798992 PMCID: PMC4831362 DOI: 10.1111/jcmm.12772] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Accepted: 12/01/2015] [Indexed: 12/18/2022] Open
Abstract
Pituitary adenylate cyclase‐activating polypeptide (PACAP) is a structurally endogenous peptide with many biological roles. However, little is known about its presence or effects in human adipose‐derived stem cells (hADSCs). In this study, the expression of PACAP type I receptor (PAC1R) was first confirmed in hADSCs. Maxadilan, a specific agonist of PAC1R, could increase hADSC proliferation as determined by Cell Counting Kit‐8 and cell cycle analysis and promote migration as shown in wound‐healing assays. Maxadilan also showed anti‐apoptotic activity in hADSCs against serum withdrawal‐induced apoptosis based on Annexin V/propidium iodide analysis and mitochondrial membrane potential assays. The anti‐apoptotic effects of maxadilan correlated with the down‐regulation of Cleaved Caspase 3 and Caspase 9 as well as up‐regulation of Bcl‐2. The chemical neural differentiation potential could be enhanced by maxadilan as indicated through quantitative PCR, Western blot and cell morphology analysis. Moreover, cytokine neural redifferentiation of hADSCs treated with maxadilan acquired stronger neuron‐like functions with higher voltage‐dependent tetrodotoxin‐sensitive sodium currents, higher outward potassium currents and partial electrical impulses as determined using whole‐cell patch clamp recordings. Maxadilan up‐regulated the Wnt/β‐catenin signalling pathway associated with dimer‐dependent activity of PAC1R, promoting cell viability that was inhibited by XAV939, and it also activated the protein kinase A (PKA) signalling pathway associated with ligand‐dependent activity of PAC1R, enhancing cell viability and neural differentiation potential that was inhibited by H‐89. In summary, these results demonstrated that PAC1R is present in hADSCs, and maxadilan could enhance hADSC viability and neural differentiation potential in neural differentiation medium.
Collapse
Affiliation(s)
- Xiaoling Guo
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
| | - Rongjie Yu
- Department of Cell Biology, Jinan University, Guangzhou, China
| | - Ying Xu
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Ruiling Lian
- Department of Ophthalmology, The First Clinical Medical College of Jinan University, Guangzhou, China
| | - Yankun Yu
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Zekai Cui
- Department of Cell Biology, Jinan University, Guangzhou, China
| | - Qingshan Ji
- Department of Ophthalmology, Affiliated Anhui Provincial Hospital of Anhui Medical University, Hefei, China
| | - Junhe Chen
- Department of Mathematics, South China University of Technology, Guangzhou, China
| | - Zhijie Li
- Eye Institute, Medical College of Jinan University, Guangzhou, China
| | - Hongwei Liu
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
| | - Jiansu Chen
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China.,Department of Ophthalmology, The First Clinical Medical College of Jinan University, Guangzhou, China.,Eye Institute, Medical College of Jinan University, Guangzhou, China
| |
Collapse
|
19
|
Banki E, Pakai E, Gaszner B, Zsiboras C, Czett A, Bhuddi PRP, Hashimoto H, Toth G, Tamas A, Reglodi D, Garami A. Characterization of the thermoregulatory response to pituitary adenylate cyclase-activating polypeptide in rodents. J Mol Neurosci 2014; 54:543-54. [PMID: 24994541 DOI: 10.1007/s12031-014-0361-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 06/23/2014] [Indexed: 12/19/2022]
Abstract
Administration of the long form (38 amino acids) of pituitary adenylate cyclase-activating polypeptide (PACAP38) into the central nervous system causes hyperthermia, suggesting that PACAP38 plays a role in the regulation of deep body temperature (T b). In this study, we investigated the thermoregulatory role of PACAP38 in details. First, we infused PACAP38 intracerebroventricularly to rats and measured their T b and autonomic thermoeffector responses. We found that central PACAP38 infusion caused dose-dependent hyperthermia, which was brought about by increased thermogenesis and tail skin vasoconstriction. Compared to intracerebroventricular administration, systemic (intravenous) infusion of the same dose of PACAP38 caused significantly smaller hyperthermia, indicating a central site of action. We then investigated the thermoregulatory phenotype of mice lacking the Pacap gene (Pacap (-/-)). Freely moving Pacap (-/-) mice had higher locomotor activity throughout the day and elevated deep T b during the light phase. When the Pacap (-/-) mice were loosely restrained, their metabolic rate and T b were lower compared to their wild-type littermates. We conclude that PACAP38 causes hyperthermia via activation of the autonomic cold-defense thermoeffectors through central targets. Pacap (-/-) mice express hyperkinesis, which is presumably a compensatory mechanism, because under restrained conditions, these mice are hypometabolic and hypothermic compared to controls.
Collapse
Affiliation(s)
- Eszter Banki
- Department of Anatomy PTE-MTA "Lendulet" PACAP Research Team, Medical School, University of Pecs, Pecs, Hungary
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Koga M, Mizuno Y, Watanabe I, Kawakami H, Goto T. Role of VPAC2 receptor in monocrotaline-induced pulmonary hypertension in rats. J Appl Physiol (1985) 2014; 117:383-91. [PMID: 24947028 DOI: 10.1152/japplphysiol.00861.2013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Pulmonary hypertension (PH) is associated with significant morbidity and mortality. Vasoactive intestinal peptide (VIP) and pituitary adenylyl cyclase activating peptide (PACAP) have pulmonary vasodilatory and positive inotropic effects via receptors VPAC1 and VPAC2, which possess a similar affinity for both peptides, and PAC1, a PACAP-preferring receptor. VIP is a promising option for PH treatment; however, various physiological effects of VIP have limited its clinical use. We investigated the effects of VPAC1 and VPAC2 selective agonists VIP and PACAP to explore more appropriate means of treatment for PH. We examined hemodynamic changes in right ventricular systolic pressure (RVSP), systemic blood pressure (SBP), total pulmonary resistance index (TPRI), total systemic resistance index, and cardiac index (CI) in response to their agonists with monocrotaline (MCT)-induced PH and explored involvement of VIP/PACAP expression and receptors in PH. Sprague-Dawley rats were divided into the MCT group (administered MCT 60 mg/kg) and control group. In MCT-induced PH, decreased VIP and PACAP were associated with upregulation of VPAC1, VPAC2, and PAC1 in lung tissues. Intravenous injection of VPAC2-selective agonist BAY 55-9837 and VIP, but not [Ala(11,22,28)]VIP, improved the CI. The decrease in SBP with VPAC2 agonist was significantly less than that in the control. Although they decreased SBP, these agonists hardly affected RVSP in the control. Activation of VPAC2 receptor with BAY 55-9837 effectively improved RVSP, TPRI, and CI in MCT-induced PH, suggesting a VPAC2 agonist as a possible promising treatment for PH.
Collapse
Affiliation(s)
- Motokazu Koga
- Department of Anesthesiology and Critical Care Medicine, Division of Bio-Functional Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yusuke Mizuno
- Department of Anesthesiology and Critical Care Medicine, Division of Bio-Functional Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Itaru Watanabe
- Department of Anesthesiology and Critical Care Medicine, Division of Bio-Functional Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hiromasa Kawakami
- Department of Anesthesiology and Critical Care Medicine, Division of Bio-Functional Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takahisa Goto
- Department of Anesthesiology and Critical Care Medicine, Division of Bio-Functional Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
21
|
PACAP38 suppresses cortical damage in mice with traumatic brain injury by enhancing antioxidant activity. J Mol Neurosci 2014; 54:370-9. [PMID: 24907941 DOI: 10.1007/s12031-014-0309-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 04/14/2014] [Indexed: 10/25/2022]
Abstract
The production of reactive oxygen species (ROS) and the resulting oxidative stress in mice in response to a controlled cortical impact (CCI) are typical exacerbating factors associated with traumatic brain injury (TBI). Pituitary adenylate cyclase-activating polypeptide 38 (PACAP38) is a multifunctional peptide that has been shown to exhibit neuroprotective effects in response to a diverse range of injuries to neuronal cells. We recently reported that PACAP38 might regulate oxidative stress in mice. The aim of the present study was to determine whether PACAP38 exerts neuroprotective effects by regulating oxidative stress in mice with TBI. Reactive oxidative metabolites (ROMs) and biological antioxidant potential (BAP) were measured in male C57Bl/6 mice before and 3, 4, and 24 h after CCI. PACAP38 was administered intravenously immediately following CCI, and immunostaining for the oxidative stress indicator nitrotyrosine (NT), and for neuronal death as an indicator of the area affected by TBI, was measured 24 h later. Western blot experiments to determine antioxidant activity [as indicated by superoxide dismutase-2 (SOD-2) and glutathione peroxidase 1 (GPx-1)] in the neocortical region were also performed 3 h post-CCI. Results showed that plasma BAP and ROM levels were dramatically increased 3 h after CCI. PACAP38 suppressed the extent of TBI and NT-positive regions 24 h after CCI, and increased SOD-2 and GPx-1 levels in both hemispheres. Taken together, these results suggest that increasing antioxidant might be involving in the neuroprotective effect of PACAP38 in mice subjected to a CCI.
Collapse
|
22
|
Horvath G, Reglodi D, Brubel R, Halasz M, Barakonyi A, Tamas A, Fabian E, Opper B, Toth G, Cohen M, Szereday L. Investigation of the possible functions of PACAP in human trophoblast cells. J Mol Neurosci 2014; 54:320-30. [PMID: 24874580 DOI: 10.1007/s12031-014-0337-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 05/16/2014] [Indexed: 12/21/2022]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) is an endogenous neuropeptide having a widespread distribution both in the nervous system and peripheral organs including the female reproductive system. Both the peptide and its receptors have been shown in the placenta but its role in placental growth, especially its human aspects, remains unknown. The aim of the present study was to investigate the effects of PACAP on invasion, proliferation, cell survival, and angiogenesis of trophoblast cells. Furthermore, cytokine production was investigated in human decidual and peripheral blood mononuclear cells. For in vitro studies, human invasive proliferative extravillous cytotrophoblast (HIPEC) cells and HTR-8/SVneo human trophoblast cells were used. Both cell types were used for testing the effects of PACAP on invasion and cell survival in order to investigate whether the effects of PACAP in trophoblasts depend on the examined cell type. Invasion was studied by standardized invasion assay. PACAP increased proliferation in HIPEC cells, but not in HTR-8 cells. Cell viability was examined using MTT test, WST-1 assay, and annexin V/propidium iodide flow cytometry assay. Survival of HTR-8/SVneo cells was studied under oxidative stress conditions induced by hydrogen peroxide. PACAP as pretreatment, but not as co-treatment, significantly increased the number of surviving HTR-8 cells. Viability of HIPEC cells was investigated using methotrexate (MTX) toxicity, but PACAP1-38 could not counteract its toxic effect. Angiogenic molecules were determined both in the supernatant and the cell lysate by angiogenesis array. In the supernatant, we found that PACAP decreased the secretion of various angiogenic markers, such as angiopoietin, angiogenin, activin, endoglin, ADAMTS-1, and VEGF. For the cytokine assay, human decidual and peripheral blood lymphocytes were separated and treated with PACAP1-38. Th1 and Th2 cytokines were analyzed with CBA assay and the results showed that there were no significant differences in control and PACAP-treated cells. In summary, PACAP seems to play various roles in human trophoblast cells, depending on the cell type and microenvironmental influences.
Collapse
Affiliation(s)
- G Horvath
- Department of Anatomy, MTA-PTE "Lendulet" PACAP Research Team, University of Pecs, Szigeti u 12, 7624, Pecs, Hungary
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Wang G, Dai L, Luo L, Xu W, Zhang C, Zhu Y, Chen Z, Hu W, Xu X, Pan W. Non-essential amino acids attenuate apoptosis of gastric cancer cells induced by glucose starvation. Oncol Rep 2014; 32:332-40. [PMID: 24858809 DOI: 10.3892/or.2014.3205] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 04/23/2014] [Indexed: 11/06/2022] Open
Abstract
Energy and nutrition are essential requirements for all living cells, including cancer cells. In the initiating stage of cancer in organs, cancer cells grow fast and have inadequate supplies of glucose, oxygen and other nutrients due to deficient angiogenesis. Anaerobic conditions cause cancer cells to rely on glycolysis, which produces pyruvate and ATP that can be used by cancer cells to survive. However, glucose starvation may result in apoptosis or necrosis of cancer cells. It has been reported that autophagy is a consequence of glucose starvation and that amino acids are products of autophagy. The present study investigated whether amino acids may represent an alternative energy source for cancer cells undergoing glucose starvation. With non-essential amino acids, growth inhibition and apoptosis of gastric cancer cells induced by glucose starvation were attenuated compared with that of cells undergoing glucose starvation without amino acids, as measured by cell viability, apoptosis rates, membrane potential of mitochondria, and apoptosis-related genes. Meanwhile, both mitochondrial DNA copy number and amino acid transporter genes were increased compared with those in control cells. Non-essential amino acids prevented gastric cancer cells from glucose starvation-induced apoptosis.
Collapse
Affiliation(s)
- Gang Wang
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Lei Dai
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Laisheng Luo
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Wen Xu
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Chenjing Zhang
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Yimiao Zhu
- Department of Gastroenterology, The Second Affiliated Hospital Binjiang Campus, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Zhongting Chen
- Department of Pharmacy, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Wen Hu
- Department of Gastroenterology, The Second Affiliated Hospital Binjiang Campus, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Xiang Xu
- Department of Pharmacy, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Wensheng Pan
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| |
Collapse
|
24
|
Tanaka S, Miyagi T, Dohi E, Seki T, Hide I, Sotomaru Y, Saeki Y, Antonio Chiocca E, Matsumoto M, Sakai N. Developmental expression of GPR3 in rodent cerebellar granule neurons is associated with cell survival and protects neurons from various apoptotic stimuli. Neurobiol Dis 2014; 68:215-27. [PMID: 24769160 DOI: 10.1016/j.nbd.2014.04.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 03/08/2014] [Accepted: 04/15/2014] [Indexed: 11/15/2022] Open
Abstract
G-protein coupled receptor 3 (GPR3), GPR6, and GPR12 belong to a family of constitutively active Gs-coupled receptors that activate 3'-5'-cyclic adenosine monophosphate (cAMP) and are highly expressed in the brain. Among these receptors, the endogenous expression of GPR3 in cerebellar granule neurons (CGNs) is increased following development. GPR3 is important for neurite outgrowth and neural maturation; however, the physiological functions of GPR3 remain to be fully elucidated. Here, we investigated the survival and antiapoptotic functions of GPR3 under normal and apoptosis-inducing culture conditions. Under normal culture conditions, CGNs from GPR3-knockout mice demonstrated lower survival than did CGNs from wild-type or GPR3-heterozygous mice. Cerebellar sections from GPR3-/- mice at P7, P14, and P21 revealed more caspase-3-positive neurons in the internal granular layer than in cerebellar sections from wild-type mice. Conversely, in a potassium-deprivation model of apoptosis, increased expression of these three receptors promoted neuronal survival. The antiapoptotic effect of GPR3 was also observed under hypoxic (1% O2/5% CO2) and reactive oxygen species (ROS)-induced apoptotic conditions. We further investigated the signaling pathways involved in the GPR3-mediated antiapoptotic effect. The addition of the PKA inhibitor KT5720, the MAP kinase inhibitor U0126, and the PI3 kinase inhibitor LY294002 abrogated the GPR3-mediated antiapoptotic effect in a potassium-deprivation model of apoptosis, whereas the PKC inhibitor Gö6976 did not affect the antiapoptotic function of GPR3. Furthermore, downregulation of endogenous GPR3 expression in CGNs resulted in a marked reduction in the basal levels of ERK and Akt phosphorylation under normal culture conditions. Finally, we used a transient middle cerebral artery occlusion (tMCAO) model in wild-type and GPR3-knockout mice to determine whether GPR3 expression modulates neuronal survival after brain ischemia. After tMCAO, GPR3-knockout mice exhibited a significantly larger infarct area than did wild-type mice. Collectively, these in vitro and in vivo results suggest that the developmental expression of constitutively active Gs-coupled GPR3 activates the ERK and Akt signaling pathways at the basal level, thereby protecting neurons from apoptosis that is induced by various stimuli.
Collapse
Affiliation(s)
- Shigeru Tanaka
- Department of Molecular and Pharmacological Neuroscience, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; Department of Clinical Neuroscience and Therapeutics, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan.
| | - Tatsuhiro Miyagi
- Department of Molecular and Pharmacological Neuroscience, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Eisuke Dohi
- Department of Clinical Neuroscience and Therapeutics, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Takahiro Seki
- Department of Molecular and Pharmacological Neuroscience, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Izumi Hide
- Department of Molecular and Pharmacological Neuroscience, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Yusuke Sotomaru
- Natural Science Center for Basic Research and Development, Hiroshima University, Hiroshima 734-8551, Japan
| | | | - E Antonio Chiocca
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Institute for the Neurosciences at the Brigham, Brigham and Women's/Faulkner Hospital and Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Masayasu Matsumoto
- Department of Clinical Neuroscience and Therapeutics, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Norio Sakai
- Department of Molecular and Pharmacological Neuroscience, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| |
Collapse
|
25
|
Pituitary adenylate cyclase activating polypeptide (PACAP) signalling exerts chondrogenesis promoting and protecting effects: implication of calcineurin as a downstream target. PLoS One 2014; 9:e91541. [PMID: 24643018 PMCID: PMC3958376 DOI: 10.1371/journal.pone.0091541] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 02/13/2014] [Indexed: 01/20/2023] Open
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) is an important neurotrophic factor influencing differentiation of neuronal elements and exerting protecting role during traumatic injuries or inflammatory processes of the central nervous system. Although increasing evidence is available on its presence and protecting function in various peripheral tissues, little is known about the role of PACAP in formation of skeletal components. To this end, we aimed to map elements of PACAP signalling in developing cartilage under physiological conditions and during oxidative stress. mRNAs of PACAP and its receptors (PAC1,VPAC1, VPAC2) were detectable during differentiation of chicken limb bud-derived chondrogenic cells in micromass cell cultures. Expression of PAC1 protein showed a peak on days of final commitment of chondrogenic cells. Administration of either the PAC1 receptor agonist PACAP 1-38, or PACAP 6-38 that is generally used as a PAC1 antagonist, augmented cartilage formation, stimulated cell proliferation and enhanced PAC1 and Sox9 protein expression. Both variants of PACAP elevated the protein expression and activity of the Ca-calmodulin dependent Ser/Thr protein phosphatase calcineurin. Application of PACAPs failed to rescue cartilage formation when the activity of calcineurin was pharmacologically inhibited with cyclosporine A. Moreover, exogenous PACAPs prevented diminishing of cartilage formation and decrease of calcineurin activity during oxidative stress. As an unexpected phenomenon, PACAP 6-38 elicited similar effects to those of PACAP 1-38, although to a different extent. On the basis of the above results, we propose calcineurin as a downstream target of PACAP signalling in differentiating chondrocytes either in normal or pathophysiological conditions. Our observations imply the therapeutical perspective that PACAP can be applied as a natural agent that may have protecting effect during joint inflammation and/or may promote cartilage regeneration during degenerative diseases of articular cartilage.
Collapse
|
26
|
Nemeth A, Szabadfi K, Fulop B, Reglodi D, Kiss P, Farkas J, Szalontai B, Gabriel R, Hashimoto H, Tamas A. Examination of calcium-binding protein expression in the inner ear of wild-type, heterozygous and homozygous pituitary adenylate cyclase-activating polypeptide (PACAP)-knockout mice in kanamycin-induced ototoxicity. Neurotox Res 2013; 25:57-67. [PMID: 24155155 DOI: 10.1007/s12640-013-9428-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 09/23/2013] [Accepted: 09/24/2013] [Indexed: 11/24/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide with diverse biological effects. It also occurs and exerts protective effects in sensory organs; however, little is known about its effects in the auditory system. Recently, we have shown that PACAP protects cochlear cells against oxidative-stress-induced apoptosis and homozygous PACAP-deficient animals show stronger expression of Ca(2+)-binding proteins in the hair cells of the inner ear, but there are no data about the consequences of the lack of endogenous PACAP in different ototoxic insults such as aminoglycoside-induced toxicity. In this study, we examined the effect of kanamycin treatment on Ca(2+)-binding protein expression in hair cells of wild-type, heterozygous and homozygous PACAP-deficient mice. We treated 5-day-old mice with kanamycin, and 2 days later, we examined the Ca(2+)-binding protein expression of the hair cells with immunohistochemistry. We found stronger expression of Ca(2+)-binding proteins in the hair cells of control heterozygous and homozygous PACAP-deficient mice compared with wild-type animals. Kanamycin induced a significant increase in Ca(2+)-binding protein expression in wild-type and heterozygous PACAP-deficient mice, but the baseline higher expression in homozygous PACAP-deficient mice did not show further changes after the treatment. Elevated endolymphatic Ca(2+) is deleterious for the cochlear function, against which the high concentration of Ca(2+)-buffers in hair cells may protect. Meanwhile, the increased immunoreactivity of Ca(2+)-binding proteins in the absence of PACAP provide further evidence for the important protective role of PACAP in ototoxicity, but further investigations are necessary to examine the exact role of endogenous PACAP in ototoxic insults.
Collapse
Affiliation(s)
- A Nemeth
- Department of Oto-rhino-laryngology, University of Pecs, Pecs, Hungary
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Nedvig K, Weber G, Nemeth J, Kovacs K, Reglodi D, Kemeny A, Ferencz A. Changes of PACAP immunoreactivities and cytokine levels after PACAP-38 containing intestinal preservation and autotransplantation. J Mol Neurosci 2012; 48:788-94. [PMID: 22899163 DOI: 10.1007/s12031-012-9870-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 07/31/2012] [Indexed: 01/28/2023]
Abstract
Small bowel is one of the most sensitive organs to ischemia-reperfusion injury, which is a significant problem during transplantation. Pituitary adenylate cyclase-activating polypeptide (PACAP) has cytoprotective effect in ischemic injuries of various tissues. The aim of our study was to measure changes of PACAP-38 and PACAP-27 immunoreactivities and cytokine levels in intestinal grafts stored in PACAP-38-containing preservation solution. Small bowel autotransplantation was performed on male Wistar rats. Grafts were stored in University of Wisconsin (UW) solution at 4 °C for 1 h (group (G)I), for 3 h (GII), and for 6 h (GIII) and in PACAP-38-containing UW solution for 1 h (GIV), for 3 h (GV), and for 6 h (GVI). After preservation, performing vessel anastomosis reperfusion began, which lasted 3 h in each group. Tissue biopsies were collected after laparotomy (control) and at the end of the reperfusion periods. Intestinal PACAP-38 and PACAP-27 immunoreactivities were measured by radioimmunoassay. To measure cytokines from tissue homogenates, we used rat cytokine array and Luminex Multiplex Immunoassay. Levels of PACAP-38 and PACAP-27 immunoreactivity decreased after 1 and 3 h preservation compared to control levels. This decrease was significant following 6 h cold storage (p < 0.05). Values remained significantly higher in grafts stored in PACAP-38-containing UW. Cytokine array revealed that expression of the soluble intercellular adhesion molecule-1 (CD54) and L-selectin (CD62L/LECAM-1) was increased in GIII. Both 6 h cold storage in PACAP-38-containing UW solution and 3 h reperfusion caused strong reduction in these cytokines activation in GVI. RANTES (CCL5) levels were increased in all groups. Strong activation of the tissue inhibitor of metalloproteinase-1 was in GIII. However, PACAP-38-containing cold storage could decrease its activation in GVI. Furthermore, strong activation of the tissue inhibitor of metalloproteinase-1 was detected in 6 h preserved grafts without PACAP-38 (GIII). PACAP-38-containing cold storage could decrease its activation in GVI. Our present study showed that PACAP-38 and PACAP-27 immunoreactivities decreased in a time-dependent manner during intestinal cold preservation, which could be ameliorated by administration of exogenous PACAP-38 to the preservation solution. Moreover, PACAP-38 could attenuate tissue cold ischemic injury-induced changes in cytokine expression.
Collapse
Affiliation(s)
- Klara Nedvig
- Department of Surgical Research and Techniques, University of Pecs, Pecs, Hungary
| | | | | | | | | | | | | |
Collapse
|
28
|
Szanto Z, Sarszegi Z, Reglodi D, Nemeth J, Szabadfi K, Kiss P, Varga A, Banki E, Csanaky K, Gaszner B, Pinter O, Szalai Z, Tamas A. PACAP immunoreactivity in human malignant tumor samples and cardiac diseases. J Mol Neurosci 2012; 48:667-73. [PMID: 22648511 DOI: 10.1007/s12031-012-9815-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 05/15/2012] [Indexed: 01/21/2023]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) is a pleiotropic and multifunctional neuropeptide having important roles in various physiological processes. Recent trends in PACAP research point to the clinical introduction of PACAP or its analogs/fragments possibly in the near future. Recently, we have shown the presence of PACAP in human plasma, milk, placenta, and follicular fluid samples. However, relatively few data are available on PACAP in human tissues from patients with different disorders. The aim of the present study was to determine, by radioimmunoassay, the tissue level of PACAP38-like immunoreactivity (LI) and PACAP27-LI in different primary non-small cell lung cancer, colon tumor samples, and in cardiac muscle samples from patients suffering from ischemic heart disease and valvular disorders. We also labeled the PAC1 receptors in human cardiac cells. All samples showed significantly higher PACAP38-LI compared with PACAP27-LI. We found significantly lower levels of PACAP38-LI and PACAP27-LI in tumoral and peripheral samples compared with normal healthy tissue in both lung and colon cancers. Further investigations are necessary to describe the exact function of PACAP in oncogenesis. We showed that PACAP38-LI and PACAP27-LI are significantly higher in ischemic heart diseases compared with valvular abnormalities, suggesting that PACAP might play a role in ischemic heart disorders.
Collapse
Affiliation(s)
- Z Szanto
- Surgery Clinic, University of Pecs, Pecs, Hungary
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
PACAP is an Endogenous Protective Factor—Insights from PACAP-Deficient Mice. J Mol Neurosci 2012; 48:482-92. [DOI: 10.1007/s12031-012-9762-0] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Accepted: 03/22/2012] [Indexed: 01/07/2023]
|
30
|
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. [PMID: 21621841 DOI: 10.1016/j.npep.2011.05.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Accepted: 05/03/2011] [Indexed: 10/18/2022]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) is a widespread neuropeptide with diverse effects in the nervous system and peripheral organs. One of the most well-studied effects of PACAP is its cytoprotective action, against different harmful stimuli in a wide variety of cells and tissues. PACAP occurs in the urinary system, from the kidney to the lower urinary tract. The present review focuses on the nephroprotective effects of PACAP and summarizes data obtained regarding the protective effects of PACAP in different models of kidney pathologies. In vitro data show that PACAP protects tubular cells against oxidative stress, myeloma light chain, cisplatin, cyclosporine-A and hypoxia. In vivo data provide evidence for its protective effects in ischemia/reperfusion, cisplatin, cyclosporine-A, myeloma kidney injury, diabetic nephropathy and gentamicin-induced kidney damage. Results accumulated on the renoprotective effects of PACAP suggest that PACAP is an emerging candidate for treatment of human kidney pathologies.
Collapse
Affiliation(s)
- Dora Reglodi
- Department of Anatomy, University of Pecs, Szigeti u 12, 7624 Pecs, Hungary.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Zhao Z, Yu R, Yang J, Liu X, Tan M, Li H, Chen J. Maxadilan prevents apoptosis in iPS cells and shows no effects on the pluripotent state or karyotype. PLoS One 2012; 7:e33953. [PMID: 22457805 PMCID: PMC3311553 DOI: 10.1371/journal.pone.0033953] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Accepted: 02/20/2012] [Indexed: 11/19/2022] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a structurally endogenous peptide with many biological roles. Maxadilan, a 61-amino acid vasodilatory peptide, specifically activates the PACAP type I receptor (PAC1). Although PAC1 has been identified in embryonic stem cells, little is known about its presence or effects in human induced pluripotent stem (iPS) cells. In the present study, we investigated the expression of PAC1 in human iPS cells by reverse transcriptase polymerase chain reaction (RT-PCR) and western blot analysis. To study the physiological effects mediated by PAC1, we evaluated the role of maxadilan in preventing apoptotic cell death induced by ultraviolet C (UVC). After exposure to UVC, the iPS cells showed a marked reduction in cell viability and a parallel increase of apoptotic cells, as demonstrated by WST-8 analysis, annexin V/propidium iodide (PI) analysis and the terminal transferase dUTP nick end labeling (TUNEL) assay. The addition of 30 nM of maxadilan dramatically increased iPS cell viability and reduced the percentage of apoptotic cells. The anti-apoptotic effects of maxadilan were correlated to the downregulation of caspase-3 and caspase-9. Concomitantly, immunofluorescence, western blot analysis, real-time quantitative polymerase chain reaction (RT-qPCR) analysis and in vitro differentiation results showed that maxadilan did not affect the pluripotent state of iPS cells. Moreover, karyotype analysis showed that maxadilan did not affect the karyotype of iPS cells. In summary, these results demonstrate that PAC1 is present in iPS cells and that maxadilan effectively protects iPS cells against UVC-induced apoptotic cell death while not affecting the pluripotent state or karyotype.
Collapse
Affiliation(s)
- Zhiyi Zhao
- Ophthalmology Department, First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of ophthalmology, Medical College, Jinan University, Guangzhou, China
- Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - Rongjie Yu
- Bio-engineering Institute of Jinan University, Jinan University, Guangzhou, China
| | - Jiayin Yang
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xiaofei Liu
- Bio-engineering Institute of Jinan University, Jinan University, Guangzhou, China
| | - Meihua Tan
- Ophthalmology Department, First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of ophthalmology, Medical College, Jinan University, Guangzhou, China
- Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - HongYang Li
- Ophthalmology Department, First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of ophthalmology, Medical College, Jinan University, Guangzhou, China
- Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - Jiansu Chen
- Ophthalmology Department, First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of ophthalmology, Medical College, Jinan University, Guangzhou, China
- Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
- * E-mail:
| |
Collapse
|
32
|
Tamas A, Szabadfi K, Nemeth A, Fulop B, Kiss P, Atlasz T, Gabriel R, Hashimoto H, Baba A, Shintani N, Helyes Z, Reglodi D. Comparative Examination of Inner Ear in Wild Type and Pituitary Adenylate Cyclase Activating Polypeptide (PACAP)-Deficient Mice. Neurotox Res 2011; 21:435-44. [DOI: 10.1007/s12640-011-9298-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2011] [Revised: 11/17/2011] [Accepted: 12/06/2011] [Indexed: 12/30/2022]
|
33
|
Lorentz CU, Woodward WR, Tharp K, Habecker BA. Altered norepinephrine content and ventricular function in p75NTR-/- mice after myocardial infarction. Auton Neurosci 2011; 164:13-9. [PMID: 21646052 PMCID: PMC3167025 DOI: 10.1016/j.autneu.2011.05.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Revised: 05/09/2011] [Accepted: 05/11/2011] [Indexed: 01/08/2023]
Abstract
Cardiac sympathetic neurons stimulate heart rate and the force of contraction through release of norepinephrine. Nerve growth factor modulates sympathetic transmission through activation of TrkA and p75NTR. Nerve growth factor plays an important role in post-infarct sympathetic remodeling. We used mice lacking p75NTR to examine the effect of altered nerve growth factor signaling on sympathetic neuropeptide expression, cardiac norepinephrine, and ventricular function after myocardial infarction. Infarct size was similar in wildtype and p75NTR-/- mice after ischemia-reperfusion surgery. Likewise, mRNAs encoding vasoactive intestinal peptide, galanin, and pituitary adenylate cyclase activating peptides were identical in wildtype and p75NTR-/- cardiac sympathetic neurons, as was expression of the TrkA neurotrophin receptor. Norepinephrine content was elevated in the base of the p75NTR-/- ventricle compared to wildtype, but levels were identical below the site of occlusion. Left ventricular pressure, dP/dt(MAX), and dP/dt(MIN) were measured under isoflurane anesthesia 3 and 7 days after surgery. Ventricular pressure decreased significantly 3 days after infarction, and deficits in dP/dt(MAX) were revealed by stimulating beta receptors with dobutamine and release of endogenous norepinephrine with tyramine. dP/dt(MIN) was not altered by genotype or surgical group. Few differences were observed between genotypes 3 days after surgery, in contrast to low pressure and dP/dt(MAX) previously reported in control p75NTR-/- animals. Seven days after surgery ventricular pressure and dP/dt(MAX) were significantly lower in p75NTR-/- hearts compared to WT hearts. Thus, the lack of p75NTR did not enhance cardiac function after myocardial infarction.
Collapse
Affiliation(s)
- Christina U. Lorentz
- Department of Physiology & Pharmacology, Oregon Health & Science University, Portland, OR 97239, USA
| | - William R. Woodward
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Kevin Tharp
- Department of Physiology & Pharmacology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Beth A. Habecker
- Department of Physiology & Pharmacology, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
34
|
Alston EN, Parrish DC, Hasan W, Tharp K, Pahlmeyer L, Habecker BA. Cardiac ischemia-reperfusion regulates sympathetic neuropeptide expression through gp130-dependent and independent mechanisms. Neuropeptides 2011; 45:33-42. [PMID: 21035185 PMCID: PMC3053070 DOI: 10.1016/j.npep.2010.10.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2010] [Revised: 10/04/2010] [Accepted: 10/04/2010] [Indexed: 11/23/2022]
Abstract
Cardiac function is regulated by a balance of sympathetic and parasympathetic transmission. Neuropeptide Y (NPY) and galanin (GAL) released from cardiac sympathetic neurons inhibits parasympathetic transmission in the heart. Sympathetic peptides may contribute to autonomic imbalance, which is characterized by increased sympathetic and decreased parasympathetic transmission and contributes to life threatening cardiovascular pathologies. Several gp130 cytokines are increased in the heart after myocardial infarction (MI), and these cytokines stimulate neuropeptide expression in sympathetic neurons. We used mice whose sympathetic neurons lack the gp130 receptor (gp130(DBH-Cre/lox) mice) to ask if cytokine activation of gp130 regulated neuropeptide expression in cardiac sympathetic nerves after MI. Myocardial infarction decreased NPY mRNA through a gp130 independent mechanism and increased VIP and PACAP mRNA via gp130, while GAL mRNA was unchanged. Immunohistochemistry revealed a gp130-dependent increase in PACAP38 in cells of the stellate ganglion after MI, and PACAP was detected in pre-ganglionic fibers of all genotypes and surgical groups. VIP was identified in a few sympathetic nerve fibers in all genotypes and surgical groups. GAL and PACAP38 were not detected in sham hearts, but peptide immunoreactivity was high in the infarct three days after MI. Surprisingly, peptides were abundant in cells that co-labeled with macrophage markers F4/80 and MAC2, but were not detected in sympathetic axons. PACAP protects cardiac myocytes from apoptosis, and GAL stimulates axon regeneration in addition to inhibiting parasympathetic transmission. Thus, these peptides may play an important role in cardiac and neuronal remodeling after ischemia-reperfusion.
Collapse
Affiliation(s)
| | | | | | | | | | - Beth A. Habecker
- Address correspondence to: Beth A. Habecker, Ph.D., Dept. of Physiology & Pharmacology, L334, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, TEL: (503) 494-0497, FAX: (503) 494-4352,
| |
Collapse
|
35
|
PACAP protects against TNFα-induced cell death in olfactory epithelium and olfactory placodal cell lines. Mol Cell Neurosci 2010; 45:345-54. [PMID: 20654718 DOI: 10.1016/j.mcn.2010.07.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Revised: 07/09/2010] [Accepted: 07/13/2010] [Indexed: 11/20/2022] Open
Abstract
In mouse olfactory epithelium (OE), pituitary adenylate cyclase-activating peptide (PACAP) protects against axotomy-induced apoptosis. We used mouse OE to determine whether PACAP protects neurons during exposure to the inflammatory cytokine TNFα. Live slices of neonatal mouse OE were treated with 40 ng/ml TNFα ± 40nM PACAP for 6h and dying cells were live-labeled with 0.5% propidium iodide. TNFα significantly increased the percentage of dying cells while co-incubation with PACAP prevented cell death. PACAP also prevented TNFα-mediated cell death in the olfactory placodal (OP) cell lines, OP6 and OP27. Although OP cell lines express all three PACAP receptors (PAC1, VPAC1,VPAC2), PACAP's protection of these cells from TNFα was mimicked by the specific PAC1 receptor agonist maxadilan and abolished by the PAC1 antagonist PACAP6-38. Treatment of OP cell lines with blockers or activators of the PLC and AC/MAPKK pathways revealed that PACAP-mediated protection from TNFα involved both pathways. PACAP may therefore function through PAC1 receptors to protect neurons from cell death during inflammatory cytokine release in vivo as would occur upon viral infection or allergic rhinitis-associated injury.
Collapse
|
36
|
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 2010; 43:35-43. [DOI: 10.1007/s12031-010-9427-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2010] [Accepted: 07/07/2010] [Indexed: 12/27/2022]
|
37
|
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; 42:450-8. [PMID: 20449689 DOI: 10.1007/s12031-010-9374-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Accepted: 04/13/2010] [Indexed: 10/19/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP), a neuropeptide with survival-promoting actions, has been observed in endocrine organs and is thought to play a role in reproductive functions, including pregnancy. PACAP occurs in two forms, 27 and 38 amino acid residues, with PACAP38 being the predominant form in human tissues. In the present study, we determined the concentrations of PACAP38 and PACAP27 in first-trimester and full-term human placentas using radioimmunoassay. We found high levels of PACAP38 and lower levels of PACAP27 in different parts of the full-term human placenta. PACAP38 content increased in the placenta during pregnancy, both on the maternal side and on the fetal side. The effects of PACAP on the survival of JAR human choriocarcinoma cells were investigated using flow cytometry and MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) cell viability assay in cells exposed to the widely used chemotherapeutic agent methotrexate (MTX). It was found that PACAP neither influenced the survival of JAR cytotrophoblast cells nor affected cellular response to the death-inducing effect of the chemotherapeutic agent MTX. The present observations further support the significance of PACAP in the human placenta. The observation that PACAP did not influence the effects of MTX may have future clinical importance, showing that PACAP does not decrease the effects of certain chemotherapeutic agents.
Collapse
|
38
|
Racz B, Reglodi D, Horvath G, Szigeti A, Balatonyi B, Roth E, Weber G, Alotti N, Toth G, Gasz B. Protective Effect of PACAP Against Doxorubicin-Induced Cell Death in Cardiomyocyte Culture. J Mol Neurosci 2010; 42:419-27. [DOI: 10.1007/s12031-010-9349-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2010] [Accepted: 03/04/2010] [Indexed: 12/01/2022]
|
39
|
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
|
40
|
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]
|
41
|
Horvath G, Racz B, Reglodi D, Kovacs K, Kiss P, Gallyas F, Bognar Z, Szabo A, Magyarlaki T, Laszlo E, Lubics A, Tamas A, Toth G, Szakaly P. Effects of PACAP on mitochondrial apoptotic pathways and cytokine expression in rats subjected to renal ischemia/reperfusion. J Mol Neurosci 2010; 42:411-8. [PMID: 20229361 DOI: 10.1007/s12031-010-9342-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2009] [Accepted: 02/11/2010] [Indexed: 10/19/2022]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) is a neuropeptide with highly efficient cytoprotective actions. Its neuroprotective effects are well-known, but PACAP is able to exert similar actions in non-neuronal cells. Recently, we have shown that PACAP prolongs renal ischemic time, decreases mortality, and attenuates tubular degeneration in a rat model of renal ischemia/reperfusion, but the mechanism of renoprotection is not yet known. Therefore, the aim of the present study was to obtain further insight into the renoprotective effects of PACAP by examining its direct effects of PACAP on mitochondrial permeability transition in vitro and on the expression of the anti-apoptotic Bcl-2 and cytokines/chemokines in kidney tissues following 45 and 60 min renal ischemia and reperfusion in vivo. We found that PACAP did not have any direct effect on mitochondrial permeability transition. Cytokine array revealed that the expression of a few cytokines/chemokines was strongly increased after ischemia/reperfusion, which was ameliorated by PACAP treatment. Furthermore, in rats subjected to renal ischemia, PACAP treatment counteracted the ischemia/reperfusion-induced decrease of the anti-apoptotic Bcl-2, both after 45 and 60 min ischemia, as analyzed by Western blot. In summary, we showed that PACAP could attenuate tissue injury involving both anti-inflammatory and anti-apoptotic effects, but not directly acting on mitochondrial permeability transition.
Collapse
Affiliation(s)
- Gabriella Horvath
- Department of Anatomy, School of Medicine, University of Pecs, Szigeti u 12, Pecs, Hungary.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Giunta S, Castorina A, Adorno A, Mazzone V, Carnazza ML, D'Agata V. PACAP and VIP affect NF1 expression in rat malignant peripheral nerve sheath tumor (MPNST) cells. Neuropeptides 2010; 44:45-51. [PMID: 19919880 DOI: 10.1016/j.npep.2009.10.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2009] [Revised: 09/14/2009] [Accepted: 10/15/2009] [Indexed: 10/20/2022]
Abstract
In our previous study we have identified PACAP, VIP and their receptors in rat malignant peripheral nerve sheath tumor (MPNST) cells, thus showing anti-apoptotic roles. Recently it has been shown that the tumor suppressor neurofibromin, encoded by the Neurofibromatosis type I (NF1) gene, promotes MPNST cells sensitivity to apoptosis after serum withdrawal. In the present study we investigated whether PACAP or VIP negatively regulate NF1 expression under normal or serum-dependent pro-apoptotic culture conditions. Results indicated that serum itself significantly influenced gene and protein levels. In fact, the low NF1 levels of cells cultured in normal serum-containing medium were remarkably increased in cells switched to low- or no-serum after 24h and 48 h. Treatment with 100 nM PACAP or VIP did not affect NF1 expression when using normal amounts of serum, whereas it significantly inhibited transcript and protein levels both in low- or no-serum cultured cells. In particular, PACAP reduced NF1 levels already after 24h in low-serum cultured cells, while VIP showed a similar effect only after serum deprivation. However, both PACAP and VIP downregulated gene and protein levels within 48 h either in low-dose and serum-starved cells. Results were confirmed by fluorescence microscopy, showing that 100 nM PACAP or VIP attenuated neurofibromin cytoplasmic localization only in low- or no-serum cultured cells. The present study provides a comprehensive analysis of both neuropeptides effect on NF1 expression in normal, low- or serum-starved MPNST cells, ameliorating the hypothesis that resistance to apoptosis in serum-deprived cells might be correlated to PACAP-/VIP-induced NF1 inhibition.
Collapse
Affiliation(s)
- Salvatore Giunta
- Department of Anatomy, Diagnostic Pathology, Legal Medicine, Hygiene and Public Health, University of Catania, Catania, Italy
| | | | | | | | | | | |
Collapse
|
43
|
Szigeti A, Hocsak E, Rapolti E, Racz B, Boronkai A, Pozsgai E, Debreceni B, Bognar Z, Bellyei S, Sumegi B, Gallyas F. Facilitation of mitochondrial outer and inner membrane permeabilization and cell death in oxidative stress by a novel Bcl-2 homology 3 domain protein. J Biol Chem 2010; 285:2140-51. [PMID: 19901022 PMCID: PMC2804370 DOI: 10.1074/jbc.m109.015222] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2009] [Revised: 11/01/2009] [Indexed: 11/06/2022] Open
Abstract
We identified a sequence homologous to the Bcl-2 homology 3 (BH3) domain of Bcl-2 proteins in SOUL. Tissues expressed the protein to different extents. It was predominantly located in the cytoplasm, although a fraction of SOUL was associated with the mitochondria that increased upon oxidative stress. Recombinant SOUL protein facilitated mitochondrial permeability transition and collapse of mitochondrial membrane potential (MMP) and facilitated the release of proapoptotic mitochondrial intermembrane proteins (PMIP) at low calcium and phosphate concentrations in a cyclosporine A-dependent manner in vitro in isolated mitochondria. Suppression of endogenous SOUL by diced small interfering RNA in HeLa cells increased their viability in oxidative stress. Overexpression of SOUL in NIH3T3 cells promoted hydrogen peroxide-induced cell death and stimulated the release of PMIP but did not enhance caspase-3 activation. Despite the release of PMIP, SOUL facilitated predominantly necrotic cell death, as revealed by annexin V and propidium iodide staining. This necrotic death could be the result of SOUL-facilitated collapse of MMP demonstrated by JC-1 fluorescence. Deletion of the putative BH3 domain sequence prevented all of these effects of SOUL. Suppression of cyclophilin D prevented these effects too, indicating that SOUL facilitated mitochondrial permeability transition in vivo. Overexpression of Bcl-2 and Bcl-x(L), which can counteract the mitochondria-permeabilizing effect of BH3 domain proteins, also prevented SOUL-facilitated collapse of MMP and cell death. These data indicate that SOUL can be a novel member of the BH3 domain-only proteins that cannot induce cell death alone but can facilitate both outer and inner mitochondrial membrane permeabilization and predominantly necrotic cell death in oxidative stress.
Collapse
Affiliation(s)
- Andras Szigeti
- From the Institute of Oncotherapy and
- Department of Biochemistry and Medical Chemistry, University of Pécs, 12 Szigeti Street, Pécs H-7624, Hungary
| | - Eniko Hocsak
- Department of Biochemistry and Medical Chemistry, University of Pécs, 12 Szigeti Street, Pécs H-7624, Hungary
| | - Edit Rapolti
- Department of Biochemistry and Medical Chemistry, University of Pécs, 12 Szigeti Street, Pécs H-7624, Hungary
| | - Boglarka Racz
- Department of Biochemistry and Medical Chemistry, University of Pécs, 12 Szigeti Street, Pécs H-7624, Hungary
| | - Arpad Boronkai
- From the Institute of Oncotherapy and
- Department of Biochemistry and Medical Chemistry, University of Pécs, 12 Szigeti Street, Pécs H-7624, Hungary
| | - Eva Pozsgai
- Department of Biochemistry and Medical Chemistry, University of Pécs, 12 Szigeti Street, Pécs H-7624, Hungary
| | - Balazs Debreceni
- Department of Biochemistry and Medical Chemistry, University of Pécs, 12 Szigeti Street, Pécs H-7624, Hungary
| | - Zita Bognar
- Department of Biochemistry and Medical Chemistry, University of Pécs, 12 Szigeti Street, Pécs H-7624, Hungary
| | - Szabolcs Bellyei
- From the Institute of Oncotherapy and
- Department of Biochemistry and Medical Chemistry, University of Pécs, 12 Szigeti Street, Pécs H-7624, Hungary
| | - Balazs Sumegi
- Department of Biochemistry and Medical Chemistry, University of Pécs, 12 Szigeti Street, Pécs H-7624, Hungary
| | - Ferenc Gallyas
- Department of Biochemistry and Medical Chemistry, University of Pécs, 12 Szigeti Street, Pécs H-7624, Hungary
| |
Collapse
|
44
|
Mori H, Nakamachi T, Ohtaki H, Yofu S, Sato A, Endo K, Iso Y, Suzuki H, Takeyama Y, Shintani N, Hashimoto H, Baba A, Shioda S. Cardioprotective Effect of Endogenous Pituitary Adenylate Cyclase-Activating Polypeptide on Doxorubicin-Induced Cardiomyopathy in Mice. Circ J 2010; 74:1183-90. [DOI: 10.1253/circj.cj-09-1024] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Hiroyoshi Mori
- Division of Cardiology, Department of Internal Medicine, Showa University Fujigaoka Hospital
- Department of First Anatomy, Showa University School of Medicine
| | - Tomoya Nakamachi
- Department of First Anatomy, Showa University School of Medicine
- Center for Biotechnology, Showa University
| | - Hirokazu Ohtaki
- Department of First Anatomy, Showa University School of Medicine
| | - Sachiko Yofu
- Department of First Anatomy, Showa University School of Medicine
| | - Atsushi Sato
- Department of First Anatomy, Showa University School of Medicine
| | - Kimi Endo
- Department of First Anatomy, Showa University School of Medicine
| | - Yoshitaka Iso
- Division of Cardiology, Department of Internal Medicine, Showa University Fujigaoka Hospital
| | - Hiroshi Suzuki
- Division of Cardiology, Department of Internal Medicine, Showa University Fujigaoka Hospital
| | - Youichi Takeyama
- Division of Cardiology, Department of Internal Medicine, Showa University Fujigaoka Hospital
| | - Norihito Shintani
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Hitoshi Hashimoto
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Akemichi Baba
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Seiji Shioda
- Department of First Anatomy, Showa University School of Medicine
| |
Collapse
|
45
|
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
|
46
|
Horvath G, Mark L, Brubel R, Szakaly P, Racz B, Kiss P, Tamas A, Helyes Z, Lubics A, Hashimoto H, Baba A, Shintani N, Furjes G, Nemeth J, Reglodi D. Mice deficient in pituitary adenylate cyclase activating polypeptide display increased sensitivity to renal oxidative stress in vitro. Neurosci Lett 2009; 469:70-4. [PMID: 19932736 DOI: 10.1016/j.neulet.2009.11.046] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2009] [Revised: 11/13/2009] [Accepted: 11/17/2009] [Indexed: 12/24/2022]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) is a multifunctional neuropeptide, showing widespread occurrence in the nervous system and also in peripheral organs. The neuroprotective effects of PACAP are well-established in different neuronal systems against noxious stimuli in vitro and in vivo. Recently, its general cytoprotective actions have been recognized, including renoprotective effects. However, the effect of endogenous PACAP in the kidneys is not known. The main aim of the present study was to investigate whether the lack of this endogenous neuropeptide influences survival of kidney cells against oxidative stress. First, we determined the presence of endogenous PACAP from mouse kidney homogenates by mass spectrometry and PACAP-like immunoreactivity by radioimmunoassay. Second, primary cultures were isolated from wild type and PACAP deficient mice and cell viability was assessed following oxidative stress induced by 0.5, 1.5 and 3mM H(2)O(2). Our mass spectrometry and radioimmunoassay results show that PACAP is endogenously present in the kidney. The main part of our study revealed that the sensitivity of cells from PACAP deficient mice was increased to oxidative stress: both after 2 or 4h of exposure, cell viability was significantly reduced compared to that from control wild type mice. This increased sensitivity of kidneys from PACAP deficient mice could be counteracted by exogenously given PACAP38. These results show, for the first time, that endogenous PACAP protects against oxidative stress in the kidney, and that PACAP may act as a stress sensor in renal cells. These findings further support the general cytoprotective nature of this neuropeptide.
Collapse
|
47
|
Atlasz T, Szabadfi K, Kiss P, Tamas A, Toth G, Reglodi D, Gabriel R. Evaluation of the protective effects of PACAP with cell-specific markers in ischemia-induced retinal degeneration. Brain Res Bull 2009; 81:497-504. [PMID: 19751807 DOI: 10.1016/j.brainresbull.2009.09.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2009] [Accepted: 09/08/2009] [Indexed: 01/26/2023]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) is a neurotrophic and neuroprotective peptide that has been shown to exert protective effects in different neuronal injuries, such as traumatic brain injury, models of neurodegenerative diseases and cerebral ischemia. We have provided evidence that PACAP is neuroprotective in several models of retinal degeneration in vivo. In our previous studies we showed that PACAP treatment significantly ameliorated the damaging effects of permanent bilateral common carotid artery occlusion (BCCAO). In the present study cell-type-specific markers were used in the same models in order to further specify the protective effects of PACAP. In rats BCCAO led to severe degeneration of all retinal layers that was attenuated by PACAP (100 pmol) administered unilaterally immediately following BCCAO into the vitreous body of one eye. Retinas were processed for immunohistochemistry after 3 weeks. Immunolabeling was executed for vesicular glutamate transporter 1 (VGLUT 1), vesicular gamma-aminobutyric acid transporter (VGAT), protein kinase Calpha (PKCalpha), glial fibrillary acidic protein (GFAP) and calcium-binding proteins, such as calbindin, calretinin, parvalbumin. In BCCAO retinas, intensity of immunopositivity for all antisera was dramatically decreased, except in the case of GFAP. In PACAP-treated retinas, immunostaining was similar to that of the control animals. In summary, our study presented immunohistochemical identification of cell types sensitive to chronic retinal hypoperfusion and the protective effects of PACAP. This analysis revealed that the retinoprotective effects of PACAP are not phenotype-specific, but it rather influences general cytoprotective pathways irrespective of the neuronal subtypes in the retina subjected to chronic hypoperfusion.
Collapse
Affiliation(s)
- Tamas Atlasz
- Department of Experimental Zoology and Neurobiology, University of Pecs, Pecs, Hungary.
| | | | | | | | | | | | | |
Collapse
|
48
|
Roth E, Wéber G, Kiss P, Horváth G, Tóth G, Gasz B, Ferencz A, Gallyas F, Reglodi D, Rácz B. Effects of PACAP and preconditioning against ischemia/reperfusion-induced cardiomyocyte apoptosis in vitro. Ann N Y Acad Sci 2009; 1163:512-6. [PMID: 19456402 DOI: 10.1111/j.1749-6632.2008.03635.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The neuropeptide pituitary adenylate cyclase activating polypeptide (PACAP) and its receptors are widely expressed in the nervous system and various other tissues. PACAP exerts strong anti-apoptotic effects in neuronal cell lines and, according to recent data, also in non-neuronal cells. The peptide is present in the cardiovascular system and has various distinct effects. We have demonstrated earlier that PACAP has protective effects against in vitro ischemia/reperfusion-induced apoptosis in cardiomyocytes. Preconditioning with brief intermittent periods of ischemia is known to provide protection against ischemic injury. The aim of the present study was to investigate whether PACAP could enhance the protective effect of preconditioning against in vitro ischemic injury. Cultured cardiomyocytes were exposed to brief preconditioning ischemia followed by 2 h ischemia and 4 h reperfusion. Both PACAP treatment and preconditioning alone significantly increased cell viability and decreased the ratio of cell death. Pretreatment with PACAP was found to further reduce the level of cleaved caspase-8 but it did not lead to additional survival rate when compared to cells treated with PACAP or preconditioning alone. These results show that although both PACAP and preconditioning have a protective effect against ischemia/reperfusion-induced cardiomyocyte apoptosis, their effects are not additive.
Collapse
Affiliation(s)
- E Roth
- Department of Surgical Research and Techniques, University of Pécs, Pécs, Hungary
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Boronkai A, Brubel R, Racz B, Tamas A, Kiss P, Horvath G, Lubics A, Szigeti A, Bellyei S, Toth G, Lakatos A, Reglodi D. Effects of pituitary adenylate cyclase activating polypeptide on the survival and signal transduction pathways in human choriocarcinoma cells. Ann N Y Acad Sci 2009; 1163:353-7. [PMID: 19456358 DOI: 10.1111/j.1749-6632.2008.03630.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The pituitary adenylate cyclase activating polypeptide (PACAP) has several effects in endocrine and reproductive organs, including the placenta. PACAP is generally known as a survival-promoting peptide acting on divergent signal transduction pathways. However, its effects on the survival and signaling mechanisms of trophoblast cells are not known. In the present study we found that 1-h pretreatment with PACAP38 did not significantly influence the survival of JAR cytotrophoblast cells. However, the survival rate of cells exposed to oxidative stress or CoCl(2)-induced in vitro hypoxia showed a significant further decrease in PACAP-treated cells, implying that PACAP sensitizes the cells to these stressors. This was not observed in the case of lipopolysaccharide or ethanol treatment. Western blot data revealed that, in cells exposed to oxidative stress, PACAP treatment decreased phosphorylation of all extracellular signal-regulated kinase (ERK), phospho-jun N-terminal kinase (JNK), protein kinase B, p38 mitogen-activated protein kinase (MAPK), and phospho-glycogen synthase kinase (GSK) and the expression of bax. The overall effect seems to be a sensitizing effect in almost all examined pathways when oxidative stress was applied, which may explain the enhancing effect of PACAP on cell death in contrast to most other cell types examined so far. Our data show that the signaling mechanism of PACAP may be different in trophoblast cells to that observed in other cell lines.
Collapse
Affiliation(s)
- A Boronkai
- Department of Experiemntal Zoology and Neurobiology, University of Pécs, Pécs, Hungary
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Sanchez A, Rao HV, Grammas P. PACAP38 protects rat cortical neurons against the neurotoxicity evoked by sodium nitroprusside and thrombin. ACTA ACUST UNITED AC 2008; 152:33-40. [PMID: 18682263 DOI: 10.1016/j.regpep.2008.07.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2008] [Revised: 07/02/2008] [Accepted: 07/08/2008] [Indexed: 10/21/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) 38 is a multifunctional anti-inflammatory and anti-apoptotic neuropeptide widely distributed in the nervous system. The objective of this study is to determine whether PACAP38 is neuroprotective against sodium nitroprusside (SNP) and thrombin, two mechanistically distinct neurotoxic agents. Treatment of primary cortical neuronal cultures with 1 mM SNP for 4 h causes neuronal cell death that is significantly reduced by 100 nM PACAP38. PACAP38 down-regulates SNP-induced cell cycle protein (cyclin E) expression and up-regulates p57(KIP2), a cyclin-dependent kinase inhibitor as well as the anti-apoptotic protein Bcl-2. Similarly, neuronal death induced by 100 nM thrombin or the thrombin receptor activating peptide (TRAP 6) is reduced by PACAP38 treatment. Thrombin-stimulated cell cycle protein (cdk4) expression is decreased by PACAP38 while PACAP38 inhibits thrombin-mediated reduction of p57(KIP2). However, the decrease in Bcl-2 evoked by thrombin is not affected by PACAP38. Finally, both SNP and thrombin (or TRAP) increase caspase 3 activity, an effect that is decreased by PACAP38. These data show that PACAP38 supports neuronal survival in vitro suppressing cell cycle progression and enhancing anti-apoptotic proteins. Our results support the possibility that PACAP could be a useful therapeutic agent for reducing neuronal cell death in neurodegenerative diseases.
Collapse
Affiliation(s)
- Alma Sanchez
- Department of Neuropsychiatry, Texas Tech University Health Sciences Center, Lubbock TX 79430, USA
| | | | | |
Collapse
|