1
|
Kantati YT, Kodjo MK, Lefranc B, Basille-Dugay M, Hupin S, Schmitz I, Leprince J, Gbeassor M, Vaudry D. Neuroprotective Effect of Sterculia setigera Leaves Hydroethanolic Extract. J Mol Neurosci 2024; 74:44. [PMID: 38630337 DOI: 10.1007/s12031-024-02222-6] [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: 08/19/2023] [Accepted: 04/06/2024] [Indexed: 04/19/2024]
Abstract
Plants are a valuable source of information for pharmacological research and new drug discovery. The present study aimed to evaluate the neuroprotective potential of the leaves of the medicinal plant Sterculia setigera. In vitro, the effect of Sterculia setigera leaves dry hydroethanolic extract (SSE) was tested on cultured cerebellar granule neurons (CGN) survival when exposed to hydrogen peroxide (H2O2) or 6-hydroxydopamine (6-OHDA), using the viability probe fluorescein diacetate (FDA), a lactate dehydrogenase (LDH) activity assay, an immunocytochemical staining against Gap 43, and the quantification of the expression of genes involved in apoptosis, necrosis, or oxidative stress. In vivo, the effect of intraperitoneal (ip) injection of SSE was assessed on the developing brain of 8-day-old Wistar rats exposed to ethanol neurotoxicity by measuring caspase-3 activity on cerebellum homogenates, the expression of some genes in tissue extracts, the thickness of cerebellar cortical layers and motor coordination. In vitro, SSE protected CGN against H2O2 and 6-OHDA-induced cell death at a dose of 10 µg/mL, inhibited the expression of genes Casp3 and Bad, and upregulated the expression of Cat and Gpx7. In vivo, SSE significantly blocked the deleterious effect of ethanol by reducing the activity of caspase-3, inhibiting the expression of Bax and Tp53, preventing the reduction of the thickness of the internal granule cell layer of the cerebellar cortex, and restoring motor functions. Sterculia setigera exerts neuroactive functions as claimed by traditional medicine and should be a good candidate for the development of a neuroprotective treatment against neurodegenerative diseases.
Collapse
Affiliation(s)
- Yendubé T Kantati
- Univ Rouen Normandie, Normandie Univ, NorDiC UMR 1239, 76000, Inserm, Rouen, France
- Laboratory of Physiology/Pharmacology, Physiopathology Bioactive Substances and Innocuity Research Unit (PBSI), Faculty of Sciences, of Lomé, Lomé, Togo, 01BP 1515
| | - Magloire K Kodjo
- Laboratory of Physiology/Pharmacology, Physiopathology Bioactive Substances and Innocuity Research Unit (PBSI), Faculty of Sciences, of Lomé, Lomé, Togo, 01BP 1515
| | - Benjamin Lefranc
- Univ Rouen Normandie, Normandie Univ, NorDiC UMR 1239, 76000, Inserm, Rouen, France
- Univ Rouen Normandie, CNRS, Normandie Univ, HeRacLeS US 51 UAR 2026, 76000, Inserm, Rouen, France
| | - Magali Basille-Dugay
- Univ Rouen Normandie, Normandie Univ, NorDiC UMR 1239, 76000, Inserm, Rouen, France
| | - Sébastien Hupin
- UMR 6014, Normandie Université, COBRA, Université de Rouen, INSA de Rouen-Normandie, CNRS, IRCOF, 3038, Mont Saint Aignan Cedex, FR, France
| | - Isabelle Schmitz
- UMR 6014, Normandie Université, COBRA, Université de Rouen, INSA de Rouen-Normandie, CNRS, IRCOF, 3038, Mont Saint Aignan Cedex, FR, France
- UMR 6270, Univ Rouen Normandie, INSA Rouen Normandie, CNRS, Normandie Univ, 76000, Rouen, France
| | - Jérôme Leprince
- Univ Rouen Normandie, Normandie Univ, NorDiC UMR 1239, 76000, Inserm, Rouen, France
- Univ Rouen Normandie, CNRS, Normandie Univ, HeRacLeS US 51 UAR 2026, 76000, Inserm, Rouen, France
| | - Messanvi Gbeassor
- Laboratory of Physiology/Pharmacology, Physiopathology Bioactive Substances and Innocuity Research Unit (PBSI), Faculty of Sciences, of Lomé, Lomé, Togo, 01BP 1515
| | - David Vaudry
- Univ Rouen Normandie, CNRS, Normandie Univ, HeRacLeS US 51 UAR 2026, 76000, Inserm, Rouen, France.
- UMR 1245, Laboratory of Cancer and Brain Genomics, Univ Rouen Normandie, Normandie Univ, 76000, Inserm, Rouen, France.
| |
Collapse
|
2
|
Barra T, Falanga A, Bellavita R, Pisano J, Laforgia V, Prisco M, Galdiero S, Valiante S. Neuroprotective Effects of gH625-lipoPACAP in an In Vitro Fluid Dynamic Model of Parkinson’s Disease. Biomedicines 2022; 10:biomedicines10102644. [PMID: 36289905 PMCID: PMC9599564 DOI: 10.3390/biomedicines10102644] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 10/06/2022] [Accepted: 10/12/2022] [Indexed: 11/16/2022] Open
Abstract
Parkinson’s disease (PD) is an aggressive and devastating age-related disorder. Although the causes are still unclear, several factors, including genetic and environmental, are involved. Except for symptomatic drugs, there are not, to date, any real cures for PD. For this purpose, it is necessary develop a model to better study this disease. Neuroblastoma cell line, SH-SY5Y, differentiated with retinoic acid represents a good in vitro model to explore PD, since it maintains growth cells to differentiated neurons. In the present study, SH-SY5Y cells were treated with 1-methyl-4-phenylpyridinium (MPP+), a neurotoxin that induces Parkinsonism, and the neuroprotective effects of pituitary adenylate cyclase-activating polypeptide (PACAP), delivered by functionalized liposomes in a blood–brain barrier fluid dynamic model, were evaluated. We demonstrated PACAP neuroprotective effects when delivered by gH625-liposome on MPP+-damaged SH-SY5Y spheroids.
Collapse
Affiliation(s)
- Teresa Barra
- Department of Biology, University of Naples Federico II, 80125 Naples, Italy
| | - Annarita Falanga
- Department of Agricultural Sciences, University of Naples Federico II, 80055 Portici, Italy
| | - Rosa Bellavita
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Jessica Pisano
- Department of Biology, University of Naples Federico II, 80125 Naples, Italy
| | - Vincenza Laforgia
- Department of Biology, University of Naples Federico II, 80125 Naples, Italy
| | - Marina Prisco
- Department of Biology, University of Naples Federico II, 80125 Naples, Italy
| | - Stefania Galdiero
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Salvatore Valiante
- Department of Biology, University of Naples Federico II, 80125 Naples, Italy
- Correspondence: ; Tel.: +39-081-2535169
| |
Collapse
|
3
|
Pandey S, Mudgal J. A Review on the Role of Endogenous Neurotrophins and Schwann Cells in Axonal Regeneration. J Neuroimmune Pharmacol 2022; 17:398-408. [PMID: 34843075 PMCID: PMC9810669 DOI: 10.1007/s11481-021-10034-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 11/13/2021] [Indexed: 01/13/2023]
Abstract
Injury to the peripheral nerve is traditionally referred to acquired nerve injury as they are the result of physical trauma due to laceration, stretch, crush and compression of nerves. However, peripheral nerve injury may not be completely limited to acquired physical trauma. Peripheral nerve injury equally implies clinical conditions like Guillain-Barré syndrome (GBS), Carpal tunnel syndrome, rheumatoid arthritis and diabetes. Physical trauma is commonly mono-neuropathic as it engages a single nerve and produces focal damage, while in the context of pathological conditions the damage is divergent involving a group of the nerve causing polyneuropathy. Damage to the peripheral nerve can cause a diverse range of manifestations from sensory impairment to loss of function with unpredictable recovery patterns. Presently no treatment option provides complete or functional recovery in nerve injury, as nerve cells are highly differentiated and inert to regeneration. However, the regenerative phenotypes in Schwann cells get expressed when a signalling cascade is triggered by neurotrophins. Neurotrophins are one of the promising biomolecules that are released naturally post-injury with the potential to exhibit better functional recovery. Pharmacological intervention modulating the expression of these neurotrophins such as brain-derived neurotrophic factor (BDNF) and pituitary adenylyl cyclase-activating peptide (PACAP) can prove to be a significant treatment option as endogenous compounds which may have remarkable innate advantage showing maximum 'biological relevance'.
Collapse
Affiliation(s)
- Samyak Pandey
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India, 576104
| | - Jayesh Mudgal
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India, 576104.
| |
Collapse
|
4
|
Apostol CR, Bernard K, Tanguturi P, Molnar G, Bartlett MJ, Szabò L, Liu C, Ortiz JB, Saber M, Giordano KR, Green TRF, Melvin J, Morrison HW, Madhavan L, Rowe RK, Streicher JM, Heien ML, Falk T, Polt R. Design and Synthesis of Brain Penetrant Glycopeptide Analogues of PACAP With Neuroprotective Potential for Traumatic Brain Injury and Parkinsonism. FRONTIERS IN DRUG DISCOVERY 2022; 1. [PMID: 35237767 PMCID: PMC8887546 DOI: 10.3389/fddsv.2021.818003] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
There is an unmet clinical need for curative therapies to treat neurodegenerative disorders. Most mainstay treatments currently on the market only alleviate specific symptoms and do not reverse disease progression. The Pituitary adenylate cyclase-activating polypeptide (PACAP), an endogenous neuropeptide hormone, has been extensively studied as a potential regenerative therapeutic. PACAP is widely distributed in the central nervous system (CNS) and exerts its neuroprotective and neurotrophic effects via the related Class B GPCRs PAC1, VPAC1, and VPAC2, at which the hormone shows roughly equal activity. Vasoactive intestinal peptide (VIP) also activates these receptors, and this close analogue of PACAP has also shown to promote neuronal survival in various animal models of acute and progressive neurodegenerative diseases. However, PACAP's poor pharmacokinetic profile (non-linear PK/PD), and more importantly its limited blood-brain barrier (BBB) permeability has hampered development of this peptide as a therapeutic. We have demonstrated that glycosylation of PACAP and related peptides promotes penetration of the BBB and improves PK properties while retaining efficacy and potency in the low nanomolar range at its target receptors. Furthermore, judicious structure-activity relationship (SAR) studies revealed key motifs that can be modulated to afford compounds with diverse selectivity profiles. Most importantly, we have demonstrated that select PACAP glycopeptide analogues (2LS80Mel and 2LS98Lac) exert potent neuroprotective effects and anti-inflammatory activity in animal models of traumatic brain injury and in a mild-toxin lesion model of Parkinson's disease, highlighting glycosylation as a viable strategy for converting endogenous peptides into robust and efficacious drug candidates.
Collapse
Affiliation(s)
- Christopher R Apostol
- Department of Chemistry and Biochemistry, BIO5, The University of Arizona, Tucson, AZ, United States
| | - Kelsey Bernard
- Graduate Interdisciplinary Program in Physiological Sciences, The University of Arizona, Tucson, AZ, United States
| | | | - Gabriella Molnar
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ, United States
| | - Mitchell J Bartlett
- Department of Neurology, College of Medicine, The University of Arizona, Tucson, AZ, United States
| | - Lajos Szabò
- Department of Chemistry and Biochemistry, BIO5, The University of Arizona, Tucson, AZ, United States
| | - Chenxi Liu
- Department of Chemistry and Biochemistry, BIO5, The University of Arizona, Tucson, AZ, United States
| | - J Bryce Ortiz
- Barrow Neurological Institute at Phoenix Children's Hospital, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,Department of Child Health, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,Phoenix Veteran Affairs Health Care System, Phoenix, AZ, United States
| | - Maha Saber
- Barrow Neurological Institute at Phoenix Children's Hospital, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,Department of Child Health, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
| | - Katherine R Giordano
- Barrow Neurological Institute at Phoenix Children's Hospital, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,Department of Child Health, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,Phoenix Veteran Affairs Health Care System, Phoenix, AZ, United States
| | - Tabitha R F Green
- Department of Child Health, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
| | - James Melvin
- Department of Child Health, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,Department of Biological Sciences, University of Bath, Bath, United Kingdom
| | - Helena W Morrison
- College of Nursing, University of Arizona, Tucson, AZ, United States
| | - Lalitha Madhavan
- Graduate Interdisciplinary Program in Physiological Sciences, The University of Arizona, Tucson, AZ, United States.,Department of Neurology, College of Medicine, The University of Arizona, Tucson, AZ, United States
| | - Rachel K Rowe
- Barrow Neurological Institute at Phoenix Children's Hospital, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,Department of Child Health, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - John M Streicher
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ, United States
| | - Michael L Heien
- Department of Chemistry and Biochemistry, BIO5, The University of Arizona, Tucson, AZ, United States
| | - Torsten Falk
- Graduate Interdisciplinary Program in Physiological Sciences, The University of Arizona, Tucson, AZ, United States.,Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ, United States.,Department of Neurology, College of Medicine, The University of Arizona, Tucson, AZ, United States
| | - Robin Polt
- Department of Chemistry and Biochemistry, BIO5, The University of Arizona, Tucson, AZ, United States
| |
Collapse
|
5
|
Shili I, Hamdi Y, Marouani A, Ben Lasfar Z, Ghrairi T, Lefranc B, Leprince J, Vaudry D, Olfa MK. Long-term protective effect of PACAP in a fetal alcohol syndrome (FAS) model. Peptides 2021; 146:170630. [PMID: 34481915 DOI: 10.1016/j.peptides.2021.170630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/13/2021] [Accepted: 08/14/2021] [Indexed: 12/29/2022]
Abstract
Prenatal ethanol exposure provokes teratogenic effects, due to oxidative stress and massive neuronal apoptosis in the developing brain that result in lifelong behavioral abnormalities. PACAP exerts anti-oxidative and neuroprotective activities on neuronal cells, and prevents ethanol neurotoxicity. The present study focused on the ability of PACAP to protect the brain of 30-day-old mice (P30) from prenatal alcohol exposure induced oxidative damage and toxicity. Pregnant mice were divided randomly into 4 groups, i.e. control group, ethanol group (1.5 g/kg ip daily injection), PACAP group (5 μg intrauterine daily injection) and an ethanol plus PACAP group. Offspring prenatally exposed to ethanol had decreased body weight and reduced cell survival. Moreover, production of ROS was sharply enhanced in the brain of prenatal ethanol-exposed animals, associated with an elevation in the activity of the antioxidant enzymes, and an increase of oxidative damages as shown by the accumulation of the lipid oxidation marker malondialdehyde and of protein carbonyl compounds. Intrauterine administration of PACAP during the gestational period restored the endogenous antioxidant system, prevented ROS overproduction and promoted the survival of dissociated cells from animals prenatally exposed to ethanol. Behavioral tests revealed that P30 animals exposed to ethanol during the prenatal period exhibited reduced motor activity, altered exploratory interest and increased anxiety. However, PACAP treatment significantly attenuated these behavioral impairments. This study demonstrates that PACAP exerts a potent neuroprotective effect against alcohol toxicity during brain development, and indicates that PACAP and/or PACAP analogs might be a useful tool for treatment of alcohol intoxication during pregnancy.
Collapse
Affiliation(s)
- Ilhem Shili
- University Tunis El Manar, Faculty of Sciences of Tunis, LR18ES03, Laboratory of Neurophysiology, Cellular Physiopathology and Biomelcules Valorisation, 2092 Tunis, Tunisia; Normandie Univ, UNIROUEN, Inserm, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal Death and Cell Plasticity Team, 76000 Rouen, France
| | - Yosra Hamdi
- University Tunis El Manar, Faculty of Sciences of Tunis, LR18ES03, Laboratory of Neurophysiology, Cellular Physiopathology and Biomelcules Valorisation, 2092 Tunis, Tunisia
| | - Ammar Marouani
- Institut Pasteur Tunisia, Laboratory of Venins and Toxines, B.P. 74, 1002 Tunis-Belvédère, Tunisia
| | - Zakaria Ben Lasfar
- Institut Pasteur Tunisia, Laboratory of Venins and Toxines, B.P. 74, 1002 Tunis-Belvédère, Tunisia
| | - Taoufik Ghrairi
- University Tunis El Manar, Faculty of Sciences of Tunis, LR18ES03, Laboratory of Neurophysiology, Cellular Physiopathology and Biomelcules Valorisation, 2092 Tunis, Tunisia
| | - Benjamin Lefranc
- Normandie Univ, UNIROUEN, Inserm, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal Death and Cell Plasticity Team, 76000 Rouen, France; Normandie Univ, UNIROUEN, Regional Cell Imaging Platform of Normandy (PRIMACEN), 76000 Rouen, France
| | - Jérôme Leprince
- Normandie Univ, UNIROUEN, Inserm, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal Death and Cell Plasticity Team, 76000 Rouen, France; Normandie Univ, UNIROUEN, Regional Cell Imaging Platform of Normandy (PRIMACEN), 76000 Rouen, France
| | - David Vaudry
- Normandie Univ, UNIROUEN, Inserm, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal Death and Cell Plasticity Team, 76000 Rouen, France; Normandie Univ, UNIROUEN, Regional Cell Imaging Platform of Normandy (PRIMACEN), 76000 Rouen, France.
| | - Masmoudi-Kouki Olfa
- University Tunis El Manar, Faculty of Sciences of Tunis, LR18ES03, Laboratory of Neurophysiology, Cellular Physiopathology and Biomelcules Valorisation, 2092 Tunis, Tunisia.
| |
Collapse
|
6
|
Apostol CR, Tanguturi P, Szabò LZ, Varela D, Gilmartin T, Streicher JM, Polt R. Synthesis and In Vitro Characterization of Glycopeptide Drug Candidates Related to PACAP 1-23. Molecules 2021; 26:4932. [PMID: 34443519 PMCID: PMC8401035 DOI: 10.3390/molecules26164932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 01/05/2023] Open
Abstract
The search for efficacious treatment of neurodegenerative and progressive neuroinflammatory diseases continues, as current therapies are unable to halt or reverse disease progression. PACAP represents one potential therapeutic that provides neuroprotection effects on neurons, and also modulates inflammatory responses and circulation within the brain. However, PACAP is a relatively long peptide hormone that is not trivial to synthesize. Based on previous observations that the shortened isoform PACAP1-23 is capable of inducing neuroprotection in vitro, we were inspired to synthesize shortened glycopeptide analogues of PACAP1-23. Herein, we report the synthesis and in vitro characterization of glycosylated PACAP1-23 analogues that interact strongly with the PAC1 and VPAC1 receptors, while showing reduced activity at the VPAC2 receptor.
Collapse
Affiliation(s)
- Christopher R. Apostol
- Department of Chemistry and Biochemistry, BIO5, The University of Arizona, 1306 E. University Blvd, Tucson, AZ 85721, USA; (C.R.A.); (L.Z.S.)
| | - Parthasaradhireddy Tanguturi
- Department of Pharmacology, College of Medicine, The University of Arizona, 1501 N. Campbell Ave, Tucson, AZ 85724, USA; (P.T.); (J.M.S.)
| | - Lajos Z. Szabò
- Department of Chemistry and Biochemistry, BIO5, The University of Arizona, 1306 E. University Blvd, Tucson, AZ 85721, USA; (C.R.A.); (L.Z.S.)
| | - Daniel Varela
- Facultat de Quìmica Tarragona, Universitat Rovera I Virgili, 43007 Barcelona, Spain; (D.V.); (T.G.)
| | - Thiago Gilmartin
- Facultat de Quìmica Tarragona, Universitat Rovera I Virgili, 43007 Barcelona, Spain; (D.V.); (T.G.)
| | - John M. Streicher
- Department of Pharmacology, College of Medicine, The University of Arizona, 1501 N. Campbell Ave, Tucson, AZ 85724, USA; (P.T.); (J.M.S.)
| | - Robin Polt
- Department of Chemistry and Biochemistry, BIO5, The University of Arizona, 1306 E. University Blvd, Tucson, AZ 85721, USA; (C.R.A.); (L.Z.S.)
| |
Collapse
|
7
|
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
|
8
|
Pleiotropic pituitary adenylate cyclase-activating polypeptide (PACAP): Novel insights into the role of PACAP in eating and drug intake. Brain Res 2019; 1729:146626. [PMID: 31883848 PMCID: PMC6953419 DOI: 10.1016/j.brainres.2019.146626] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 12/20/2019] [Accepted: 12/24/2019] [Indexed: 01/30/2023]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) was discovered thirty years ago, but its role in eating and drug use disorders has only recently begun to be investigated. The present review develops the hypothesis that, although PACAP normally functions to tightly regulate intake, inhibiting it through negative feedback, this relationship can become dysregulated with the development of dependence, such that PACAP instead acts through positive feedback to promote excessive intake. We propose that repeated exposure to palatable food and drugs of abuse can alter the downstream responses of specific populations of neurons to stimulation by PACAP, leading to the perpetuation of the addiction cycle. Thus, this review will first describe published literature on homeostatic food intake, which shows that PACAP suppresses food intake, while its levels are themselves increased by overfeeding. Next, it will present literature on palatable food, cocaine, alcohol, and nicotine, which overall demonstrates that PACAP in specific limbic brain regions can promote their seeking and intake and itself is stimulated by their intake. Then, it will present literature on affective behavior, which shows that chronic stress increases levels of PACAP, which then promotes anxiety and depression, factors that can trigger substance seeking. Finally, the review will address mechanisms through which chronic substance exposure may dysregulate the PACAP system, proposing that it alters expression of PACAP receptor splice variants. While many questions remain to be addressed, the current evidence suggests that PACAP could be a viable medication target for the treatment of binge eating and drug and alcohol use disorders.
Collapse
|
9
|
Stathaki M, Stamatiou ME, Magioris G, Simantiris S, Syrigos N, Dourakis S, Koutsilieris M, Armakolas A. The role of kisspeptin system in cancer biology. Crit Rev Oncol Hematol 2019; 142:130-140. [PMID: 31401420 DOI: 10.1016/j.critrevonc.2019.07.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 02/01/2019] [Accepted: 07/18/2019] [Indexed: 02/08/2023] Open
Abstract
Kisspeptins are a family of neuropeptides that are known to be critical in puberty initiation and ovulation. Apart from that kisspeptin derived peptides (KPs) are also known for their antimetastatic activities in several malignancies. Herein we report recent evidence of the role of kisspeptins in cancer biology and we examine the prospective of targeting the kisspeptin pathways leading to a better prognosis in patients with malignant diseases.
Collapse
Affiliation(s)
- Martha Stathaki
- Physiology Laboratory, Athens Medical School, National and Kapodestrian University of Athens, Greece
| | - Maria Evanthia Stamatiou
- Physiology Laboratory, Athens Medical School, National and Kapodestrian University of Athens, Greece
| | - George Magioris
- Physiology Laboratory, Athens Medical School, National and Kapodestrian University of Athens, Greece
| | - Spyridon Simantiris
- Physiology Laboratory, Athens Medical School, National and Kapodestrian University of Athens, Greece
| | - Nikolaos Syrigos
- Physiology Laboratory, Athens Medical School, National and Kapodestrian University of Athens, Greece
| | - Spyridon Dourakis
- 2nd Academic Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens School of Medicine Hippokration General Hospital Athens Greece, Greece
| | - Michael Koutsilieris
- Physiology Laboratory, Athens Medical School, National and Kapodestrian University of Athens, Greece
| | - Athanasios Armakolas
- Physiology Laboratory, Athens Medical School, National and Kapodestrian University of Athens, Greece.
| |
Collapse
|
10
|
Denes V, Hideg O, Nyisztor Z, Lakk M, Godri Z, Berta G, Geck P, Gabriel R. The Neuroprotective Peptide PACAP1-38 Contributes to Horizontal Cell Development in Postnatal Rat Retina. Invest Ophthalmol Vis Sci 2019; 60:770-778. [PMID: 30795011 DOI: 10.1167/iovs.18-25719] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose PACAP1-38, a member of the secretin/glucagon superfamily, is expressed in the developing retina with documented neuroprotective effects. However, its function in retinal cell differentiation has yet to be elucidated. Our goals, therefore, were to identify PAC1 expressing cells morphologically, investigate the PACAP1-38 action functionally, and establish PACAP1-38 regulated events developmentally during the first postnatal week in rat retina. Methods P1 retinal sections or whole mounts of Wistar rats were used to reveal PAC1 and calbindin immunoreactive structures. P1, P3, or P7 pups were injected intravitreally with 100 pmol PACAP1-38. Tissues were harvested 24 hours post-treatment, then processed for calbindin immunohistochemistry to determine horizontal cell number, or 6, 12, 24 hours post-treatment for real-time PCR and immunoblots to detect PCNA expression. To localize proliferating cells, anti-PCNA antibody was applied. Results We showed various PAC1 expressing cells in RPE, NBL, and GCL in P1 retina including calbindin positive horizontal cells. We found that PACAP1-38 induced a marked cell number increase at P3 and P7 and showed upregulated cell proliferation as its mechanism; however, it was ineffective at P1. PACAP1-38 induced proliferative cells localized in the NBL, and double-marker studies demonstrated that the induced proliferative cells were horizontal cells. Conclusions PACAP1-38 appears to act in retinal differentiation by inducing mitosis selectively in a time and cell specific manner through PAC1. The control of horizontal cell proliferation raises the novel possibilities that (1) PACAP1-38 may be a major player in retinal patterning and (2) PACAP signaling may be critical in retinoblastoma.
Collapse
Affiliation(s)
- Viktoria Denes
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary
| | - Orsolya Hideg
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary
| | - Zsolt Nyisztor
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary
| | - Monika Lakk
- Department of Ophthalmology and Visual Sciences, School of Medicine, University of Utah, Salt Lake City, Utah, United States
| | - Zoltan Godri
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary
| | - Gergely Berta
- Department of Medical Biology and Central Electron Microscope Laboratory, Medical School, University of Pécs, Pécs, Hungary
| | - Peter Geck
- Department of Immunology, School of Medicine, Tufts University, Boston, Massachusetts, United States
| | - Robert Gabriel
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary
| |
Collapse
|
11
|
Reglodi D, Toth D, Vicena V, Manavalan S, Brown D, Getachew B, Tizabi Y. Therapeutic potential of PACAP in alcohol toxicity. Neurochem Int 2019; 124:238-244. [PMID: 30682380 DOI: 10.1016/j.neuint.2019.01.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 12/15/2018] [Accepted: 01/21/2019] [Indexed: 12/19/2022]
Abstract
Alcohol addiction is a worldwide concern as its detrimental effects go far beyond the addicted individual and can affect the entire family as well as the community. Considerable effort is being expended in understanding the neurobiological basis of such addiction in hope of developing effective prevention and/or intervention strategies. In addition, organ damage and neurotoxicological effects of alcohol are intensely investigated. Pharmacological approaches, so far, have only provided partial success in prevention or treatment of alcohol use disorder (AUD) including the neurotoxicological consequences of heavy drinking. Pituitary adenylate cyclase-activating polypeptide (PACAP) is an endogenous 38 amino-acid neuropeptide with demonstrated protection against neuronal injury, trauma as well as various endogenous and exogenous toxic agents including alcohol. In this mini-review, following a brief presentation of alcohol addiction and its neurotoxicity, the potential of PACAP as a therapeutic intervention in toxicological consequences of this devastating disorder is discussed.
Collapse
Affiliation(s)
- Dora Reglodi
- Department of Anatomy, MTA-PTE PACAP Research Team, University of Pecs Medical School, Hungary.
| | - Denes Toth
- Department of Forensic Medicine, University of Pecs Medical School, Hungary
| | - Viktoria Vicena
- Department of Anatomy, MTA-PTE PACAP Research Team, University of Pecs Medical School, Hungary
| | - Sridharan Manavalan
- Department of Anatomy, MTA-PTE PACAP Research Team, University of Pecs Medical School, Hungary; Department of Basic Sciences, National University of Health Sciences, Florida, USA
| | - Dwayne Brown
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, USA
| | - Bruk Getachew
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, USA
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, USA
| |
Collapse
|
12
|
Nyisztor Z, Denes V, Kovacs-Valasek A, Hideg O, Berta G, Gabriel R. Pituitary Adenylate Cyclase Activating Polypeptide (PACAP1-38) Exerts Both Pro and Anti-Apoptotic Effects on Postnatal Retinal Development in Rat. Neuroscience 2018; 385:59-66. [PMID: 29906550 DOI: 10.1016/j.neuroscience.2018.06.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 06/01/2018] [Accepted: 06/04/2018] [Indexed: 02/07/2023]
Abstract
PACAP1-38, a ubiquitous and multifunctional regulator has been in the focus of neurotoxicity research due to its impressive neuroprotective potential. Although the literature extensively demonstrated its repressive effect on the apoptotic machinery in neurodegenerative models, there is a striking absence of analysis on its role in normal development. We performed quantitative analyses on caspase activity in developing retina upon 100, 50, 25 or 1 pmol intravitreal PACAP1-38 injection from postnatal day 1 (P1) through P7 in Wistar rats. Retinas were harvested at 6, 12, 18, 24 or 48 h post-injection. Apoptotic activity was revealed using fluorescent caspase 3/7 enzyme assay, western blots and TUNEL assay. Unexpectedly, we found that 100 pmol PACAP1-38 increased the activity of caspase 3/7 at P1 and P5 whereas it had no effect at P7. At P3, as a biphasic effect, PACAP1-38 repressed active caspase 3/7 at 18 h post-injection while increased their activity in 24 h post-injection. Amounts, smaller than 100 pmol, could not inhibit apoptosis whereas 50, 25 or 1 pmol PACAP1-38 could evoke significant elevation in caspase 3/7 activity. TUNEL-positive cells appeared in the proximal part of inner nuclear as well as ganglion cell layers in response to PACAP1-38 treatment. The fundamental novelty of these results is that PACAP1-38 induces apoptosis during early postnatal retinogenesis. The dose as well as stage-dependent response suggests that PACAP1-38 has a Janus face in apoptosis regulation. It not only inhibits development-related apoptosis, but as a long-term effect, facilitates it.
Collapse
Affiliation(s)
- Zsolt Nyisztor
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary
| | - Viktoria Denes
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary.
| | - Andrea Kovacs-Valasek
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary
| | - Orsolya Hideg
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary
| | - Gergely Berta
- Institute of Medical Biology, School of Medicine, University of Pécs, Pécs, Hungary
| | - Robert Gabriel
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary
| |
Collapse
|
13
|
Protective effects of pituitary adenylate cyclase activating polypeptide against neurotoxic agents. Neurotoxicology 2018; 66:185-194. [DOI: 10.1016/j.neuro.2018.03.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 03/09/2018] [Accepted: 03/27/2018] [Indexed: 01/28/2023]
|
14
|
Kovács-Valasek A, Szabadfi K, Dénes V, Szalontai B, Tamás A, Kiss P, Szabó A, Setalo G, Reglődi D, Gábriel R. Accelerated retinal aging in PACAP knock-out mice. Neuroscience 2017; 348:1-10. [PMID: 28215987 DOI: 10.1016/j.neuroscience.2017.02.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 01/31/2017] [Accepted: 02/02/2017] [Indexed: 12/26/2022]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) is a neurotrophic and neuroprotective peptide. PACAP and its receptors are widely distributed in the retina. A number of reports provided evidence that PACAP is neuroprotective in retinal degenerations. The current study compared retina cell type-specific differences in young (3-4months) and aged adults (14-16months), of wild-type (WT) mice and knock-out (KO) mice lacking endogenous PACAP production during the course of aging. Histological, immunocytochemical and Western blot examinations were performed. The staining for standard neurochemical markers (tyrosine hydroxylase for dopaminergic cells, calbindin 28 kDa for horizontal cells, protein kinase Cα for rod bipolar cells) of young adult PACAP KO retinas showed no substantial alterations compared to young adult WT retinas, except for the specific PACAP receptor (PAC1-R) staining. We could not detect PAC1-R immunoreactivity in bipolar and horizontal cells in young adult PACAP KO animals. Some other age-related changes were observed only in the PACAP KO mice only. These alterations included horizontal and rod bipolar cell dendritic sprouting into the photoreceptor layer and decreased ganglion cell number. Also, Müller glial cells showed elevated GFAP expression compared to the aging WT retinas. Furthermore, Western blot analyses revealed significant differences between the phosphorylation state of ERK1/2 and JNK in KO mice, indicating alterations in the MAPK signaling pathway. These results support the conclusion that endogenous PACAP contributes to protection against aging of the nervous system.
Collapse
Affiliation(s)
- Andrea Kovács-Valasek
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary; Department of János Szentágothai Research Center, University of Pécs, Pécs, Hungary
| | - Krisztina Szabadfi
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary
| | - Viktória Dénes
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary
| | - Bálint Szalontai
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary
| | - Andrea Tamás
- Department of Anatomy, University of Pécs, Pécs, Hungary
| | - Péter Kiss
- Department of Anatomy, University of Pécs, Pécs, Hungary
| | - Aliz Szabó
- Department of Biochemistry and Medicinal Chemistry, University of Pécs, Pécs, Hungary
| | - Gyorgy Setalo
- Department of Medical Biology, University of Pécs, Pécs, Hungary; Department of János Szentágothai Research Center, University of Pécs, Pécs, Hungary
| | - Dóra Reglődi
- Department of Anatomy, University of Pécs, Pécs, Hungary
| | - Robert Gábriel
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary; Department of János Szentágothai Research Center, University of Pécs, Pécs, Hungary.
| |
Collapse
|
15
|
PACAP Protects the Adolescent and Adult Mice Brain from Ethanol Toxicity and Modulates Distinct Sets of Genes Regulating Similar Networks. Mol Neurobiol 2016; 54:7534-7548. [PMID: 27826748 DOI: 10.1007/s12035-016-0204-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 10/11/2016] [Indexed: 12/30/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a 38-amino acid neuropeptide which has been shown to exert various neuroprotective actions in vitro and in vivo; however, the ability of endogenous PACAP to prevent cell death in vivo remains to be elucidated. To explore the capacity of endogenous PACAP to prevent ethanol toxicity, adolescent and adult PACAP knockout (KO) mice were injected with ethanol in a binge drinking-like manner. Biochemical analyses revealed that ethanol administration induced an increase in the production of reactive oxygen species and the activity of caspase-3 in PACAP KO mice in an age-independent manner. In order to characterize the mechanisms underlying the sensitivity of PACAP KO mice, a whole-genome microarray analysis was performed to compare gene regulations induced by ethanol in adolescent and adult wild-type and PACAP KO mice. Gene expression substantially differed between adolescent and adult wild-type mice, suggesting distinct effects of ethanol according to the state of brain maturation. Interestingly, in adolescent and adult PACAP KO mice, the set of genes regulated were also markedly different but seemed to inhibit some similar regulatory network processes associated in particular with DNA repair and cell cycle. These data imply that ethanol induces serious DNA damages and cell cycle alteration in PACAP KO mice. This hypothesis, based on the transcriptomic data, could be confirmed by functional studies which showed that cell proliferation decreased in adolescent and adult PACAP KO mice treated with ethanol but recovered after a 30-day withdrawal period. These data, obtained with PACAP KO animals, demonstrate that endogenous PACAP protects the brain of adolescent and adult mice from alcohol toxicity and modulates distinct sets of genes according to the maturation status of the brain.
Collapse
|
16
|
Lakk M, Denes V, Gabriel R. Pituitary Adenylate Cyclase-Activating Polypeptide Receptors Signal via Phospholipase C Pathway to Block Apoptosis in Newborn Rat Retina. Neurochem Res 2015; 40:1402-9. [PMID: 25975365 DOI: 10.1007/s11064-015-1607-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 04/03/2015] [Accepted: 05/07/2015] [Indexed: 12/22/2022]
Abstract
Glutamate induced cell death mechanisms gained considerable attention lately as excessive release of extracellular glutamate was reported to cause neurodegeneration in brain areas including the retina. Conversely, pituitary adenylate cyclase-activating polypeptide (PACAP) was shown to provide neuroprotection through anti-apoptotic effects in the glutamate-model and also in other degeneration assays. Although PACAP is known to orchestrate complex intracellular signaling primarily through cAMP production, the mechanism that mediates the anti-apoptotic effect in glutamate excitotoxicity remains to be clarified. To study this mechanism we induced retinal neurodegeneration in newborn Wistar rats by subcutaneous monosodium-glutamate injection. 100 pmol PACAP and enzyme inhibitors were administered intravitreally. Levels of caspase 3, 9, and phospho-protein kinase A were assessed by Western blots. Changes in cAMP levels were detected employing a competitive immunoassay. We found that cAMP blockade by an adenylyl-cyclase inhibitor (2',4'-dideoxy-adenosine) did not abrogate the neuroprotective effect of PACAP1-38. We show that following intravitreal PACAP1-38 treatment cAMP was unaltered, consistent with the inhibitor results and phospho-protein kinase A, an effector of the cAMP pathway was also unaffected. On the other hand, blockade of the alternative phosphatidylcholine-specific PLC pathway using an inhibitor (D609CAS) abrogated the neuroprotective effects of PACAP1-38. Our results highlight PACAP1-38 ability in protecting retinal cells against apoptosis through diverse signaling cascades. It seems that at picomolar concentrations, PACAP does not trigger cAMP production, but nonetheless, exerts a significant anti-apoptotic effect through PLC activation. In conclusion, PACAP1-38 may signal via both AC and PLC activation producing the same protective outcome.
Collapse
Affiliation(s)
- Monika Lakk
- Department of Experimental Zoology and Neurobiology, University of Pécs, 6 Ifjúság Street, 7601, Pécs, Hungary
| | | | | |
Collapse
|
17
|
Brown D, Tamas A, Reglodi D, Tizabi Y. PACAP Protects Against Inflammatory-Mediated Toxicity in Dopaminergic SH-SY5Y Cells: Implication for Parkinson’s Disease. Neurotox Res 2014; 26:230-9. [DOI: 10.1007/s12640-014-9468-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 03/21/2014] [Accepted: 04/01/2014] [Indexed: 12/31/2022]
|
18
|
Huang H, Wang H, Figueiredo-Pereira ME. Regulating the ubiquitin/proteasome pathway via cAMP-signaling: neuroprotective potential. Cell Biochem Biophys 2014; 67:55-66. [PMID: 23686612 DOI: 10.1007/s12013-013-9628-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The cAMP-signaling pathway has been under intensive investigation for decades. It is a wonder that such a small simple molecule like cAMP can modulate a vast number of diverse processes in different types of cells. The ubiquitous involvement of cAMP-signaling in a variety of cellular events requires tight spatial and temporal control of its generation, propagation, compartmentalization, and elimination. Among the various steps of the cAMP-signaling pathway, G-protein-coupled receptors, adenylate cyclases, phosphodiesterases, the two major cAMP targets, i.e., protein kinase A and exchange protein activated by cAMP, as well as the A-kinase anchoring proteins, are potential targets for drug development. Herein we review the recent progress on the regulation and manipulation of different steps of the cAMP-signaling pathway. We end by focusing on the emerging role of cAMP-signaling in modulating protein degradation via the ubiquitin/proteasome pathway. New discoveries on the regulation of the ubiquitin/proteasome pathway by cAMP-signaling support the development of new therapeutic approaches to prevent proteotoxicity in chronic neurodegenerative disorders and other human disease conditions associated with impaired protein turnover by the ubiquitin/proteasome pathway and the accumulation of ubiquitin-protein aggregates.
Collapse
Affiliation(s)
- He Huang
- Department of Biological Sciences, Hunter College and Graduate Center, City University of New York, 695 Park Avenue, New York, NY 10065, USA
| | | | | |
Collapse
|
19
|
PACAP protects against salsolinol-induced toxicity in dopaminergic SH-SY5Y cells: implication for Parkinson's disease. J Mol Neurosci 2013; 50:600-7. [PMID: 23625270 DOI: 10.1007/s12031-013-0015-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 04/15/2013] [Indexed: 01/14/2023]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is an endogenous 38 amino acid containing neuropeptide with various cytoprotective functions including neuroprotection. Administration of PACAP has been shown to reduce damage induced by ischemia, trauma, or exogenous toxic substances. Moreover, mice deficient in PACAP are more vulnerable to damaging insults. In this study, we sought to determine whether PACAP may also be protective against salsolinol-induced toxicity in SH-SY5Y cells and, if so, elucidate its mechanism(s) of action. Salsolinol (SALS) is an endogenous dopamine metabolite with selective toxicity to nigral dopaminergic neurons, which are directly implicated in Parkinson's disease (PD). SH-SY5Y cells, derived from human neuroblastoma cells, express high levels of dopaminergic activity and are used extensively as a model to study these neurons. Exposure of SH-SY5Y cells to 400 μM SALS for 24 h resulted in approximately 50 % cell death that was mediated by apoptosis as determined by cell flow cytometry and increases in caspase-3 levels. Cellular toxicity was also associated with reductions in brain-derived neurotrophic factor and phosphorylated cyclic AMP response element-binding protein. Pretreatment with PACAP dose-dependently attenuated SALS-induced toxicity and the associated apoptosis and the chemical changes. PACAP receptor antagonist PACAP6-38, in turn, dose-dependently blocked the effects of PACAP. Neither PACAP nor PACAP antagonist had any effect of its own on cellular viability. These results suggest the protective effects of PACAP in a cellular model of PD. Hence, PACAP or its agonists could be of therapeutic benefit in PD.
Collapse
|
20
|
Luo J. Mechanisms of ethanol-induced death of cerebellar granule cells. THE CEREBELLUM 2012; 11:145-54. [PMID: 20927663 DOI: 10.1007/s12311-010-0219-0] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Maternal ethanol exposure during pregnancy may cause fetal alcohol spectrum disorders (FASD). FASD is the leading cause of mental retardation. The most deleterious effect of fetal alcohol exposure is inducing neuroapoptosis in the developing brain. Ethanol-induced loss of neurons in the central nervous system underlies many of the behavioral deficits observed in FASD. The cerebellum is one of the brain areas that are most susceptible to ethanol during development. Ethanol exposure causes a loss of both cerebellar Purkinje cells and granule cells. This review focuses on the toxic effect of ethanol on cerebellar granule cells (CGC) and the underlying mechanisms. Both in vitro and in vivo studies indicate that ethanol induces apoptotic death of CGC. The vulnerability of CGC to ethanol-induced death diminishes over time as neurons mature. Several mechanisms for ethanol-induced apoptosis of CGC have been suggested. These include inhibition of N-methyl-D-aspartate receptors, interference with signaling by neurotrophic factors, induction of oxidative stress, modulation of retinoid acid signaling, disturbance of potassium channel currents, thiamine deficiency, and disruption of translational regulation. Cultures of CGC provide an excellent system to investigate cellular/molecular mechanisms of ethanol-induced neurodegeneration and to evaluate interventional strategies. This review will also discuss the approaches leading to neuroprotection against ethanol-induced neuroapoptosis.
Collapse
Affiliation(s)
- Jia Luo
- Department of Internal Medicine, University of Kentucky College of Medicine, Lexington, KY 40536, USA.
| |
Collapse
|
21
|
Pinilla L, Aguilar E, Dieguez C, Millar RP, Tena-Sempere M. Kisspeptins and Reproduction: Physiological Roles and Regulatory Mechanisms. Physiol Rev 2012; 92:1235-316. [DOI: 10.1152/physrev.00037.2010] [Citation(s) in RCA: 529] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Procreation is essential for survival of species. Not surprisingly, complex neuronal networks have evolved to mediate the diverse internal and external environmental inputs that regulate reproduction in vertebrates. Ultimately, these regulatory factors impinge, directly or indirectly, on a final common pathway, the neurons producing the gonadotropin-releasing hormone (GnRH), which stimulates pituitary gonadotropin secretion and thereby gonadal function. Compelling evidence, accumulated in the last few years, has revealed that kisspeptins, a family of neuropeptides encoded by the Kiss1 gene and produced mainly by neuronal clusters at discrete hypothalamic nuclei, are pivotal upstream regulators of GnRH neurons. As such, kisspeptins have emerged as important gatekeepers of key aspects of reproductive maturation and function, from sexual differentiation of the brain and puberty onset to adult regulation of gonadotropin secretion and the metabolic control of fertility. This review aims to provide a comprehensive account of the state-of-the-art in the field of kisspeptin physiology by covering in-depth the consensus knowledge on the major molecular features, biological effects, and mechanisms of action of kisspeptins in mammals and, to a lesser extent, in nonmammalian vertebrates. This review will also address unsolved and contentious issues to set the scene for future research challenges in the area. By doing so, we aim to endow the reader with a critical and updated view of the physiological roles and potential translational relevance of kisspeptins in the integral control of reproductive function.
Collapse
Affiliation(s)
- Leonor Pinilla
- Department of Cell Biology, Physiology and Immunology, University of Córdoba; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III; and Instituto Maimónides de Investigaciones Biomédicas, Córdoba, Spain; Department of Physiology, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain; and Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Enrique Aguilar
- Department of Cell Biology, Physiology and Immunology, University of Córdoba; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III; and Instituto Maimónides de Investigaciones Biomédicas, Córdoba, Spain; Department of Physiology, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain; and Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Carlos Dieguez
- Department of Cell Biology, Physiology and Immunology, University of Córdoba; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III; and Instituto Maimónides de Investigaciones Biomédicas, Córdoba, Spain; Department of Physiology, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain; and Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Robert P. Millar
- Department of Cell Biology, Physiology and Immunology, University of Córdoba; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III; and Instituto Maimónides de Investigaciones Biomédicas, Córdoba, Spain; Department of Physiology, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain; and Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Manuel Tena-Sempere
- Department of Cell Biology, Physiology and Immunology, University of Córdoba; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III; and Instituto Maimónides de Investigaciones Biomédicas, Córdoba, Spain; Department of Physiology, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain; and Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
22
|
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]
|
23
|
Szabadfi K, Atlasz T, Kiss P, Danyadi B, Tamas A, Helyes Z, Hashimoto H, Shintani N, Baba A, Toth G, Gabriel R, Reglodi D. Mice deficient in pituitary adenylate cyclase activating polypeptide (PACAP) are more susceptible to retinal ischemic injury in vivo. Neurotox Res 2011; 21:41-8. [PMID: 21717232 DOI: 10.1007/s12640-011-9254-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Revised: 06/14/2011] [Accepted: 06/16/2011] [Indexed: 12/19/2022]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) is a neuroprotective peptide exerting protective effects in neuronal injuries. We have provided evidence that PACAP is neuroprotective in several models of retinal degeneration in vivo. Our previous studies showed that PACAP treatment ameliorated the damaging effects of chronic hypoperfusion modeled by permanent bilateral carotid artery occlusion. We have also demonstrated in earlier studies that treatment with PACAP antagonists further aggravates retinal lesions. It has been shown that PACAP deficient mice have larger infarct size in cerebral ischemia. The aim of this study was to compare the degree of retinal damage in wild type and PACAP deficient mice in ischemic retinal insult. Mice underwent 10 min of bilateral carotid artery occlusion followed by 2-week reperfusion period. Retinas were then processed for histological analysis. It was found that PACAP deficient mice had significantly greater retinal damage, as shown by the thickness of the whole retina, the morphometric analysis of the individual retinal layers, and the cell numbers in the inner nuclear and ganglion cell layers. Exogenous PACAP administration could partially protect against retinal degeneration in PACAP deficient mice. These results clearly show that endogenous PACAP reacts as a stress-response peptide that is necessary for endogenous protection against different retinal insults.
Collapse
Affiliation(s)
- K Szabadfi
- Department of Experimental Zoology, University of Pecs, Pecs, Hungary
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|