1
|
Magrì B, D'Amico AG, Maugeri G, Morello G, La Cognata V, Saccone S, Federico C, Cavallaro S, D'Agata V. Neuroprotective effect of the PACAP-ADNP axis on SOD1G93A mutant motor neuron death induced by trophic factors deprivation. Neuropeptides 2023; 102:102386. [PMID: 37856900 DOI: 10.1016/j.npep.2023.102386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/04/2023] [Accepted: 10/07/2023] [Indexed: 10/21/2023]
Abstract
Amyotrophic lateral Sclerosis (ALS) is a neurodegenerative disease characterized by progressive degeneration of motor neurons in the central nervous system. Mutations in the gene encoding Cu/Zn superoxide dismutase (SOD1) account for approximately in 20% of familial ALS cases. The pathological mechanisms underlying the toxicity induced by mutated SOD1 are still unknown. However, it has been hypothesized that oxidative stress (OS) has a crucial role in motor neuron degeneration in ALS patients. Moreover, it has been described that SOD1 mutation interferes expression of nuclear factor erythroid 2-related factor 2 (Nrf2), a protective key modulator against OS and reactive oxygen species (ROS) formation. The protective effect of pituitary adenylate cyclase-activating peptide (PACAP) has been demonstrated in various neurological disorders, including ALS. Some of its effects are mediated by the stimulation of an intracellular factor known as activity-dependent protein (ADNP). The role of PACAP-ADNP axis on mutated SOD1 motor neuron degeneration has not been explored, yet. The present study aimed to investigate whether PACAP prevented apoptotic cell death induced by growth factor deprivation through ADNP activation and whether the peptidergic axis can counteract the OS insult. By using an in vitro model of ALS, we demonstrated that PACAP by binding to PAC1 receptor (PAC1R) prevented motor neuron death induced by serum deprivation through induction of the ADNP expression via PKC stimulation. Furthermore, we have also demonstrated that the PACAP/ADNP axis counteracted ROS formation by inducing translocation of the Nfr2 from the cytoplasm to the nucleus. In conclusion, our study provides new insights regarding the protective role of PACAP-ADNP in ALS.
Collapse
Affiliation(s)
- Benedetta Magrì
- Department of Biomedical and Biotechnological Sciences, Section of Anatomy, Histology and Movement Sciences, University of Catania, Italy
| | | | - Grazia Maugeri
- Department of Biomedical and Biotechnological Sciences, Section of Anatomy, Histology and Movement Sciences, University of Catania, Italy
| | - Giovanna Morello
- Institute for Biomedical Research and Innovation, National Research Council, Via P. Gaifami 18, Catania 95126, Italy
| | - Valentina La Cognata
- Institute for Biomedical Research and Innovation, National Research Council, Via P. Gaifami 18, Catania 95126, Italy
| | - Salvatore Saccone
- Department of Biological, Geological and Environmental Sciences, Section of Animal Biology, University of Catania, Catania 95123, Italy
| | - Concetta Federico
- Department of Biological, Geological and Environmental Sciences, Section of Animal Biology, University of Catania, Catania 95123, Italy
| | - Sebastiano Cavallaro
- Institute for Biomedical Research and Innovation, National Research Council, Via P. Gaifami 18, Catania 95126, Italy
| | - Velia D'Agata
- Department of Biomedical and Biotechnological Sciences, Section of Anatomy, Histology and Movement Sciences, University of Catania, Italy.
| |
Collapse
|
2
|
Wang Q, Wang Y, Li S, Shi J. PACAP-Sirtuin3 alleviates cognitive impairment through autophagy in Alzheimer's disease. Alzheimers Res Ther 2023; 15:184. [PMID: 37891608 PMCID: PMC10605376 DOI: 10.1186/s13195-023-01334-2] [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: 07/28/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023]
Abstract
BACKGROUND Autophagy is vital in the pathogenesis of neurodegeneration. Thus far, no studies have specifically investigated the relationship between pituitary adenylate cyclase-activating polypeptide (PACAP) and autophagy, particularly in the context of Alzheimer's disease (AD). This study used in vitro and in vivo models, along with clinical samples, to explore interactions between PACAP and autophagy in AD. METHODS AD model mice were administered 6 μl of 0.1 mg/ml PACAP liquid intranasally for 4 weeks, then subjected to behavioral analyses to assess the benefits of PACAP treatment. The underlying mechanisms of PACAP-induced effects were investigated by methods including real-time quantitative polymerase chain reaction, RNA sequencing, immunofluorescence, and western blotting. Exosomes were extracted from human serum and subjected to enzyme-linked immunosorbent assays to examine autophagy pathways. The clinical and therapeutic implications of PACAP and autophagy were extensively investigated throughout the experiment. RESULTS Impaired autophagy was a critical step in amyloid β (Aβ) and Tau deposition; PACAP enhanced autophagy and attenuated cognitive impairment. RNA sequencing revealed three pathways that may be involved in AD progression: PI3K-AKT, mTOR, and AMPK. In vivo and in vitro studies showed that sirtuin3 knockdown diminished the ability of PACAP to restore normal autophagy function, resulting in phagocytosis dysregulation and the accumulation of pTau, Tau, and Aβ. Additionally, the autophagic biomarker MAP1LC3 demonstrated a positive association with PACAP in human serum. CONCLUSIONS PACAP reverses AD-induced cognitive impairment through autophagy, using sirtuin3 as a key mediator. MAP1LC3 has a positive relationship with PACAP in humans. These findings provide insights regarding potential uses of intranasal PACAP and sirtuin3 agonists in AD treatment. TRIAL REGISTRATION NCT04320368.
Collapse
Affiliation(s)
- Qing Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, No. 119, South 4Th Ring West Road, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Yue Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, No. 119, South 4Th Ring West Road, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Shiping Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, No. 119, South 4Th Ring West Road, Fengtai District, Beijing, 100070, China.
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
| | - Jiong Shi
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, No. 119, South 4Th Ring West Road, Fengtai District, Beijing, 100070, China.
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
| |
Collapse
|
3
|
Wu Y, Guo S, Wang K, Kang J. The interaction of peptide inhibitors and Aβ protein: Binding mode analysis, inhibition of the formation of Aβ aggregates, and then exert neuroprotective effects. Front Aging Neurosci 2023; 15:1139418. [PMID: 37113572 PMCID: PMC10126514 DOI: 10.3389/fnagi.2023.1139418] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 03/20/2023] [Indexed: 04/29/2023] Open
Abstract
Introduction The misfolding and aggregation of β-amyloid (Aβ) easily form Aβ fibers, which are continuously deposited in the brain, leading to the massive generation of amyloid plaques, severely destroying neuronal connections, and promoting Alzheimer's disease (AD) The occurrence and development of AD is one of the pathogenesis of AD. There is an urgent need to develop inhibitors against Aβ aggregation, which is hopefully a potential way to treat AD. Methods In this study, we first found the crystal structure of the Aβ1-42 receptor protein from the RCSB PDB protein structure database and used the SYBYL X2.0 software for molecular docking, and then used the Peptide Ranker, Innovagen, DPL, and ToxinPred online websites to perform peptides. Predict the activity score, toxicity and water solubility, and then calculate the affinity constant KD value of polypeptide and Aβ through Surface Plasmon Resonance (SPR) experiment. Subsequently, the CCK-8 kit method was used to determine the toxicity of different concentrations of peptides (3.125, 6.25, 12.5, 25, 50, 100, 200 μM) to PC12 cells, and then the peptides and Aβ according to different concentration ratios (1:4, 1:2, 1:1, 1:0.5, 1:0.25, 0:4), this method is also used to detect the effect of peptides on Aβ-induced neurotoxicity. The thioflavin T (ThT) fluorescence method was used to detect the effects of peptides (50 μM) on Aβ (25 μM) aggregation inhibitory effect. Results The results showed that the CScore of YVRHLKYVRHLK peptide molecule docking was 10.0608, the predicted activity score was 0.20, and the KD value was 5.385 × 10-5. The ThT and CCK-8 kit method found that the peptide itself is less toxic to PC12 cells at a concentration of 50 μM, and it has a significant inhibitory effect on the formation of Aβ1-42 aggregates when incubated with Aβ1-42 at a ratio of 1:1 (p < 0.05) and can significantly reduce the PC12 cytotoxicity induced by Aβ1-42 (p < 0.05). Conclusion In conclusion, the polypeptide YVRHLKYVRHLK designed in this study has a neuroprotective effect on PC12 cytotoxicity induced by Aβ1-42. Graphical Abstract.
Collapse
Affiliation(s)
- Yuchen Wu
- College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, Zhengzhou, China
| | - Shuang Guo
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Kunli Wang
- College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, Zhengzhou, China
| | - Jingjing Kang
- College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, Zhengzhou, China
- *Correspondence: Jingjing Kang,
| |
Collapse
|
4
|
Gopalakrishna R, Lin CY, Oh A, Le C, Yang S, Hicks A, Kindy MS, Mack WJ, Bhat NR. cAMP-induced decrease in cell-surface laminin receptor and cellular prion protein attenuates amyloid-β uptake and amyloid-β-induced neuronal cell death. FEBS Lett 2022; 596:2914-2927. [PMID: 35971617 PMCID: PMC9712173 DOI: 10.1002/1873-3468.14467] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/21/2022] [Accepted: 07/31/2022] [Indexed: 02/07/2023]
Abstract
Previous studies have shown that amyloid-β oligomers (AβO) bind with high affinity to cellular prion protein (PrPC ). The AβO-PrPC complex binds to cell-surface co-receptors, including the laminin receptor (67LR). Our current studies revealed that in Neuroscreen-1 cells, 67LR is the major co-receptor involved in the cellular uptake of AβO and AβΟ-induced cell death. Both pharmacological (dibutyryl-cAMP, forskolin and rolipram) and physiological (pituitary adenylate cyclase-activating polypeptide) cAMP-elevating agents decreased cell-surface PrPC and 67LR, thereby attenuating the uptake of AβO and the resultant neuronal cell death. These cAMP protective effects are dependent on protein kinase A, but not dependent on the exchange protein directly activated by cAMP. Conceivably, cAMP protects neuronal cells from AβO-induced cytotoxicity by decreasing cell-surface-associated PrPC and 67LR.
Collapse
Affiliation(s)
- Rayudu Gopalakrishna
- Department of Integrative Anatomical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA,Corresponding author: Department of Integrative Anatomical Sciences, 1333 San Pablo Street, Keck School of Medicine, Los Angeles, CA 90089, USA, Phone: 1 + 323-442-1770; Fax: 1 + 323-442-1771:
| | - Charlotte Y. Lin
- Department of Integrative Anatomical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Andrew Oh
- Department of Integrative Anatomical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Calvin Le
- Department of Integrative Anatomical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Seolyn Yang
- Department of Integrative Anatomical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Alexandra Hicks
- Department of Integrative Anatomical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Mark S. Kindy
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA; James A. Haley VA Medical Center, Tampa, FL 33612, USA
| | - William J. Mack
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Narayan R. Bhat
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
5
|
Tian Y, Lu Y, Cao Y, Dang C, Wang N, Tian K, Luo Q, Guo E, Luo S, Wang L, Li Q. Identification of diagnostic signatures associated with immune infiltration in Alzheimer’s disease by integrating bioinformatic analysis and machine-learning strategies. Front Aging Neurosci 2022; 14:919614. [PMID: 35966794 PMCID: PMC9372364 DOI: 10.3389/fnagi.2022.919614] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 07/11/2022] [Indexed: 12/04/2022] Open
Abstract
Objective As a chronic neurodegenerative disorder, Alzheimer’s disease (AD) is the most common form of progressive dementia. The purpose of this study was to identify diagnostic signatures of AD and the effect of immune cell infiltration in this pathology. Methods The expression profiles of GSE109887, GSE122063, GSE28146, and GSE1297 were downloaded from the Gene Expression Omnibus (GEO) database to obtain differentially expressed genes (DEGs) between AD and control brain samples. Functional enrichment analysis was performed to reveal AD-associated biological functions and key pathways. Besides, we applied the Least Absolute Shrinkage Selection Operator (LASSO) and support vector machine-recursive feature elimination (SVM-RFE) analysis to screen potential diagnostic feature genes in AD, which were further tested in AD brains of the validation cohort (GSE5281). The discriminatory ability was then assessed by the area under the receiver operating characteristic curves (AUC). Finally, the CIBERSORT algorithm and immune cell infiltration analysis were employed to assess the inflammatory state of AD. Results A total of 49 DEGs were identified. The functional enrichment analysis revealed that leukocyte transendothelial migration, cytokine receptor interaction, and JAK-STAT signaling pathway were enriched in the AD group. MAF basic leucine zipper transcription factor F (MAFF), ADCYAP1, and ZFP36L1 were identified as the diagnostic biomarkers of AD with high discriminatory ability (AUC = 0.850) and validated in AD brains (AUC = 0.935). As indicated from the immune cell infiltration analysis, naive B cells, plasma cells, activated/resting NK cells, M0 macrophages, M1 macrophages, resting CD4+ T memory cells, resting mast cells, memory B cells, and resting/activated dendritic cells may participate in the development of AD. Additionally, all diagnostic signatures presented different degrees of correlation with different infiltrating immune cells. Conclusion MAFF, ADCYAP1, and ZFP36L1 may become new candidate biomarkers of AD, which were closely related to the pathogenesis of AD. Moreover, the immune cells mentioned above may play crucial roles in disease occurrence and progression.
Collapse
Affiliation(s)
- Yu Tian
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Gerontology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yaoheng Lu
- Department of General Surgery, Chengdu Integrated Traditional Chinese Medicine and Western Medicine Hospital, Chengdu, China
| | - Yuze Cao
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Chun Dang
- West China Medical Publishers, West China Hospital, Sichuan University, Chengdu, China
| | - Na Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kuo Tian
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qiqi Luo
- Department of Gerontology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Erliang Guo
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shanshun Luo
- Department of Gerontology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Shanshun Luo,
| | - Lihua Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Lihua Wang,
| | - Qian Li
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- *Correspondence: Qian Li,
| |
Collapse
|
6
|
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
|
7
|
Sunkaria A, Bhardwaj S. Sleep Disturbance and Alzheimer's Disease: The Glial Connection. Neurochem Res 2022; 47:1799-1815. [PMID: 35303225 DOI: 10.1007/s11064-022-03578-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/27/2022] [Accepted: 03/09/2022] [Indexed: 12/28/2022]
Abstract
Poor quality and quantity of sleep are very common in elderly people throughout the world. Growing evidence has suggested that sleep disturbances could accelerate the process of neurodegeneration. Recent reports have shown a positive correlation between sleep deprivation and amyloid-β (Aβ)/tau aggregation in the brain of Alzheimer's patients. Glial cells have long been implicated in the progression of Alzheimer's disease (AD) and recent findings have also suggested their role in regulating sleep homeostasis. However, how glial cells control the sleep-wake balance and exactly how disturbed sleep may act as a trigger for Alzheimer's or other neurological disorders have recently gotten attention. In an attempt to connect the dots, the present review has highlighted the role of glia-derived sleep regulatory molecules in AD pathogenesis. Role of glia in sleep disturbance and Alzheimer's progression.
Collapse
Affiliation(s)
- Aditya Sunkaria
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, Punjab, 143005, India.
| | - Supriya Bhardwaj
- Department of Dermatology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| |
Collapse
|
8
|
Solés-Tarrés I, Cabezas-Llobet N, Lefranc B, Leprince J, Alberch J, Vaudry D, Xifró X. Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) Protects Striatal Cells and Improves Motor Function in Huntington’s Disease Models: Role of PAC1 Receptor. Front Pharmacol 2022; 12:797541. [PMID: 35153755 PMCID: PMC8832515 DOI: 10.3389/fphar.2021.797541] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/31/2021] [Indexed: 12/21/2022] Open
Abstract
Huntington’s disease (HD) is a hereditary neurodegenerative disorder caused by the expression of mutant huntingtin (mHtt). One of the main features of HD is the degeneration of the striatum that leads to motor discoordination. Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide that acts through three receptors named PAC1R, VPAC1R, and VPAC2R. In the present study, we first investigated the effect of PACAP on STHdhQ7/Q7 and STHdhQ111/Q111 cells that express wild-type Htt with 7 and mHtt with 111 glutamines, respectively. Then we explored the capacity of PACAP to rescue motor symptoms in the R6/1, a murine model of HD. We found that PACAP treatment (10–7 M) for 24 h protects STHdhQ111/Q111 cells from mHtt-induced apoptosis. This effect is associated with an increase in PAC1R transcription, phosphorylation of ERK and Akt, and an increase of intracellular c-fos, egr1, CBP, and BDNF protein content. Moreover, the use of pharmacological inhibitors revealed that activation of ERK and Akt mediates these antiapoptotic and neurotrophic effects of PACAP. To find out PAC1R implication, we treated STHdh cells with vasoactive intestinal peptide (VIP), which exhibits equal affinity for VPAC1R and VPAC2R, but lower affinity for PAC1R, in contrast to PACAP which has same affinity for the three receptors. VIP reduced cleaved caspase-3 protein level, without promoting the expression of c-fos, egr1, CBP, and the neurotrophin BDNF. We next measured the protein level of PACAP receptors in the striatum and cortex of R6/1 mice. We observed a specific reduction of PAC1R at the onset of motor symptoms. Importantly, the intranasal administration of PACAP to R6/1 animals restored the motor function and increased the striatal levels of PAC1R, CBP, and BDNF. In conclusion, PACAP exerts antiapoptotic and neurotrophic effects in striatal neurons mainly through PAC1R. This effect in HD striatum allows the recovery of motor function and point out PAC1R as a therapeutic target for treatment of HD.
Collapse
Affiliation(s)
- Irene Solés-Tarrés
- New Therapeutic Targets Group, Department of Medical Science, Faculty of Medicine, University of Girona, Girona, Spain
| | - Núria Cabezas-Llobet
- New Therapeutic Targets Group, Department of Medical Science, Faculty of Medicine, University of Girona, Girona, Spain
| | - Benjamin Lefranc
- Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal Death and Cell Plasticity Team, UNIROUEN, Inserm, Normandie University, Rouen, France
- Regional Cell Imaging Platform of Normandy (PRIMACEN), UNIROUEN, Normandie University, Rouen, France
| | - Jérôme Leprince
- Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal Death and Cell Plasticity Team, UNIROUEN, Inserm, Normandie University, Rouen, France
- Regional Cell Imaging Platform of Normandy (PRIMACEN), UNIROUEN, Normandie University, Rouen, France
| | - Jordi Alberch
- Departament de Biomedicina, Institut de Neurociències, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
- Institut D’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - David Vaudry
- Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal Death and Cell Plasticity Team, UNIROUEN, Inserm, Normandie University, Rouen, France
- Regional Cell Imaging Platform of Normandy (PRIMACEN), UNIROUEN, Normandie University, Rouen, France
| | - Xavier Xifró
- New Therapeutic Targets Group, Department of Medical Science, Faculty of Medicine, University of Girona, Girona, Spain
- *Correspondence: Xavier Xifró,
| |
Collapse
|
9
|
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
|
10
|
Hu S, Huang S, Ma J, Li D, Zhao Z, Zheng J, Li M, Wang Z, Sun W, Shi X. Correlation of Decreased Serum Pituitary Adenylate Cyclase-Activating Polypeptide and Vasoactive Intestinal Peptide Levels With Non-motor Symptoms in Patients With Parkinson's Disease. Front Aging Neurosci 2021; 13:689939. [PMID: 34566619 PMCID: PMC8457255 DOI: 10.3389/fnagi.2021.689939] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/16/2021] [Indexed: 12/05/2022] Open
Abstract
Objective: Pituitary adenylate-cyclase activating polypeptide (PACAP) and vasoactive intestinal peptide (VIP) are two neuropeptides that exhibit anti-inflammatory and neuroprotective properties, modulating the production of cytokines and chemokines, and the behavior of immune cells. However, the relationship between PACAP and VIP levels and Parkinson’s disease (PD) are not clear. The aim of the current study was to evaluate serum PACAP and VIP levels in PD patients and to analysis the correlation between neuropeptide levels and non-motor symptoms. Methods: In this cross-sectional study, we enrolled 72 patients with idiopathic PD and 71 healthy volunteers. Serum PACAP and VIP levels were measured using an enzyme-linked immunosorbent assay (ELISA) kit. Non-motor symptoms were assessed with the Non-Motor Symptoms Scale (NMSS) for PD, including total and single-item scores. Results: The serum PACAP levels of PD patients were significantly lower than those of healthy controls [(76.02 ± 43.78) pg/ml vs. (154.96 ± 76.54) pg/ml, P < 0.001]; and the serum VIP levels of PD patients were also significantly lower than those of healthy controls [(109.56 ± 15.39) pg/ml vs. (136.46 ± 24.16) pg/ml, P < 0.001]. PACAP levels were inversely correlated only with the score on NMSS item five, assessing Attention/memory (r = −0.276, P < 0.05) and lower serum PACAP levels were detected in the cognitive dysfunction subgroup than in the cognitively intact subgroup [(61.87 ± 32.66) pg/ml vs. (84.51 ± 47.59) pg/ml, P < 0.05]; meanwhile, VIP levels were inversely correlated with the NMSS total score (r = −0.285, P < 0.05) and the single-item scores for item one, assessing Cardiovascular (r = −0.257, P < 0.05) and item three, assessing Mood/cognition (r = −0.373, P < 0.05), and lower serum VIP levels were detected in the anxiety subgroup and depression subgroup than in the non-anxiety subgroup and non-depression subgroup, respectively [(107.45 ± 15.40) pg/ml vs. (116.41 ± 13.67) pg/ml, P < 0.05]; [(104.45 ± 15.26) pg/ml vs. (113.43 ± 14.52) pg/ml, P < 0.05]. Conclusion: The serum PACAP and VIP levels of PD patients were significantly lower than those of healthy controls. The non-motor symptoms significantly negatively correlated with serum PACAP level was cognitive dysfunction, while mood disorder was significantly correlated with serum VIP level.
Collapse
Affiliation(s)
- Shiyu Hu
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China.,Department of Neurology, People's Hospital of Henan University, Zhengzhou, China
| | - Shen Huang
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China.,Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianjun Ma
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China.,Department of Neurology, People's Hospital of Henan University, Zhengzhou, China.,Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Dongsheng Li
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China.,Department of Neurology, People's Hospital of Henan University, Zhengzhou, China.,Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenxiang Zhao
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China.,Department of Neurology, People's Hospital of Henan University, Zhengzhou, China.,Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Jinhua Zheng
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China.,Department of Neurology, People's Hospital of Henan University, Zhengzhou, China.,Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Mingjian Li
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China.,Department of Neurology, People's Hospital of Henan University, Zhengzhou, China
| | - Zhidong Wang
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China.,Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenhua Sun
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China.,Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoxue Shi
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China.,Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
11
|
VIP Stabilizes the Cytoskeleton of Schlemm's Canal Endothelia via Reducing Caspase-3 Mediated ZO-1 Endolysosomal Degradation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9397960. [PMID: 34552687 PMCID: PMC8452417 DOI: 10.1155/2021/9397960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/05/2021] [Accepted: 08/23/2021] [Indexed: 11/17/2022]
Abstract
Objectives In glaucomatous eyes, the main aqueous humor (AH) outflow pathway is damaged by accumulated oxidative stress arising from the microenvironment, vascular dysregulation, and aging, which results in increased outflow resistance and ocular hypertension. Schlemm's canal (SC) serves as the final filtration barrier of the main AH outflow pathway. The present study is aimed at investigating the possible regulation of vasoactive intestinal peptide (VIP) on the cytoskeleton by stabilizing ZO-1 in SC. Methods Model of chronic ocular hypertension (COH) induced by episcleral venous cauterization was treated with topical VIP. The ultrastructure of junctions, ZO-1 levels, and permeability of the SC inner wall to FITC-dextran (70 kDa) were detected in the COH models. The F-actin distribution, F/G-actin ratio, and ZO-1 degradation pathway in human umbilical vein endothelial cells (HUVECs) and HEK 293 cells were investigated. Results ZO-1 in the outer wall of the SC was less than that in the inner wall. COH elicited junction disruption, ZO-1 reduction, and increased permeability of the SC inner wall to FITC-dextran in rats. ZO-1 plays an essential role in maintaining the F/G-actin ratio and F-actin distribution. VIP treatment attenuated the downregulation of ZO-1 associated with COH or H2O2-induced oxidative damage. In H2O2-stimulated HUVECs, the caspase-3 inhibitor prevents ZO-1 disruption. Caspase-3 activation promoted endolysosomal degradation of ZO-1. Furthermore, a decrease in caspase-3 activation and cytoskeleton redistribution was demonstrated in VIP + H2O2-treated cells. The knockdown of ZO-1 or the overexpression of caspase-3 blocked the effect of VIP on the cytoskeleton. Conclusion This study provides insights into the role of VIP in stabilizing the interaction between the actin cytoskeleton and cell junctions and may provide a promising targeted strategy for glaucoma treatment.
Collapse
|
12
|
Perlikowska R. Whether short peptides are good candidates for future neuroprotective therapeutics? Peptides 2021; 140:170528. [PMID: 33716091 DOI: 10.1016/j.peptides.2021.170528] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 02/18/2021] [Accepted: 02/27/2021] [Indexed: 02/07/2023]
Abstract
Neurodegenerative diseases are a broad group of largely debilitating, and ultimately terminal conditions resulting in progressive degeneration of different brain regions. The observed damages are associated with cell death, structural and functional deficits of neurons, or demyelination. The concept of neuroprotection concerns the administration of the agent, which should reverse some of the damage or prevent further adverse changes. A growing body of evidence suggested that among many classes of compounds considered as neuroprotective agents, peptides derived from natural materials or their synthetic analogs are good candidates. They presented a broad spectrum of activities and abilities to act through diverse mechanisms of action. Biologically active peptides have many properties, including antioxidant, antimicrobial, antiinflammatory, and immunomodulatory effects. Peptides with pro-survival and neuroprotective activities, associated with inhibition of oxidative stress, apoptosis, inflammation and are able to improve cell viability or mitochondrial functions, are also promising molecules of particular interest to the pharmaceutical industries. Peptide multiple activities open the way for broad application potential as therapeutic agents or ingredients of health-promoting functional foods. Significantly, synthetic peptides can be remodeled in numerous ways to have desired features, such as increased solubility or biological stability, as well as selectivity towards a specific receptor, and finally better membrane penetration. This review summarized the most common features of major neurodegenerative disorders, their causes, consequences, and reported new neuroprotective drug development approaches. The author focused on the unique perspectives in neuroprotection and provided a concise survey of short peptides proposed as novel therapeutic agents against various neurodegenerative diseases.
Collapse
Affiliation(s)
- Renata Perlikowska
- Department of Biomolecular Chemistry, Faculty of Medicine, Medical University of Lodz, 92-215, Lodz, Poland.
| |
Collapse
|
13
|
Ben-Shushan S, Miller Y. Neuropeptides: Roles and Activities as Metal Chelators in Neurodegenerative Diseases. J Phys Chem B 2021; 125:2796-2811. [PMID: 33570949 PMCID: PMC8389909 DOI: 10.1021/acs.jpcb.0c11151] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Neurodegenerative diseases, such as Alzheimer's disease (AD) and Parkinson's disease (PD), are characterized by deposits of amyloid proteins. The homeostasis of metal ions is crucial for the normal biological functions in the brain. However, in AD and PD, the imbalance of metal ions leads to formation of amyloid deposits. In the past four decades, there has been extensive effort to design compound agents than can chelate metal ions with the aim of preventing the formation of the amyloid deposits. Unfortunately, the compounds to date that were designed were not successful candidates to be used in clinical trials. Neuropeptides are small molecules that are produced and released by neurons. It has been shown that neuropeptides have neuroprotective effects in the brain and reduce the formation of amyloid deposits. This Review Article is focused on the function of neuropeptides as metal chelators. Experimental and computational studies demonstrated that neuropeptides could bind metal ions, such as Cu2+ and Zn2+. This Review Article provides perspectives and initiates future studies to investigate the role of neuropeptides as metal chelators in neurodegenerative diseases.
Collapse
Affiliation(s)
- Shira Ben-Shushan
- Department of Chemistry, Ben-Gurion University of the Negev, P.O. Box 653, Be'er Sheva 84105, Israel.,Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Be'er Sheva 84105, Israel
| | - Yifat Miller
- Department of Chemistry, Ben-Gurion University of the Negev, P.O. Box 653, Be'er Sheva 84105, Israel.,Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Be'er Sheva 84105, Israel
| |
Collapse
|
14
|
Ruel J, Guitton MJ, Gratias P, Lenoir M, Shen S, Puel JL, Brabet P, Wang J. Endogenous Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) Plays a Protective Effect Against Noise-Induced Hearing Loss. Front Cell Neurosci 2021; 15:658990. [PMID: 33828461 PMCID: PMC8019930 DOI: 10.3389/fncel.2021.658990] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/02/2021] [Indexed: 01/07/2023] Open
Abstract
Pituitary adenylyl cyclase-activating polypeptide (PACAP) is a member of the vasoactive intestinal polypeptide (VIP)-the secretin-glucagon family of neuropeptides. They act through two classes of receptors: PACAP type 1 (PAC1) and type 2 (VPAC1 and VPAC2). Among their pleiotropic effects throughout the body, PACAP functions as neuromodulators and neuroprotectors, rescuing neurons from apoptosis, mostly through the PAC1 receptor. To explore the potential protective effect of endogenous PACAP against Noise-induced hearing loss (NIHL), we used a knockout mouse model lacking PAC1 receptor expression (PACR1−/−) and a transgenic humanized mouse model expressing the human PAC1 receptor (TgHPAC1R). Based on complementary approaches combining electrophysiological, histochemical, and molecular biological evaluations, we show PAC1R expression in spiral ganglion neurons and in cochlear apical cells of the organ of Corti. Wild-type (WT), PAC1R−/−, and TgHPAC1R mice exhibit similar auditory thresholds. For most of the frequencies tested after acute noise damage, however, PAC1R−/− mice showed a larger elevation of the auditory threshold than did their WT counterparts. By contrast, in a transgene copy number-dependent fashion, TgHPAC1R mice showed smaller noise-induced elevations of auditory thresholds compared to their WT counterparts. Together, these findings suggest that PACAP could be a candidate for endogenous protection against noise-induced hearing loss.
Collapse
Affiliation(s)
- Jérôme Ruel
- Institute for Neurosciences of Montpellier (INM), University Montpellier, INSERM, Montpellier, France.,Laboratoire de Neurosciences Cognitives, UMR7291 CNRS, Aix-Marseille Université, Marseille, France
| | - Matthieu J Guitton
- CERVO Brain Research Center, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| | - Paul Gratias
- Institute for Neurosciences of Montpellier (INM), University Montpellier, INSERM, Montpellier, France
| | - Marc Lenoir
- Institute for Neurosciences of Montpellier (INM), University Montpellier, INSERM, Montpellier, France
| | - Sanbing Shen
- Regenerative Medicine Institute, National University of Ireland (NUI), Galway, Ireland
| | - Jean-Luc Puel
- Institute for Neurosciences of Montpellier (INM), University Montpellier, INSERM, Montpellier, France
| | - Philippe Brabet
- Institute for Neurosciences of Montpellier (INM), University Montpellier, INSERM, Montpellier, France
| | - Jing Wang
- Institute for Neurosciences of Montpellier (INM), University Montpellier, INSERM, Montpellier, France
| |
Collapse
|
15
|
Naito A. Fibril Formation by Glucagon in Solution and in Membrane Environments. Mol Pharmacol 2020. [DOI: 10.5772/intechopen.91681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Glucagon is a 29-amino acid peptide hormone secreted by pancreatic α-cells and interacts with specific receptors located in various organs. Glucagon tends to form gel-like fibril aggregates that are cytotoxic because they activate apoptotic signaling pathways. First, fibril formation by glucagon in acidic solution is discussed in light of morphological and structural changes during elapsed time. Second, we provide kinetic analyses using a two-step autocatalytic reaction mechanism; the first step is a homogeneous nuclear formation process, and the second step is an autocatalytic heterogeneous fibril elongation process. Third, the processes of fibril formation by glucagon in a membrane environment are discussed based on the structural changes in the fibrils. In the presence of bicelles in acidic solution, glucagon interacts with the bicelles and forms fibril intermediates on the bicelle surface and grows into elongated fibrils. Glucagon-dimyristoylphosphatidylcholine (DMPC) bilayers in neutral solution mimic the environment for fibril formation by glucagon under near-physiological condition. Under these conditions, glucagon forms fibril intermediates that grow into elongated fibrils inside the lipid bilayer. Many days after preparing the glucagon-DMPC bilayer sample, the fibrils form networks inside and outside the bilayer. Furthermore, fibril intermediates strongly interact with lipid bilayers to form small particles.
Collapse
|
16
|
Solés-Tarrés I, Cabezas-Llobet N, Vaudry D, Xifró X. Protective Effects of Pituitary Adenylate Cyclase-Activating Polypeptide and Vasoactive Intestinal Peptide Against Cognitive Decline in Neurodegenerative Diseases. Front Cell Neurosci 2020; 14:221. [PMID: 32765225 PMCID: PMC7380167 DOI: 10.3389/fncel.2020.00221] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 06/22/2020] [Indexed: 12/23/2022] Open
Abstract
Cognitive impairment is one of the major symptoms in most neurodegenerative disorders such as Alzheimer’s (AD), Parkinson (PD), and Huntington diseases (HD), affecting millions of people worldwide. Unfortunately, there is no treatment to cure or prevent the progression of those diseases. Cognitive impairment has been related to neuronal cell death and/or synaptic plasticity alteration in important brain regions, such as the cerebral cortex, substantia nigra, striatum, and hippocampus. Therefore, compounds that can act to protect the neuronal loss and/or to reestablish the synaptic activity are needed to prevent cognitive decline in neurodegenerative diseases. Pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal peptide (VIP) are two highly related multifunctional neuropeptides widely distributed in the central nervous system (CNS). PACAP and VIP exert their action through two common receptors, VPAC1 and VPAC2, while PACAP has an additional specific receptor, PAC1. In this review article, we first presented evidence showing the therapeutic potential of PACAP and VIP to fight the cognitive decline observed in models of AD, PD, and HD. We also reviewed the main transduction pathways activated by PACAP and VIP receptors to reduce cognitive dysfunction. Furthermore, we identified the therapeutic targets of PACAP and VIP, and finally, we evaluated different novel synthetic PACAP and VIP analogs as promising pharmacological tools.
Collapse
Affiliation(s)
- Irene Solés-Tarrés
- New Therapeutic Targets Group (TargetsLab), Department of Medical Science, Faculty of Medicine, Universitat de Girona, Girona, Spain
| | - Núria Cabezas-Llobet
- New Therapeutic Targets Group (TargetsLab), Department of Medical Science, Faculty of Medicine, Universitat de Girona, Girona, Spain
| | - David Vaudry
- Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal Death and Cell Plasticity Team, Normandie University, UNIROUEN, Inserm, Rouen, France
| | - Xavier Xifró
- New Therapeutic Targets Group (TargetsLab), Department of Medical Science, Faculty of Medicine, Universitat de Girona, Girona, Spain
| |
Collapse
|
17
|
Farkhondeh T, Khan H, Aschner M, Samini F, Pourbagher-Shahri AM, Aramjoo H, Roshanravan B, Hoyte C, Mehrpour O, Samarghandian S. Impact of Cannabis-Based Medicine on Alzheimer's Disease by Focusing on the Amyloid β-Modifications: A Systematic Study. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2020; 19:334-343. [PMID: 32640965 DOI: 10.2174/1871527319666200708130745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/22/2020] [Accepted: 06/07/2020] [Indexed: 01/02/2023]
Abstract
Deposition of Amyloid-beta (Aβ) peptide in the brain is the leading source of the onset and progression of Alzheimer's Disease (AD). Recent studies have suggested that anti-amyloidogenic agents may be a suitable therapeutic strategy for AD. The current review was proposed to address the beneficial effects of cannabis-based drugs for the treatment of AD, focusing primarily on Aβ modifications. Keywords related to AD, Aβ, and cannabis-based on MeSH were identified and were searched in PubMed, Google Scholar, Scopus, Ovid-Medline, and Web of Science from inception until 15 March 2020. The full text of identified papers was obtained and assessed based on exclusion and inclusion criteria. The review is based on articles that have focused on AD and the amyloidogenic pathway. A total of 17 studies were identified based on the inclusion criteria; however, nine studies qualified for this systematic review. The maximum and minimum cannabis dosages, mostly CBD and THC in animal studies, were 0.75 and 50 mg/kg, respectively. Cannabis (CBD and THC) was injected for 10 to 21 days. The findings of the 9 articles indicated that cannabis-based drugs might modulate Aβ modifications in several AD models. Our findings establish that cannabis-based drugs inhibited the progression of AD by modulating Aβ modifications.
Collapse
Affiliation(s)
- Tahereh Farkhondeh
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences (BUMS), Birjand, Iran
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Forchheimer 2091300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Fariborz Samini
- Department of Neurosurgery, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Hamed Aramjoo
- Student Research Committee, BSc Student in Lab Sciences Technology, Birjand University of Medical Sciences, Birjand, Iran
| | - Babak Roshanravan
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
| | | | - Omid Mehrpour
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences (BUMS), Birjand, Iran,Arizona Poison & Drug Information Center, the University of Arizona, College of Pharmacy, Tucson, Arizona, AZ, USA
| | - Saeed Samarghandian
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences,
Neyshabur, Iran
| |
Collapse
|
18
|
Liao C, de Molliens MP, Schneebeli ST, Brewer M, Song G, Chatenet D, Braas KM, May V, Li J. Targeting the PAC1 Receptor for Neurological and Metabolic Disorders. Curr Top Med Chem 2019; 19:1399-1417. [PMID: 31284862 PMCID: PMC6761004 DOI: 10.2174/1568026619666190709092647] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 12/23/2018] [Accepted: 12/26/2018] [Indexed: 12/16/2022]
Abstract
The pituitary adenylate cyclase-activating polypeptide (PACAP)-selective PAC1 receptor (PAC1R, ADCYAP1R1) is a member of the vasoactive intestinal peptide (VIP)/secretin/glucagon family of G protein-coupled receptors (GPCRs). PAC1R has been shown to play crucial roles in the central and peripheral nervous systems. The activation of PAC1R initiates diverse downstream signal transduction pathways, including adenylyl cyclase, phospholipase C, MEK/ERK, and Akt pathways that regulate a number of physiological systems to maintain functional homeostasis. Accordingly, at times of tissue injury or insult, PACAP/PAC1R activation of these pathways can be trophic to blunt or delay apoptotic events and enhance cell survival. Enhancing PAC1R signaling under these conditions has the potential to mitigate cellular damages associated with cerebrovascular trauma (including stroke), neurodegeneration (such as Parkinson's and Alzheimer's disease), or peripheral organ insults. Conversely, maladaptive PACAP/PAC1R signaling has been implicated in a number of disorders, including stressrelated psychopathologies (i.e., depression, posttraumatic stress disorder, and related abnormalities), chronic pain and migraine, and metabolic diseases; abrogating PAC1R signaling under these pathological conditions represent opportunities for therapeutic intervention. Given the diverse PAC1R-mediated biological activities, the receptor has emerged as a relevant pharmaceutical target. In this review, we first describe the current knowledge regarding the molecular structure, dynamics, and function of PAC1R. Then, we discuss the roles of PACAP and PAC1R in the activation of a variety of signaling cascades related to the physiology and diseases of the nervous system. Lastly, we examine current drug design and development of peptides and small molecules targeting PAC1R based on a number of structure- activity relationship studies and key pharmacophore elements. At present, the rational design of PAC1R-selective peptide or small-molecule therapeutics is largely hindered by the lack of structural information regarding PAC1R activation mechanisms, the PACAP-PAC1R interface, and the core segments involved in receptor activation. Understanding the molecular basis governing the PACAP interactions with its different cognate receptors will undoubtedly provide a basis for the development and/or refinement of receptor-selective therapeutics.
Collapse
Affiliation(s)
- Chenyi Liao
- Department of Chemistry, University of Vermont, Burlington, VT 05405, United States
| | | | - Severin T Schneebeli
- Department of Chemistry, University of Vermont, Burlington, VT 05405, United States
| | - Matthias Brewer
- Department of Chemistry, University of Vermont, Burlington, VT 05405, United States
| | - Gaojie Song
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - David Chatenet
- INRS - Institut Armand-Frappier, 531 boul. des Prairies, Laval, QC H7V 1B7, Canada
| | - Karen M Braas
- Department of Neurological Sciences, University of Vermont, Larner College of Medicine, 149 Beaumont Avenue, Burlington, VT 05405, United States
| | - Victor May
- Department of Neurological Sciences, University of Vermont, Larner College of Medicine, 149 Beaumont Avenue, Burlington, VT 05405, United States
| | - Jianing Li
- Department of Chemistry, University of Vermont, Burlington, VT 05405, United States
| |
Collapse
|
19
|
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
|
20
|
Poujol de Molliens M, Jamadagni P, Létourneau M, Devost D, Hébert TE, Patten SA, Fournier A, Chatenet D. Design and biological assessment of membrane-tethering neuroprotective peptides derived from the pituitary adenylate cyclase-activating polypeptide type 1 receptor. Biochim Biophys Acta Gen Subj 2019; 1863:129398. [PMID: 31306709 DOI: 10.1016/j.bbagen.2019.07.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/03/2019] [Accepted: 07/10/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND The pituitary adenylate cyclase-activating polypeptide (PACAP) type 1 receptor (PAC1), a class B G protein-coupled receptor (GPCR), has emerged as a promising target for treating neurodegenerative conditions. Unfortunately, despite years of research, no PAC1-specific agonist has been discovered, as activity on two other GPCRs, VPAC1 and VPAC2, is retained with current analogs. Cell signaling is related to structural modifications in the intracellular loops (ICLs) of GPCRs. Thus, we hypothesized that peptides derived from the ICLs (called pepducins) of PAC1 might initiate, as allosteric ligands, signaling cascades after recognition of the parent receptor and modulation of its conformational landscape. METHODS Three pepducins were synthesized and evaluated for their ability to 1) promote cell survival; 2) stimulate various signaling pathways associated with PAC1 activation; 3) modulate selectively PAC1, VPAC1 or VPAC2 activation; and 4) sustain mobility and prevent death of dopaminergic neurons in a zebrafish model of neurodegeneration. RESULTS Assays demonstrated that these molecules promote SH-SY5Y cell survival, a human neuroblastoma cell line expressing PAC1, and activate signaling via Gαs and Gαq, with distinct potencies and efficacies. Also, PAC1-Pep1 and PAC1-Pep2 activated selectively PAC1-mediated Gαs stimulation. Finally, experiments, using a zebrafish neurodegeneration model, showed a neuroprotective action with all three pepducins and in particular, revealed the ability of PAC1-Pep1 and PAC1-Pep3 to preserve fish mobility and tyrosine hydroxylase expression in the brain. CONCLUSION We have developed the first neuroprotective pepducins derived from PAC1, a class B GPCR. GENERAL SIGNIFICANCE PAC1-derived pepducins represent attractive templates for the development of innovative neuroprotecting molecules.
Collapse
Affiliation(s)
- Mathilde Poujol de Molliens
- Institut National de la Recherche Scientifique, Centre Armand-Frappier, Groupe de Recherche en Ingénierie des Peptides et en Pharmacothérapie (GRIPP), Université du Québec, Ville de Laval, QC, Canada; Institut National de la Recherche Scientifique, Centre Armand-Frappier, Laboratoire d'études moléculaires et pharmacologiques des peptides (LEMPP), Université du Québec, Ville de Laval, QC, Canada
| | - Priyanka Jamadagni
- Institut National de la Recherche Scientifique, Centre Armand-Frappier, Université du Québec, Ville de Laval, QC, Canada
| | - Myriam Létourneau
- Institut National de la Recherche Scientifique, Centre Armand-Frappier, Groupe de Recherche en Ingénierie des Peptides et en Pharmacothérapie (GRIPP), Université du Québec, Ville de Laval, QC, Canada; Institut National de la Recherche Scientifique, Centre Armand-Frappier, Laboratoire d'études moléculaires et pharmacologiques des peptides (LEMPP), Université du Québec, Ville de Laval, QC, Canada
| | - Dominic Devost
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
| | - Shunmoogum A Patten
- Institut National de la Recherche Scientifique, Centre Armand-Frappier, Université du Québec, Ville de Laval, QC, Canada
| | - Alain Fournier
- Institut National de la Recherche Scientifique, Centre Armand-Frappier, Laboratoire d'études moléculaires et pharmacologiques des peptides (LEMPP), Université du Québec, Ville de Laval, QC, Canada
| | - David Chatenet
- Institut National de la Recherche Scientifique, Centre Armand-Frappier, Groupe de Recherche en Ingénierie des Peptides et en Pharmacothérapie (GRIPP), Université du Québec, Ville de Laval, QC, Canada.
| |
Collapse
|
21
|
Chen XY, Du YF, Chen L. Neuropeptides Exert Neuroprotective Effects in Alzheimer's Disease. Front Mol Neurosci 2019; 11:493. [PMID: 30687008 PMCID: PMC6336706 DOI: 10.3389/fnmol.2018.00493] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 12/21/2018] [Indexed: 01/03/2023] Open
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disorder characterized by cognitive deficits and neuronal loss. Deposition of beta-amyloid peptide (Aβ) causes neurotoxicity through the formation of plaques in brains of Alzheimer's disease. Numerous studies have indicated that the neuropeptides including ghrelin, neurotensin, pituitary adenylate cyclase-activating polypeptide (PACAP), neuropeptide Y, substance P and orexin are closely related to the pathophysiology of Alzheimer's disease. The levels of neuropeptides and their receptors change in Alzheimer's disease. These neuropeptides exert neuroprotective roles mainly through preventing Aβ accumulation, increasing neuronal glucose transport, increasing the production of neurotrophins, inhibiting endoplasmic reticulum stress and autophagy, modulating potassium channel activity and hippocampal long-term potentiation. Therefore, the neuropeptides may function as potential drug targets in the prevention and cure of Alzheimer's disease.
Collapse
Affiliation(s)
- Xin-Yi Chen
- Department of Physiology and Pathophysiology, Qingdao University, Qingdao, China.,Department of Neurology, Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Yi-Feng Du
- Department of Neurology, Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Lei Chen
- Department of Physiology and Pathophysiology, Qingdao University, Qingdao, China
| |
Collapse
|
22
|
Korkmaz OT, Ay H, Aytan N, Carreras I, Kowall NW, Dedeoglu A, Tuncel N. Vasoactive Intestinal Peptide Decreases β-Amyloid Accumulation and Prevents Brain Atrophy in the 5xFAD Mouse Model of Alzheimer's Disease. J Mol Neurosci 2018; 68:389-396. [PMID: 30498985 DOI: 10.1007/s12031-018-1226-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 11/21/2018] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by extracellular deposits of fibrillary β-amyloid (Aβ) plaques in the brain that initiate an inflammatory process resulting in neurodegeneration. The neuronal loss associated with AD results in gross atrophy of affected regions causing a progressive loss of cognitive ability and memory function, ultimately leading to dementia. Growing evidence suggests that vasoactive intestinal peptide (VIP) could be beneficial for various neurodegenerative diseases, including AD. The study investigated the effects of VIP on 5xFAD, a transgenic mouse model of AD. Toward this aim, we used 20 5xFAD mice in two groups (n = 10 each), VIP-treated (25 ng/kg i.p. injection, three times per week) and saline-treated (the drug's vehicle) following the same administration regimen. Treatment started at 1 month of age and ended 2 months later. After 2 months of treatment, the mice were euthanized, their brains dissected out, and immunohistochemically stained for Aβ40 and Aβ42 on serial sections. Then, plaque analysis and stereological morphometric analysis were performed in different brain regions. Chronic VIP administration in 5xFAD mice significantly decreased the levels of Aβ40 and Aβ42 plaques in the subiculum compared to the saline treated 5xFAD mice. VIP treatment also significantly decreased Aβ40 and Aβ42 plaques in cortical areas and significantly increased the hippocampus/cerebrum and corpus callosum/cerebrum ratio but not the cerebral cortex/cerebrum ratio. In summary, we found that chronic administration of VIP significantly decreased Aβ plaques and preserved against atrophy for related brain regions in 5xFAD AD mice.
Collapse
Affiliation(s)
- Orhan Tansel Korkmaz
- Research and Development, VA Boston Healthcare System, Boston, MA, 02130, USA. .,Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA. .,Department of Physiology, Faculty of Medicine, Eskisehir Osmangazi University, 26480, Eskisehir, Turkey. .,Faculty of Medicine, Department of Physiology, Eskisehir Osmangazi University, Odunpazari, 26040, Eskisehir, Turkey.
| | - Hakan Ay
- Department of Anatomy, Faculty of Medicine, Eskisehir Osmangazi University, 26040, Eskisehir, Turkey
| | - Nurgul Aytan
- Research and Development, VA Boston Healthcare System, Boston, MA, 02130, USA.,Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Isabel Carreras
- Research and Development, VA Boston Healthcare System, Boston, MA, 02130, USA.,Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA.,Department of Biochemistry, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Neil W Kowall
- Research and Development, VA Boston Healthcare System, Boston, MA, 02130, USA.,Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Alpaslan Dedeoglu
- Research and Development, VA Boston Healthcare System, Boston, MA, 02130, USA.,Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA.,Department of Radiology, MGH and Harvard Medical School, Boston, MA, 02114, USA
| | - Nese Tuncel
- Department of Physiology, Faculty of Medicine, Eskisehir Osmangazi University, 26480, Eskisehir, Turkey
| |
Collapse
|
23
|
AMPK Activation of PGC-1α/NRF-1-Dependent SELENOT Gene Transcription Promotes PACAP-Induced Neuroendocrine Cell Differentiation Through Tolerance to Oxidative Stress. Mol Neurobiol 2018; 56:4086-4101. [DOI: 10.1007/s12035-018-1352-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 09/13/2018] [Indexed: 12/19/2022]
|
24
|
Reglodi D, Jungling A, Longuespée R, Kriegsmann J, Casadonte R, Kriegsmann M, Juhasz T, Bardosi S, Tamas A, Fulop BD, Kovacs K, Nagy Z, Sparks J, Miseta A, Mazzucchelli G, Hashimoto H, Bardosi A. Accelerated pre-senile systemic amyloidosis in PACAP knockout mice - a protective role of PACAP in age-related degenerative processes. J Pathol 2018; 245:478-490. [PMID: 29774542 PMCID: PMC6055756 DOI: 10.1002/path.5100] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 04/10/2018] [Accepted: 05/12/2018] [Indexed: 12/14/2022]
Abstract
Dysregulation of neuropeptides may play an important role in aging‐induced impairments. Among them, pituitary adenylate cyclase‐activating polypeptide (PACAP) is a potent cytoprotective peptide that provides an endogenous control against a variety of tissue‐damaging stimuli. We hypothesized that the progressive decline of PACAP throughout life and the well‐known general cytoprotective effects of PACAP lead to age‐related pathophysiological changes in PACAP deficiency, supported by the increased vulnerability to various stressors of animals partially or totally lacking PACAP. Using young and aging CD1 PACAP knockout (KO) and wild type (WT) mice, we demonstrated pre‐senile amyloidosis in young PACAP KO animals and showed that senile amyloidosis appeared accelerated, more generalized, more severe, and affected more individuals. Histopathology showed age‐related systemic amyloidosis with mainly kidney, spleen, liver, skin, thyroid, intestinal, tracheal, and esophageal involvement. Mass spectrometry‐based proteomic analysis, reconfirmed with immunohistochemistry, revealed that apolipoprotein‐AIV was the main amyloid protein in the deposits together with several accompanying proteins. Although the local amyloidogenic protein expression was disturbed in KO animals, no difference was found in laboratory lipid parameters, suggesting a complex pathway leading to increased age‐related degeneration with amyloid deposits in the absence of PACAP. In spite of no marked inflammatory histological changes or blood test parameters, we detected a disturbed cytokine profile that possibly creates a pro‐inflammatory milieu favoring amyloid deposition. In summary, here we describe accelerated systemic senile amyloidosis in PACAP gene‐deficient mice, which might indicate an early aging phenomenon in this mouse strain. Thus, PACAP KO mice could serve as a model of accelerated aging with human relevance. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Dora Reglodi
- Department of Anatomy, MTA-PTE PACAP Research Group, University of Pecs Medical School, Pécs, Hungary
| | - Adel Jungling
- Department of Anatomy, MTA-PTE PACAP Research Group, University of Pecs Medical School, Pécs, Hungary
| | - Rémi Longuespée
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Joerg Kriegsmann
- Center for Histology, Cytology and Molecular Diagnostics, Trier, Germany.,Proteopath GmbH, Trier, Germany
| | | | - Mark Kriegsmann
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Tamas Juhasz
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Hungary
| | - Sebastian Bardosi
- Center for Histology, Cytology and Molecular Diagnostics, Trier, Germany
| | - Andrea Tamas
- Department of Anatomy, MTA-PTE PACAP Research Group, University of Pecs Medical School, Pécs, Hungary
| | - Balazs Daniel Fulop
- Department of Anatomy, MTA-PTE PACAP Research Group, University of Pecs Medical School, Pécs, Hungary
| | - Krisztina Kovacs
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, Pécs, Hungary
| | - Zsuzsanna Nagy
- Second Department of Internal Medicine, University of Pecs Medical School, Pécs, Hungary
| | - Jason Sparks
- Department of Anatomy, MTA-PTE PACAP Research Group, University of Pecs Medical School, Pécs, Hungary
| | - Attila Miseta
- Department of Laboratory Medicine and Szentagothai Research Centre, University of Pecs Medical School, Pécs, Hungary
| | - Gabriel Mazzucchelli
- Laboratory of Mass Spectrometry (LSM) - MolSys, Department of Chemistry, University of Liège, Belgium
| | - Hitoshi Hashimoto
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Japan
| | - Attila Bardosi
- Center for Histology, Cytology and Molecular Diagnostics, Trier, Germany
| |
Collapse
|
25
|
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]
|
26
|
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
|
27
|
Saleem S, Biswas SC. Tribbles Pseudokinase 3 Induces Both Apoptosis and Autophagy in Amyloid-β-induced Neuronal Death. J Biol Chem 2016; 292:2571-2585. [PMID: 28011637 DOI: 10.1074/jbc.m116.744730] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 12/22/2016] [Indexed: 11/06/2022] Open
Abstract
Amyloid-β (Aβ)-induced neuron death is considered central to the pathogenesis of Alzheimer's disease (AD). Among several death modalities, autophagy and apoptosis play important roles in Aβ-induced neuron death suggesting that there may be regulatory mechanisms that initiate both cell death pathways. However, molecules that govern both pathways have not been identified. Here, we report that, upon Aβ treatment, tribbles pseudokinase 3 (Trib3, an ortholog of Drosophila Tribbles) is up-regulated in neurons both in vivo and in vitro Increased Trib3 levels inhibited the activity of the kinase Akt by interacting with it. As a result, forkhead box O1 (FoxO1), a transcription factor that is negatively regulated by Akt, was activated, translocated to the nucleus, and induced the pro-apoptotic gene BCL2-like 11 (Bim). Conversely, FoxO1 responded to Aβ insult by binding to the Trib3 gene promoter, enhancing its expression. Our investigations further revealed that Trib3 also induces autophagy. We found that Trib3 indirectly activates unc-51-like autophagy-activating kinase1 (Ulk1) by impeding phosphorylation of, and thus inactivating, a negative regulator of Ulk1, mechanistic target of rapamycin. Ulk1 activation augmented autophagosome formation and reduced autophagy flux. Thus, Trib3 was required for formation of autophagosomes, which accumulated in neurons as autophagic flux was thwarted. Most importantly, silencing endogenous Trib3 strongly protected neurons from Aβ insult. Our results suggest that a self-amplifying feed-forward loop among Trib3, Akt, and FoxO1 in Aβ-treated neurons induces both apoptosis and autophagy, culminating in neuron death. Thus, Trib3 may serve as a potential therapeutic target for AD.
Collapse
Affiliation(s)
- Suraiya Saleem
- From the Cell Biology and Physiology Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, India
| | - Subhas Chandra Biswas
- From the Cell Biology and Physiology Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, India
| |
Collapse
|
28
|
Abstract
Neurodegenerative disorders (NDDs) are characterized by neuronal death in the brain. The mechanism of the neuronal death is too complicated to be fully understood, although in many NDDs, aging and neurotoxins are known risk factors. In the central and peripheral nervous system, vasoactive intestinal peptide (VIP), a 28-amino acid neuropeptide, is released to support neuronal survival in both physiological and pathological condition. VIP can inhibit the neurodegeneration induced by the loss of neurons. The indirect protection effect is mainly mediated by glial cells through the production of neurotrophic factor(s) and inhibition of proinflammatory mediators. By remolding the structure and improving the transfer efficiency of VIP, its nerve protective function could be further improved. Its neuroprotective action and efficacy in inhibiting a broad range of inflammatory responses make VIP or related peptides becoming a novel therapeutic method to NDDs. In this review, we aim to summarize the relationship between VIP and NDDs.
Collapse
Affiliation(s)
- Guangxiu Deng
- a National Glycoengineering Research Center , Shandong University , Jinan , China
| | - Lan Jin
- a National Glycoengineering Research Center , Shandong University , Jinan , China
| |
Collapse
|
29
|
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
|
30
|
Lee EH, Seo SR. Neuroprotective roles of pituitary adenylate cyclase-activating polypeptide in neurodegenerative diseases. BMB Rep 2015; 47:369-75. [PMID: 24856828 PMCID: PMC4163857 DOI: 10.5483/bmbrep.2014.47.7.086] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Indexed: 12/04/2022] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a pleiotropic bioactive peptide that was first isolated from an ovine hypothalamus in 1989. PACAP belongs to the secretin/glucagon/vasoactive intestinal polypeptide (VIP) superfamily. PACAP is widely distributed in the central and peripheral nervous systems and acts as a neurotransmitter, neuromodulator, and neurotrophic factor via three major receptors (PAC1, VPAC1, and VPAC2). Recent studies have shown a neuroprotective role of PACAP using in vitro and in vivo models. In this review, we briefly summarize the current findings on the neurotrophic and neuroprotective effects of PACAP in different brain injury models, such as cerebral ischemia, Parkinson’s disease (PD), and Alzheimer’s disease (AD). This review will provide information for the future development of therapeutic strategies in treatment of these neurodegenerative diseases. [BMB Reports 2014; 47(7): 369-375]
Collapse
Affiliation(s)
- Eun Hye Lee
- Department of Molecular Bioscience, College of Biomedical Science, and Institute of Bioscience & Biotechnology, Kangwon National University, Chuncheon 200-701, Korea
| | - Su Ryeon Seo
- Department of Molecular Bioscience, College of Biomedical Science, and Institute of Bioscience & Biotechnology, Kangwon National University, Chuncheon 200-701, Korea
| |
Collapse
|
31
|
Jóźwiak-Bębenista M, Kowalczyk E, Nowak JZ. The cyclic AMP effects and neuroprotective activities of PACAP and VIP in cultured astrocytes and neurons exposed to oxygen-glucose deprivation. Pharmacol Rep 2014; 67:332-8. [PMID: 25712659 DOI: 10.1016/j.pharep.2014.10.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 09/24/2014] [Accepted: 10/02/2014] [Indexed: 01/30/2023]
Abstract
BACKGROUND Pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal peptide (VIP) are endogenous peptides, widely expressed in the central and peripheral nervous system. The adenylyl cyclase (AC)/cyclic AMP (cAMP) is their main intracellular signal transduction pathway. Numerous data suggest that PACAP and VIP have considerable neuroprotective potential, indicating the possibility for their use as new therapeutic strategies in stroke treatment. The aim of this study was to evaluate the effect of oxygen-glucose deprivation (OGD) - an established in vitro model for ischemic cell stress - on PACAP and VIP-evoked receptor-mediated cAMP generation in glial and neuronal cells, and to determine whether PACAP and VIP have neuroprotective activity under these conditions. METHODS The formation of [(3)H]cAMP by PACAP, VIP and forskolin (a direct activator of AC) was measured in [(3)H]adenine prelabeled primary rat glial and neuronal cells under normoxia and OGD conditions. The effects of PACAP and VIP on cell viability were measured using the MTT conversion method, and were compared to tacrolimus (FK506), a well known neuroprotective agent. RESULTS The OGD model inhibited the PACAP and VIP-induced cAMP formation in rat astrocytes and neurons. Incubation of neuronal cells with PACAP prevented OGD-induced cell death, more efficiently than VIP and FK506. CONCLUSION The obtained results showed that hypoxia/ischemia may trigger down-regulation of the brain AC-coupled PACAP/VIP receptors, with a consequent decrease of PACAP- and/or VIP-ergic-dependent cAMP-driven signaling. Moreover, our findings indicate that PACAP and VIP can prevent the deleterious effect of OGD on rat neuronal cells.
Collapse
Affiliation(s)
- Marta Jóźwiak-Bębenista
- Department of Pharmacology and Toxicology, The Interfaculty Chair of Basic and Clinical Pharmacology, Medical University of Lodz, Łódź, Poland.
| | - Edward Kowalczyk
- Department of Pharmacology and Toxicology, The Interfaculty Chair of Basic and Clinical Pharmacology, Medical University of Lodz, Łódź, Poland
| | - Jerzy Z Nowak
- Department of Pharmacology and Toxicology, The Interfaculty Chair of Basic and Clinical Pharmacology, Medical University of Lodz, Łódź, Poland
| |
Collapse
|
32
|
Neuroprotection of a novel cyclopeptide C*HSDGIC* from the cyclization of PACAP (1-5) in cellular and rodent models of retinal ganglion cell apoptosis. PLoS One 2014; 9:e108090. [PMID: 25286089 PMCID: PMC4186886 DOI: 10.1371/journal.pone.0108090] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 08/11/2014] [Indexed: 11/22/2022] Open
Abstract
Purpose To investigate the protective effects of a novel cyclopeptide C*HSDGIC* (CHC) from the cyclization of Pituitary adenylate cyclase-activating polypeptide (PACAP) (1–5) in cellular and rodent models of retinal ganglion cell apoptosis. Methodology/Principal Findings Double-labeling immunohistochemistry was used to detect the expression of Thy-1 and PACAP receptor type 1 in a retinal ganglion cell line RGC-5. The apoptosis of RGC-5 cells was induced by 0.02 J/cm2 Ultraviolet B irradiation. MTT assay, flow cytometry, fluorescence microscopy were used to investigate the viability, the level of reactive oxygen species (ROS) and apoptosis of RGC-5 cells respectively. CHC attenuated apoptotic cell death induced by Ultraviolet B irradiation and inhibited the excessive generation of ROS. Moreover, CHC treatment resulted in decreased expression of Bax and concomitant increase of Bcl-2, as was revealed by western-blot analysis. The in vivo apoptosis of retinal ganglion cells was induced by injecting 50 mM N-methyl-D-aspartate (NMDA) (100 nmol in a 2 µL saline solution) intravitreally, and different dosages of CHC were administered. At day 7, rats in CHC+ NMDA-treated groups showed obvious aversion to light when compared to NMDA rats. Electroretinogram recordings revealed a marked decrease in the amplitudes of a-wave, b-wave, and photopic negative response due to NMDA damage. In retina receiving intravitreal NMDA and CHC co-treatment, these values were significantly increased. CHC treatment also resulted in less NMDA-induced cell loss and a decrease in the proportion of dUTP end-labeling-positive cells in ganglion cell line. Conclusions C*HSDGIC*, a novel cyclopeptide from PACAP (1–5) attenuates apoptosis in RGC-5 cells and inhibits NMDA-induced retinal neuronal death. The beneficial effects may occur via the mitochondria pathway. PACAP derivatives like CHC may serve as a promising candidate for neuroprotection in glaucoma.
Collapse
|
33
|
Pituitary adenylate cyclase-activating polypeptide protects against β-amyloid toxicity. Neurobiol Aging 2014; 35:2064-71. [PMID: 24726470 DOI: 10.1016/j.neurobiolaging.2014.03.022] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 02/28/2014] [Accepted: 03/15/2014] [Indexed: 11/21/2022]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) is a neurotrophin. However, its role in human Alzheimer's disease (AD) is largely unknown. We examined PACAP expression in postmortem human AD and triple transgenic mouse (3xTG, Psen1/APPSwe/TauP301L) brains. We established an in vitro model of primary neuronal cell culture to study the protective effects of PACAP against β-amyloid (Aβ) toxicity. We further studied the PACAP-Sirtuin 3 (Sirt3) pathway on mitochondrial function. PACAP expression was reduced in AD and 3xTG mouse brains. This reduction was inversely correlated with Aβ and tau protein levels. Treatment with PACAP effectively protected neurons against Aβ toxicity. PACAP stimulated mitochondrial Sirt3 production. Similar to PACAP, Sirt3 was reduced in AD and 3xTG brains. Knocking down Sirt3 compromised the neuroprotective effects of PACAP, and this was reversed by over-expressing Sirt3. PACAP is reduced in AD and may represent a novel therapeutic strategy.
Collapse
|
34
|
Protective effects of mangosteen extract on H2O2-induced cytotoxicity in SK-N-SH cells and scopolamine-induced memory impairment in mice. PLoS One 2013; 8:e85053. [PMID: 24386444 PMCID: PMC3874002 DOI: 10.1371/journal.pone.0085053] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 11/21/2013] [Indexed: 11/19/2022] Open
Abstract
Mangosteen extracts (ME) contain high levels of polyphenolic compounds and antioxidant activity. Protective effects of ME against β-amyloid peptide (Aβ), induced cytotoxicity have been reported. Here, we further studied the protective effects of ME against oxidative stress induced by hydrogen peroxide (H2O2) and polychlorinated biphenyls (PCBs), and demonstrated the protection against memory impairment in mice. The cytoprotective effects of ME were measured as cell viability and the reduction in ROS activity. In SK-N-SH cell cultures, 200 μg/ml ME could partially antagonize the effects of 150 or 300 µM H2O2 on cell viability, ROS level and caspase-3 activity. At 200, 400 or 800 µg/ml, ME reduced AChE activity of SK-N-SH cells to about 60% of the control. In vivo study, Morris water maze and passive avoidance tests were used to assess the memory of the animals. ME, especially at 100 mg/kg body weight, could improve the animal’s memory and also antagonize the effect of scopolamine on memory. The increase in ROS level and caspase-3 activity in the brain of scopolamine-treated mice were antagonized by the ME treatment. The study demonstrated cytoprotective effects of ME against H2O2 and PCB-52 toxicity and having AChE inhibitory effect in cell culture. ME treatment in mice could attenuate scopolamine-induced memory deficit and oxidative stress in brain.
Collapse
|
35
|
Marin R, Casañas V, Pérez JA, Fabelo N, Fernandez CE, Diaz M. Oestrogens as modulators of neuronal signalosomes and brain lipid homeostasis related to protection against neurodegeneration. J Neuroendocrinol 2013; 25:1104-15. [PMID: 23795744 DOI: 10.1111/jne.12068] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 05/22/2013] [Accepted: 06/18/2013] [Indexed: 12/19/2022]
Abstract
Oestrogens trigger several pathways at the plasma membrane that exert beneficial actions against neurodegenerative diseases, such as Alzheimer's disease and Parkinson's disease. Part of these actions takes place in lipid rafts, which are membrane domains with a singular protein and lipid composition. These microdomains also represent a preferential site for signalling protein complexes, or signalosomes. A plausible hypothesis is that the dynamic interaction of signalosomes with different extracellular ligands may be at the basis of neuronal maintenance against different neuropathologies. Oestrogen receptors are localised in neuronal lipid rafts, taking part of macromolecular complexes together with a voltage-dependent anion channel (VDAC), and other molecules. Oestradiol binding to its receptor at this level enhances neuroprotection against amyloid-β degeneration through the activation of different signal transduction pathways, including VDAC gating modulation. Moreover, part of the stability and functionality of signalling platforms lays on the distribution of lipid hallmarks in these microstructures, which modulate membrane physicochemical properties, thus favouring molecular interactions. Interestingly, recent findings indicate a potential role of oestrogens in the preservation of neuronal membrane physiology related to lipid homeostasis. Thus, oestrogens and docosahexaenoic acid may act synergistically to stabilise brain lipid structure by regulating neuronal lipid biosynthetic pathways, suggesting that part of the neuroprotective effects elicited by oestrogens occur through mechanisms aimed at preserving lipid homeostasis. Overall, oestrogen mechanisms of neuroprotection may occur not only by its interaction with neuronal protein targets through nongenomic and genomic mechanisms, but also through its participation in membrane architecture stabilisation via 'lipostatic' mechanisms.
Collapse
Affiliation(s)
- R Marin
- Department of Physiology, Laboratory of Cellular Neurobiology, University of La Laguna, La Laguna, Tenerife, Spain
| | | | | | | | | | | |
Collapse
|
36
|
Wang ZJ, Han WN, Yang GZ, Yuan L, Liu XJ, Li QS, Qi JS. The neuroprotection of Rattin against amyloid β peptide in spatial memory and synaptic plasticity of rats. Hippocampus 2013; 24:44-53. [PMID: 23996574 DOI: 10.1002/hipo.22202] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 08/21/2013] [Accepted: 08/22/2013] [Indexed: 12/22/2022]
Abstract
Rattin, a specific derivative of humanin in rats, shares the ability with HN to protect neurons against amyloid β (Aβ) peptide-induced cellular toxicity. However, it is still unclear whether Rattin can protect against Aβ-induced deficits in cognition and synaptic plasticity in rats. In the present study, we observed the effects of Rattin and Aβ31-35 on the spatial reference memory and in vivo hippocampal Long-term potentiation of rats by using Morris water maze test and hippocampal field potential recording. Furthermore, the probable molecular mechanism underlying the neuroprotective roles of Rattin was investigated. We showed that intra-hippocampal injection of Rattin effectively prevented the Aβ31-35-induced spatial memory deficits and hippocampal LTP suppression in rats; the Aβ31-35-induced activation of Caspase-3 and inhibition of STAT3 in the hippocampus were also prevented by Rattin treatment. These findings indicate that Rattin treatment can protect spatial memory and synaptic plasticity of rats against Aβ31-35-induced impairments, and the underlying protective mechanism of Rattin may be involved in STAT3 and Caspases-3 pathways. Therefore, application of Rattin or activation of its signaling pathways in the brain might be beneficial to the prevention of Aβ-related cognitive deficits.
Collapse
Affiliation(s)
- Zhao-Jun Wang
- Department of Neurobiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
37
|
|
38
|
STC1 induction by PACAP is mediated through cAMP and ERK1/2 but not PKA in cultured cortical neurons. J Mol Neurosci 2013; 46:75-87. [PMID: 21975601 DOI: 10.1007/s12031-011-9653-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Accepted: 09/15/2011] [Indexed: 10/17/2022]
Abstract
The neuroprotective actions of PACAP (pituitary adenylate cyclase-activating polypeptide) in vitro and in vivo suggest that activation of its cognate G protein coupled receptor PAC1 or downstream signaling molecules,and thus activation of PACAP target genes, could be of therapeutic benefit. Here, we show that cultured rat cortical neurons predominantly expressed the PAC1hop and null variants. PACAP receptor activation resulted in the elevation of the two second messengers cAMP and Ca(2+) and expression of the putative neuroprotectant stanniocalcin 1(STC1). PACAP signaling to the STC1 gene proceeded through the extracellular signal-regulated kinases 1 and 2(ERK1/2), but not through the cAMP-dependent protein kinase (PKA), and was mimicked by the adenylate cyclase activator forskolin. PACAP- and forskolin-mediated activation of ERK1/2 occurred through cAMP, but not PKA.These results suggest that STC1 gene induction proceeds through cAMP and ERK1/2, independently of PKA, the canonical cAMP effector. In contrast, PACAP signaling to the BDNF gene proceeded through PKA, suggesting that two different neuroprotective cAMP pathways co-exist in differentiated cortical neurons. The selective activation of a potentially neuroprotective cAMP-dependent pathway different from the canonical cAMP pathway used in many physiological processes, such as memory storage, has implications for pharmacological activation of neuroprotection in vivo.
Collapse
|
39
|
Tripathy D, Sanchez A, Yin X, Martinez J, Grammas P. Age-related decrease in cerebrovascular-derived neuroprotective proteins: effect of acetaminophen. Microvasc Res 2012; 84:278-85. [PMID: 22944728 PMCID: PMC3483357 DOI: 10.1016/j.mvr.2012.08.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 07/17/2012] [Accepted: 08/17/2012] [Indexed: 12/19/2022]
Abstract
As the population ages, the need for effective methods to maintain brain function in older adults is increasingly pressing. Vascular disease and neurodegenerative disorders commonly co-occur in older persons. Cerebrovascular products contribute to the neuronal milieu and have important consequences for neuronal viability. In this regard vascular derived neuroprotective proteins, Such as vascular endothelial growth factor (VEGF), pigment epithelium-derived factor (PEDF), and pituitary adenylate cyclase activating peptide (PACAP) are important for maintaining neuronal viability, especially in the face of injury and disease. The objective of this study is to measure and compare levels of VEGF, PEDF and PACAP released from isolated brain microvessels of Fischer 344 rats at 6, 12, 18, and 24 months of age. Addition of acetaminophen to isolated brain microvessels is employed to determine whether this drug affects vascular expression of these neuroprotective proteins. Experiments on cultured brain endothelial cells are performed to explore the mechanisms/mediators that regulate the effect of acetaminophen on endothelial cells. The data indicate cerebrovascular expression of VEGF, PEDF and PACAP significantly decreases with age. The age-associated decrease in VEGF and PEDF is ameliorated by addition of acetaminophen to isolated brain microvessels. Also, release of VEGF, PEDF, and PACAP from cultured brain endothelial cells decreases with exposure to the oxidant stressor menadione. Acetaminophen treatment upregulates VEGF, PEDF and PACAP in brain endothelial cells exposed to oxidative stress. The effect of acetaminophen on cultured endothelial cells is in part inhibited by the selective thrombin inhibitor hirudin. The results of this study suggest that acetaminophen may be a useful agent for preserving cerebrovascular function. If a low dose of acetaminophen can counteract the decrease in vascular-derived neurotrophic factors evoked by age and oxidative stress, this drug might be useful for improving brain function in the elderly.
Collapse
Affiliation(s)
- Debjani Tripathy
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Alma Sanchez
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Xiangling Yin
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Joseph Martinez
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Paula Grammas
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, Texas
| |
Collapse
|
40
|
Targeting VIP and PACAP receptor signalling: new therapeutic strategies in multiple sclerosis. ASN Neuro 2011; 3:AN20110024. [PMID: 21895607 PMCID: PMC3189630 DOI: 10.1042/an20110024] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
MS (multiple sclerosis) is a chronic autoimmune and neurodegenerative pathology of the CNS (central nervous system) affecting approx. 2.5 million people worldwide. Current and emerging DMDs (disease-modifying drugs) predominantly target the immune system. These therapeutic agents slow progression and reduce severity at early stages of MS, but show little activity on the neurodegenerative component of the disease. As the latter determines permanent disability, there is a critical need to pursue alternative modalities. VIP (vasoactive intestinal peptide) and PACAP (pituitary adenylate cyclase-activating peptide) have potent anti-inflammatory and neuroprotective actions, and have shown significant activity in animal inflammatory disease models including the EAE (experimental autoimmune encephalomyelitis) MS model. Thus, their receptors have become candidate targets for inflammatory diseases. Here, we will discuss the immunomodulatory and neuroprotective actions of VIP and PACAP and their signalling pathways, and then extensively review the structure–activity relationship data and biophysical interaction studies of these peptides with their cognate receptors.
Collapse
|
41
|
Di Michele M, Peeters K, Loyen S, Thys C, Waelkens E, Overbergh L, Hoylaerts M, Van Geet C, Freson K. Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) impairs the regulation of apoptosis in megakaryocytes by activating NF-κB: a proteomic study. Mol Cell Proteomics 2011; 11:M111.007625. [PMID: 21972247 DOI: 10.1074/mcp.m111.007625] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
We previously showed that the Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) and its receptor VPAC1 are negative regulators of megakaryopoiesis and platelet function, but their downstream signaling pathway that inhibits this process still remained unknown. A combined proteomic, transcriptomic, and bioinformatic approach was here used to elucidate the molecular mechanisms underlying PACAP signaling via VPAC1 in megakaryocytes. Two-dimensional difference gel electrophoresis and tandem MS were applied to detect differentially expressed proteins in megakaryocytic CHRF cells stimulated with PACAP. The majority of the 120 proteins modulated by PACAP belong to the class of "cell cycle and apoptosis" proteins. The up- or down-regulated expression of some proteins was confirmed by immunoblot and immunohistochemical analysis. A meta-analysis of our data and 12 other published studies was performed to evaluate signaling pathways involved in different cellular models of PACAP response. From 2384 differentially expressed genes/proteins, 83 were modulated by PACAP in at least three independent studies and Ingenuity Pathway Analysis further identified apoptosis as the highest scored network with NF-κB as a key-player. PACAP inhibited serum depletion-induced apoptosis of CHRF cells via VPAC1 stimulation. In addition, PACAP switched on NF-κB dependent gene expression since higher nuclear levels of the active NF-κB p50/p65 heterodimer were found in CHRF cells treated with PACAP. Finally, a quantitative real time PCR apoptosis array was used to study RNA from in vitro differentiated megakaryocytes from a PACAP overexpressing patient, leading to the identification of 15 apoptotic genes with a 4-fold change in expression and Ingenuity Pathway Analysis again revealed NF-κB as the central player. In conclusion, our findings suggest that PACAP interferes with the regulation of apoptosis in megakaryocytes, probably via stimulation of the NF-κB pathway.
Collapse
Affiliation(s)
| | - Karen Peeters
- Center for Molecular and Vascular Biology, Leuven, Belgium
| | - Serena Loyen
- Center for Molecular and Vascular Biology, Leuven, Belgium
| | - Chantel Thys
- Center for Molecular and Vascular Biology, Leuven, Belgium
| | | | - Lutgart Overbergh
- Laboratory for Experimental Medicine and Endocrinology, Leuven, Belgium
| | - Marc Hoylaerts
- Center for Molecular and Vascular Biology, Leuven, Belgium
| | - Christel Van Geet
- Center for Molecular and Vascular Biology, Leuven, Belgium; Department of Pediatrics, University Hospital Leuven, K.U. Leuven, Leuven, Belgium
| | | |
Collapse
|
42
|
Acquaah-Mensah GK, Taylor RC, Bhave SV. PACAP interactions in the mouse brain: implications for behavioral and other disorders. Gene 2011; 491:224-31. [PMID: 22001548 DOI: 10.1016/j.gene.2011.09.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Revised: 09/02/2011] [Accepted: 09/09/2011] [Indexed: 12/24/2022]
Abstract
As an activator of adenylate cyclase, the neuropeptide Pituitary Adenylate Cyclase Activating Peptide (PACAP) impacts levels of cyclic AMP, a key second messenger available in brain cells. PACAP is involved in certain adult behaviors. To elucidate PACAP interactions, a compendium of microarrays representing mRNA expression in the adult mouse whole brain was pooled from the Phenogen database for analysis. A regulatory network was computed based on mutual information between gene pairs using gene expression data across the compendium. Clusters among genes directly linked to PACAP, and probable interactions between corresponding proteins were computed. Database "experts" affirmed some of the inferred relationships. The findings suggest ADCY7 is probably the adenylate cyclase isoform most relevant to PACAP's action. They also support intervening roles for kinases including GSK3B, PI 3-kinase, SGK3 and AMPK. Other high-confidence interactions are hypothesized for future testing. This new information has implications for certain behavioral and other disorders.
Collapse
Affiliation(s)
- George K Acquaah-Mensah
- Department of Pharmaceutical Sciences, Massachusetts College of Pharmacy and Health Sciences, Worcester, MA 01608, USA.
| | | | | |
Collapse
|
43
|
Misaka S, Sato H, Aoki Y, Mizumoto T, Onoue S, Yamada S. Novel vasoactive intestinal peptide derivatives with improved stability protect rat alveolar L2 cells from cigarette smoke-induced cytotoxicity and apoptosis. Peptides 2011; 32:401-7. [PMID: 20977915 DOI: 10.1016/j.peptides.2010.10.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Revised: 10/17/2010] [Accepted: 10/18/2010] [Indexed: 11/24/2022]
Abstract
Vasoactive intestinal peptide (VIP) has been thought to be a promising candidate for asthma/chronic obstructive pulmonary disease (COPD), and our group previously developed several long-lasting VIP derivatives. The objective of the present study was to clarify the therapeutic potential of new VIP derivatives with improved chemical and metabolic stability. Exposure of rat alveolar L2 cells to cigarette smoke extract (CSE) for 1h led to release of lactate dehydrogenase (LDH) and decreased viability in a CSE concentration-dependent manner. There appeared to be marked induction of apoptosis after CSE exposure, as demonstrated by 59% elevation of caspase-3 activity and TUNEL staining. In contrast, a stabilized VIP derivative, [R(15,20,21), L(17)]-VIP-GRR (IK312532), at a concentration of 10(-7)M, exhibited 71% attenuation of LDH release and 85% decrease of the number of apoptotic cells. In addition to IK312532, new VIP derivatives also showed anti-apoptotic effects against CSE toxicity and marked reduction of nitric oxide production. In terms of cytoprotective effects, [R(15,20,21), L(17), A(24,25), des-N(28)]-VIP-GRR was more effective than VIP and IK312532, possibly due to the improved stability. Thus, the present study is the first to demonstrate that novel stabilized VIP derivatives exert anti-apoptotic and cytoprotective effects on CSE-induced cytotoxicity.
Collapse
Affiliation(s)
- Shingen Misaka
- Department of Pharmacokinetics, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan
| | | | | | | | | | | |
Collapse
|
44
|
Moody TW, Ito T, Osefo N, Jensen RT. VIP and PACAP: recent insights into their functions/roles in physiology and disease from molecular and genetic studies. Curr Opin Endocrinol Diabetes Obes 2011; 18:61-7. [PMID: 21157320 PMCID: PMC3075877 DOI: 10.1097/med.0b013e328342568a] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating polypeptide (PACAP) as well as the three classes of G-protein-coupled receptors mediating their effects, are widely distributed in the central nervous system (CNS) and peripheral tissues. These peptides are reported to have many effects in different tissues, which are physiological or pharmacological, and which receptor mediates which effect, has been difficult to determine, primarily due to lack of potent, stable, selective agonists/antagonists. Recently the use of animals with targeted knockout of the peptide or a specific receptor has provided important insights into their role in normal physiology and disease states. RECENT FINDINGS During the review period, considerable progress and insights has occurred in the understanding of the role of VIP/PACAP as well as their receptors in a number of different disorders/areas. Particularly, insights into their roles in energy metabolism, glucose regulation, various gastrointestinal processes including gastrointestinal inflammatory conditions and motility and their role in the CNS as well as CNS diseases has greatly expanded. SUMMARY PACAP/VIP as well as their three classes of receptors are important in many physiological/pathophysiological processes, some of which are identified in these studies using knockout animals. These studies may lead to new novel treatment approaches. Particularly important are their roles in glucose metabolism and on islets leading to possible novel approaches in diabetes; their novel anti-inflammatory, cytoprotective effects, their CNS neuroprotective effects, and their possible roles in diseases such as schizophrenia and chronic depression.
Collapse
Affiliation(s)
- Terry W. Moody
- Department of Health and Human Services, National Cancer Institute Office of the Director, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Tetsuhide Ito
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Science, Kyushu University, Fukuoka, Japan
| | - Nuramy Osefo
- Department of Health and Human Services, National Cancer Institute Office of the Director, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Robert T. Jensen
- Digestive Diseases Branch, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
45
|
Kanekar S, Gandham M, Lucero MT. PACAP protects against TNFα-induced cell death in olfactory epithelium and olfactory placodal cell lines. Mol Cell Neurosci 2010; 45:345-54. [PMID: 20654718 PMCID: PMC2962708 DOI: 10.1016/j.mcn.2010.07.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [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
Affiliation(s)
- Shami Kanekar
- Department of Physiology, University of Utah, Salt Lake City, UT 84108
| | - Mahendra Gandham
- Department of Physiology, University of Utah, Salt Lake City, UT 84108
| | - Mary T Lucero
- Department of Physiology, University of Utah, Salt Lake City, UT 84108
- Utah Brain Institute, Salt Lake City, UT 84108
- Utah Center on Aging, University of Utah, Salt Lake City, UT 84112
| |
Collapse
|
46
|
Shneider Y, Shtrauss Y, Yadid G, Pinhasov A. Differential expression of PACAP receptors in postnatal rat brain. Neuropeptides 2010; 44:509-14. [PMID: 20971507 DOI: 10.1016/j.npep.2010.09.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2010] [Revised: 09/13/2010] [Accepted: 09/14/2010] [Indexed: 12/22/2022]
Abstract
Pituitary Adenylate Cyclase Activating Polypeptide (PACAP) is a multi-functional neuropeptide that acts through activation of three common G-protein coupled receptors (VPAC1, VPAC2 and PAC1). In this study, we have investigated the gene expression profile of PAC1 isoforms (Hop1, Hip, Hip-Hop) and VPAC1, VPAC2 receptors in distinct brain regions during different stages of rat postnatal development. Using quantitative real time PCR approach we found that PAC1 isoforms were highly expressed in the cortex of newborns with marked decrease in expression during later stages of development. In contrast, mRNA levels of VPAC1, VPAC2 receptors were markedly lower in newborns in comparison to later developmental stages. Expression of PAC1 isoforms predominated in the hippocampus, while expression of VPAC1 was more prominent in the cortex and VPAC2 in the striatum and hippocampus. In addition we found that during early stages of postnatal development the expression of PAC1 receptor in the hippocampus was significantly higher in females than in males. No sex dependent differences in expression were observed for the VPAC1 and VPAC2 receptors. In summary, differential expression of PAC1, VPAC1 and VPAC2 receptors during postnatal development as well as gender dependent differences of PAC1 receptor expression in the hippocampus, will contribute to our understanding of the role of PACAP/VIP signaling system in normal brain development and function.
Collapse
Affiliation(s)
- Yevgenia Shneider
- Department of Molecular Biology, Ariel University Center of Samaria, Ariel, Israel
| | | | | | | |
Collapse
|
47
|
Bossers K, Wirz KTS, Meerhoff GF, Essing AHW, van Dongen JW, Houba P, Kruse CG, Verhaagen J, Swaab DF. Concerted changes in transcripts in the prefrontal cortex precede neuropathology in Alzheimer's disease. ACTA ACUST UNITED AC 2010; 133:3699-723. [PMID: 20889584 DOI: 10.1093/brain/awq258] [Citation(s) in RCA: 182] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Using the Braak staging for neurofibrillary changes as an objective indicator of the progression of Alzheimer's disease, we have performed a systematic search for global gene expression changes in the prefrontal cortex during the course of Alzheimer's disease. In the prefrontal cortex, senile plaques and neurofibrillary changes start to appear around Braak stage III, allowing for the detection of changes in gene expression before, during and after the onset of Alzheimer's disease neuropathology. Two distinct patterns of tightly co-regulated groups of genes were observed: (i) an increase in expression in early Braak stages, followed by a decline in expression in later stages (the UPDOWN clusters; containing 865 genes) and (ii) a decrease in expression in early Braak stages, followed by an increase in expression in later stages (the DOWNUP clusters; containing 983 genes). The most profound changes in gene expression were detected between Braak stages II and III, just before or at the onset of plaque pathology and neurofibrillary changes in the prefrontal cortex. We also observed an increase in intracellular beta amyloid staining from Braak stages I to III and a clear decrease in Braak stages IV to VI. These data suggest a link between specific gene expression clusters and Alzheimer's disease-associated neuropathology in the prefrontal cortex. Gene ontology over-representation and functional gene network analyses indicate an increase in synaptic activity and changes in plasticity during the very early pre-symptomatic stage of the disease. In later Braak stages, the decreased expression of these genes suggests a reduction in synaptic activity that coincides with the appearance of plaque pathology and neurofibrillary changes and the clinical diagnosis of mild cognitive impairment. The interaction of the ApoE genotype with the expression levels of the genes in the UPDOWN and DOWNUP clusters demonstrates that the accelerating role of ApoE-ε4 in the progression of Alzheimer's disease is reflected in the temporal changes in gene expression presented here. Since the UPDOWN cluster contains several genes involved in amyloid precursor protein processing and beta amyloid clearance that increase in expression in parallel with increased intracellular beta amyloid load, just before the onset of plaque pathology in the prefrontal cortex, we hypothesize that the temporally orchestrated increase in genes involved in synaptic activity represents a coping mechanism against increased soluble beta amyloid levels. As these gene expression changes occur before the appearance of Alzheimer's disease-associated neuropathology, they provide an excellent starting point for the identification of new targets for the development of therapeutic strategies aimed at the prevention of Alzheimer's disease.
Collapse
Affiliation(s)
- Koen Bossers
- Neuroregeneration Group, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Seki T, Itoh H, Nakamachi T, Endo K, Wada Y, Nakamura K, Shioda S. Suppression of rat retinal ganglion cell death by PACAP following transient ischemia induced by high intraocular pressure. J Mol Neurosci 2010; 43:30-4. [PMID: 20585899 DOI: 10.1007/s12031-010-9410-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Accepted: 06/11/2010] [Indexed: 01/26/2023]
Abstract
Glaucoma is a neurodegenerative disease in which increasing intraocular pressure leads to the progressive loss of retinal ganglion cells (RGCs) and blindness. Here, we report a neuroprotective effect of pituitary adenylate cyclase-activating polypeptide (PACAP) against RGC loss induced by high intraocular pressure in the rat. Vehicle or PACAP (1 fM to 1,000 pM) solution was injected into the vitreous body once after induction of a high intraocular pressure (110 mmHg). Seven days later, the number of viable RGCs was reduced to 45% of that in the intact control. However, PACAP treatment significantly reduced this RGC death in a bimodal manner, with peaks at 10 fM and 10-100 pM. The cAMP antagonist Rp-cAMP significantly blocked the neuroprotective effect of PACAP at both high and low doses, whereas the MAP kinase inhibitor PD-98059 only prevented the effect of the low dose of PACAP. These findings suggest that PACAP has bimodal effects in the neuroprotection of RGCs against ischemia and that these effects are mediated via different signaling pathways.
Collapse
Affiliation(s)
- Tamotsu Seki
- Department of Anatomy, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan
| | | | | | | | | | | | | |
Collapse
|
49
|
Dejda A, Chan P, Seaborn T, Coquet L, Jouenne T, Fournier A, Vaudry H, Vaudry D. Involvement of stathmin 1 in the neurotrophic effects of PACAP in PC12 cells. J Neurochem 2010; 114:1498-510. [PMID: 20569302 DOI: 10.1111/j.1471-4159.2010.06873.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Rat pheochromocytoma PC12 cells have been widely used to investigate the neurotrophic activities of pituitary adenylate cyclase-activating polypeptide (PACAP). In particular, PACAP has been shown to promote differentiation and to inhibit apoptosis of PC12 cells. In order to identify the mechanisms mediating these effects, we sought for proteins that are phosphorylated upon PACAP treatment. High-performance liquid chromatography and 2D gel electrophoresis analysis, coupled with mass spectrometry, revealed that stathmin 1 is strongly phosphorylated within only 5 min of exposure to PACAP. Western blot experiments confirmed that PACAP induced a robust phosphorylation of stathmin 1 in a time-dependent manner. On the other hand, PACAP decreased stathmin 1 gene expression. Investigations of the signaling mechanisms known to be activated by PACAP revealed that phosphorylation of stathmin 1 was mainly mediated through the protein kinase A and mitogen-activated protein kinase pathways. Blockage of stathmin 1 expression with small interfering RNA did not affect PC12 cell differentiation induced by PACAP but reduced the ability of the peptide to inhibit caspase 3 activity and significantly decreased its neuroprotective action. Taken together, these data demonstrate that stathmin 1 is involved in the neurotrophic effect of PACAP in PC12 cells.
Collapse
Affiliation(s)
- Agnieszka Dejda
- INSERM U982, Institut Fédératif de Recherches Multidisciplinaires sur les Peptides (IFRMP 23), Université de Rouen, Mont-Saint-Aignan, France
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Vaudry D, Falluel-Morel A, Bourgault S, Basille M, Burel D, Wurtz O, Fournier A, Chow BKC, Hashimoto H, Galas L, Vaudry H. Pituitary adenylate cyclase-activating polypeptide and its receptors: 20 years after the discovery. Pharmacol Rev 2009; 61:283-357. [PMID: 19805477 DOI: 10.1124/pr.109.001370] [Citation(s) in RCA: 862] [Impact Index Per Article: 53.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a 38-amino acid C-terminally alpha-amidated peptide that was first isolated 20 years ago from an ovine hypothalamic extract on the basis of its ability to stimulate cAMP formation in anterior pituitary cells (Miyata et al., 1989. PACAP belongs to the vasoactive intestinal polypeptide (VIP)-secretin-growth hormone-releasing hormone-glucagon superfamily. The sequence of PACAP has been remarkably well conserved during evolution from protochordates to mammals, suggesting that PACAP is involved in the regulation of important biological functions. PACAP is widely distributed in the brain and peripheral organs, notably in the endocrine pancreas, gonads, respiratory and urogenital tracts. Characterization of the PACAP precursor has revealed the existence of a PACAP-related peptide, the activity of which remains unknown. Two types of PACAP binding sites have been characterized: type I binding sites exhibit a high affinity for PACAP and a much lower affinity for VIP, whereas type II binding sites have similar affinity for PACAP and VIP. Molecular cloning of PACAP receptors has shown the existence of three distinct receptor subtypes: the PACAP-specific PAC1-R, which is coupled to several transduction systems, and the PACAP/VIP-indifferent VPAC1-R and VPAC2-R, which are primarily coupled to adenylyl cyclase. PAC1-Rs are particularly abundant in the brain, the pituitary and the adrenal gland, whereas VPAC receptors are expressed mainly in lung, liver, and testis. The development of transgenic animal models and specific PACAP receptor ligands has strongly contributed to deciphering the various actions of PACAP. Consistent with the wide distribution of PACAP and its receptors, the peptide has now been shown to exert a large array of pharmacological effects and biological functions. The present report reviews the current knowledge concerning the pleiotropic actions of PACAP and discusses its possible use for future therapeutic applications.
Collapse
Affiliation(s)
- David Vaudry
- Institut National de la Santé et de la Recherche Médicale U413, European Institute for Peptide Research (Institut Fédératif de Recherches Multidisciplinaires sur les Peptides 23), Mont-Saint-Aignan, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|