1
|
Ali NH, Al-Kuraishy HM, Al-Gareeb AI, Hadi NR, Assiri AA, Alrouji M, Welson NN, Alexiou A, Papadakis M, Batiha GES. Hypoglycemia and Alzheimer Disease Risk: The Possible Role of Dasiglucagon. Cell Mol Neurobiol 2024; 44:55. [PMID: 38977507 PMCID: PMC11230952 DOI: 10.1007/s10571-024-01489-y] [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: 03/05/2024] [Accepted: 06/21/2024] [Indexed: 07/10/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by memory impairment and cognitive dysfunctions. It has been shown that hypoglycemia can adversely affect AD neuropathology. It is well-known that chronic hyperglycemia in type 2 diabetes (T2D) is regarded as a potential risk factor for the development and progression of AD. However, the effect of recurrent hypoglycemia on the pathogenesis of AD was not deeply discussed, and how recurrent hypoglycemia affects AD at cellular and molecular levels was not intensely interpreted by the previous studies. The underlying mechanisms for hypoglycaemia-induced AD are diverse such as endothelial dysfunction, thrombosis, and neuronal injury that causing tau protein hyperphosphorylation and the accumulation of amyloid beta (Aβ) in the brain neurons. Of note, the glucagon hormone, which controls blood glucose, can also regulate the cognitive functions. Glucagon increases blood glucose by antagonizing the metabolic effect of insulin. Therefore, glucagon, through attenuation of hypoglycemia, may prevent AD neuropathology. Glucagon/GLP-1 has been shown to promote synaptogenesis, hippocampal synaptic plasticity, and learning and memory, while attenuating amyloid and tau pathologies. Therefore, activation of glucagon receptors in the brain may reduce AD neuropathology. A recent glucagon receptor agonist dasiglucagon which used in the management of hypoglycemia may be effective in preventing hypoglycemia and AD neuropathology. This review aims to discuss the potential role of dasiglucagon in treating hypoglycemia in AD, and how this drug reduce AD neuropathology.
Collapse
Affiliation(s)
- Naif H Ali
- Assistant Professor of Neurology, Department of Internal Medicine, Medical College, Najran University, Najran, Kingdom of Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Head of Jabir Ibn, Hayyan Medical University, Al-Ameer Qu./Najaf-Iraq, PO.Box13, Kufa, Iraq
| | - Najah R Hadi
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Kufa, Iraq
| | - Abdullah A Assiri
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University, Abha, Kingdom of Saudi Arabia
| | - Mohammed Alrouji
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Shaqra, 11961, Kingdom of Saudi Arabia
| | - Nermeen N Welson
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Beni-Suef University, Beni Suef, 62511, Egypt
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, 2770, Australia
- AFNP Med, 1030, Vienna, Austria
- University Centre for Research & Development, Chandigarh University, Punjab, India
- Department of Research & Development, Funogen, Athens, Greece
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, Heusnerstrasse 40, University of Witten-Herdecke, 42283, Wuppertal, Germany
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, AlBeheira, 22511, Egypt
| |
Collapse
|
2
|
Gray M, Nash KR, Yao Y. Adenylyl cyclase 2 expression and function in neurological diseases. CNS Neurosci Ther 2024; 30:e14880. [PMID: 39073001 PMCID: PMC11284242 DOI: 10.1111/cns.14880] [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: 05/09/2024] [Revised: 06/25/2024] [Accepted: 07/15/2024] [Indexed: 07/30/2024] Open
Abstract
Adenylyl cyclases (Adcys) catalyze the formation of cAMP, a secondary messenger essential for cell survival and neurotransmission pathways in the CNS. Adcy2, one of ten Adcy isoforms, is highly expressed in the CNS. Abnormal Adcy2 expression and mutations have been reported in various neurological disorders in both rodents and humans. However, due to the lack of genetic tools, loss-of-function studies of Adcy2 are scarce. In this review, we summarize recent findings on Adcy2 expression and function in neurological diseases. Specifically, we first introduce the biochemistry, structure, and function of Adcy2 briefly. Next, the expression and association of Adcy2 in human patients and rodent models of neurodegenerative diseases (Alzheimer's disease and Parkinson's disease), psychiatric disorders (Tourette syndrome, schizophrenia, and bipolar disorder), and other neurological conditions (stress-associated disorders, stroke, epilepsy, and Lesch-Nyhan Syndrome) are elaborated. Furthermore, we discuss the pros and cons of current studies as well as key questions that need to be answered in the future. We hope to provide a focused review on Adcy2 that promotes future research in the field.
Collapse
Affiliation(s)
- Marsilla Gray
- Department of Molecular Pharmacology and Physiology, Morsani College of MedicineUniversity of South FloridaTampaFloridaUSA
| | - Kevin R. Nash
- Department of Molecular Pharmacology and Physiology, Morsani College of MedicineUniversity of South FloridaTampaFloridaUSA
| | - Yao Yao
- Department of Molecular Pharmacology and Physiology, Morsani College of MedicineUniversity of South FloridaTampaFloridaUSA
| |
Collapse
|
3
|
Fedele E. Anti-Amyloid Therapies for Alzheimer's Disease and the Amyloid Cascade Hypothesis. Int J Mol Sci 2023; 24:14499. [PMID: 37833948 PMCID: PMC10578107 DOI: 10.3390/ijms241914499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 09/22/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Over the past 30 years, the majority of (pre)clinical efforts to find an effective therapy for Alzheimer's disease (AD) focused on clearing the β-amyloid peptide (Aβ) from the brain since, according to the amyloid cascade hypothesis, the peptide was (and it is still considered by many) the pathogenic determinant of this neurodegenerative disorder. However, as reviewed in this article, results from the numerous clinical trials that have tested anti-Aβ therapies to date indicate that this peptide plays a minor role in the pathogenesis of AD. Indeed, even Aducanumab and Lecanemab, the two antibodies recently approved by the FDA for AD therapy, as well as Donanemab showed limited efficacy on cognitive parameters in phase III clinical trials, despite their capability of markedly lowering Aβ brain load. Furthermore, preclinical evidence demonstrates that Aβ possesses several physiological functions, including memory formation, suggesting that AD may in part be due to a loss of function of this peptide. Finally, it is generally accepted that AD could be the result of many molecular dysfunctions, and therefore, if we keep chasing only Aβ, it means that we cannot see the forest for the trees.
Collapse
Affiliation(s)
- Ernesto Fedele
- Pharmacology and Toxicology Unit, Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy;
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| |
Collapse
|
4
|
Tropea MR, Gulisano W, Vacanti V, Arancio O, Puzzo D, Palmeri A. Nitric oxide/cGMP/CREB pathway and amyloid-beta crosstalk: From physiology to Alzheimer's disease. Free Radic Biol Med 2022; 193:657-668. [PMID: 36400326 DOI: 10.1016/j.freeradbiomed.2022.11.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/30/2022] [Accepted: 11/12/2022] [Indexed: 11/17/2022]
Abstract
The nitric oxide (NO)/cGMP pathway has been extensively studied for its pivotal role in synaptic plasticity and memory processes, resulting in an increase of cAMP response element-binding (CREB) phosphorylation, and consequent synthesis of plasticity-related proteins. The NO/cGMP/CREB signaling is downregulated during aging and neurodegenerative disorders and is affected by Amyloid-β peptide (Aβ) and tau protein, whose increase and deposition is considered the key pathogenic event of Alzheimer's disease (AD). On the other hand, in physiological conditions, the crosstalk between the NO/cGMP/PKG/CREB pathway and Aβ ensures long-term potentiation and memory formation. This review summarizes the current knowledge on the interaction between the NO/cGMP/PKG/CREB pathway and Aβ in the healthy and diseased brain, offering a new perspective to shed light on AD pathophysiology. We will focus on the synaptic mechanisms underlying Aβ physiological interplay with cGMP pathway and how this balance is corrupted in AD, as high levels of Aβ interfere with NO production and cGMP molecular signaling leading to cognitive impairment. Finally, we will discuss results from preclinical and clinical studies proposing the increase of cGMP signaling as a therapeutic strategy in the treatment of AD.
Collapse
Affiliation(s)
- Maria Rosaria Tropea
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, 95123, Italy
| | - Walter Gulisano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, 95123, Italy
| | - Valeria Vacanti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, 95123, Italy
| | - Ottavio Arancio
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, USA; Department of Pathology & Cell Biology and Department of Medicine, Columbia University, New York, NY, 10032, USA
| | - Daniela Puzzo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, 95123, Italy; Oasi Research Institute-IRCCS, Troina (EN), 94018, Italy.
| | - Agostino Palmeri
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, 95123, Italy
| |
Collapse
|
5
|
Villa V, Montalto G, Caudano F, Fedele E, Ricciarelli R. Selective inhibition of phosphodiesterase 4D increases tau phosphorylation at Ser214 residue. Biofactors 2022; 48:1111-1117. [PMID: 35561079 PMCID: PMC9790528 DOI: 10.1002/biof.1847] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 04/29/2022] [Indexed: 12/30/2022]
Abstract
Tau is a protein that normally participates in the assembly and stability of microtubules. However, it can form intraneuronal hyperphosphorylated aggregates that are hallmarks of Alzheimer's disease and other neurodegenerative disorders known as tauopathies. Tau can be phosphorylated by multiple kinases at several sites. Among such kinases, the cAMP-dependent protein kinase A (PKA) phosphorylates tau at Ser214 (pTAU-S214), an event that was shown to reduce the pathological assembly of the protein. Given that the neuronal cAMP/PKA-activated cascade is involved in synaptic plasticity and memory, and that cAMP-enhancing strategies demonstrated promising therapeutic potential for the treatment of cognitive deficits, we investigated the impact of cAMP on pTAU-S214 in N2a cells and rat hippocampal slices. Our results confirm that the activation of adenylyl cyclase increases pTAU-S214 in both model systems and, more interestingly, this effect is mimicked by GEBR-7b, a phosphodiesterase 4D inhibitor with proven pro-cognitive efficacy in rodents.
Collapse
Affiliation(s)
- Viviana Villa
- Department of Experimental Medicine, Section of General Pathology, School of Medical and Pharmaceutical SciencesUniversity of GenoaGenoaItaly
| | - Giulia Montalto
- Department of Experimental Medicine, Section of General Pathology, School of Medical and Pharmaceutical SciencesUniversity of GenoaGenoaItaly
| | - Francesca Caudano
- Department of Experimental Medicine, Section of General Pathology, School of Medical and Pharmaceutical SciencesUniversity of GenoaGenoaItaly
| | - Ernesto Fedele
- Department of Pharmacy, Section of Pharmacology and Toxicology, School of Medical and Pharmaceutical SciencesUniversity of GenoaGenoaItaly
- IRCCS Ospedale Policlinico San MartinoGenoaItaly
| | - Roberta Ricciarelli
- Department of Experimental Medicine, Section of General Pathology, School of Medical and Pharmaceutical SciencesUniversity of GenoaGenoaItaly
- IRCCS Ospedale Policlinico San MartinoGenoaItaly
| |
Collapse
|
6
|
A New Bistable Switch Model of Alzheimer’s Disease Pathogenesis. Int J Mol Sci 2022; 23:ijms23137061. [PMID: 35806088 PMCID: PMC9267076 DOI: 10.3390/ijms23137061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 02/04/2023] Open
Abstract
We propose a model to explain the pathogenesis of Alzheimer’s disease (AD) based on the theory that any disease affecting a healthy organism originates from a bistable feedback loop that shifts the system from a physiological to a pathological condition. We focused on the known double inhibitory loop involving the cellular prion protein (PrPC) and the enzyme BACE1 that produces amyloid-beta (Aβ) peptides. BACE1 is inhibited by PrPC, but its inhibitory activity is lost when PrPC binds to Aβ oligomers (Aβo). Excessive Aβo formation would switch the loop to a pathogenic condition involving the Aβo-PrPC-mGluR5 complex, Fyn kinase activation, tau, and NMDAR phosphorylation, ultimately leading to neurodegeneration. Based on the emerging role of cyclic nucleotides in Aβ production, and thereby in synaptic plasticity and cognitive processes, cAMP and cGMP can be considered as modulatory factors capable of inducing the transition from a physiological steady state to a pathogenic one. This would imply that critical pharmacological targets for AD treatment lie within pathways that lead to an imbalance of cyclic nucleotides in neurons. If this hypothesis is confirmed, it will provide precise indications for the development of preventive or therapeutic treatments for the disease.
Collapse
|
7
|
Dai Z, Hu T, Su S, Liu J, Ma Y, Zhuo Y, Fang S, Wang Q, Mo Z, Pan H, Fang J. Comparative Metabolomics Analysis Reveals Key Metabolic Mechanisms and Protein Biomarkers in Alzheimer’s Disease. Front Pharmacol 2022; 13:904857. [PMID: 35694256 PMCID: PMC9174950 DOI: 10.3389/fphar.2022.904857] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 04/19/2022] [Indexed: 12/13/2022] Open
Abstract
Alzheimer’s disease (AD) is one of the most common progressive neurodegenerative diseases, accompanied by global alterations in metabolic profiles. In the past 10 years, over hundreds of metabolomics studies have been conducted to unravel metabolic changes in AD, which provides insight into the identification of potential biomarkers for diagnosis, treatment, and prognostic assessment. However, since different species may lead to systemic abnormalities in metabolomic profiles, it is urgently needed to perform a comparative metabolomics analysis between AD animal models and human patients. In this study, we integrated 78 metabolic profiles from public literatures, including 11 metabolomics studies in different AD mouse models and 67 metabolomics studies from AD patients. Metabolites and enrichment analysis were further conducted to reveal key metabolic pathways and metabolites in AD. We totally identified 14 key metabolites and 16 pathways that are both differentially significant in AD mouse models and patients. Moreover, we built a metabolite-target network to predict potential protein markers in AD. Finally, we validated HER2 and NDF2 as key protein markers in APP/PS1 mice. Overall, this study provides a comprehensive strategy for AD metabolomics research, contributing to understanding the pathological mechanism of AD.
Collapse
Affiliation(s)
- Zhao Dai
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Rheumatology, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tian Hu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shijie Su
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinman Liu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yinzhong Ma
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yue Zhuo
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Shuhuan Fang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhizhun Mo
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- *Correspondence: Zhizhun Mo, ; Huafeng Pan, ; Jiansong Fang,
| | - Huafeng Pan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Zhizhun Mo, ; Huafeng Pan, ; Jiansong Fang,
| | - Jiansong Fang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Zhizhun Mo, ; Huafeng Pan, ; Jiansong Fang,
| |
Collapse
|
8
|
Gorina YV, Khilazheva ED, Mosyagina AI, Kharitonova EV, Kapkaeva MR, Stelmashook EV, Isaev NK, Rozanova NA, Salmina AB. Impact of Lactate on Mitochondrial Activity in Endothelial Cells Exposed in vitro to the Acute Toxic Effect of beta-Amyloid. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022030218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
9
|
Montalto G, Caudano F, Sturla L, Bruzzone S, Salis A, Damonte G, Prickaerts J, Fedele E, Ricciarelli R. Protein kinase G phosphorylates the Alzheimer's disease-associated tau protein at distinct Ser/Thr sites. Biofactors 2021; 47:126-134. [PMID: 33469985 DOI: 10.1002/biof.1705] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 11/10/2022]
Abstract
Intraneuronal accumulation of hyperphosphorylated tau is a pathological hallmark of several neurodegenerative disorders, including Alzheimer's disease. Phosphorylation plays a crucial role in modulating the tau-microtubule interaction and the ability of the protein to aggregate, but despite efforts during the past decades, the real identity of the kynases involved in vivo remains uncertain. Here, for the first time, we demonstrate that the cGMP-dependent protein kinase G (PKG) phosphorylates tau in both in vitro and in vivo models. More intriguingly, we provide evidence that PKG phosphorylates tau at Ser214 but not at Ser202, a condition that could reduce the pathological aggregation of the protein shifting tau from a pro-aggregant to a neuroprotective anti-aggregant conformation.
Collapse
Affiliation(s)
- Giulia Montalto
- Section of General Pathology, Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Francesca Caudano
- Section of General Pathology, Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Laura Sturla
- Section of Biochemistry, Department of Experimental Medicine, Center for Excellence in Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Santina Bruzzone
- Section of Biochemistry, Department of Experimental Medicine, Center for Excellence in Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Annalisa Salis
- Section of Biochemistry, Department of Experimental Medicine, Center for Excellence in Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Gianluca Damonte
- Section of Biochemistry, Department of Experimental Medicine, Center for Excellence in Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Jos Prickaerts
- Department of Psychiatric and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Ernesto Fedele
- Section of Pharmacology and Toxicology, Department of Pharmacy, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Roberta Ricciarelli
- Section of General Pathology, Department of Experimental Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
10
|
Zhao Y, Chen H, Iqbal J, Liu X, Zhang H, Xiao S, Jin N, Yao F, Shen L. Targeted metabolomics study of early pathological features in hippocampus of triple transgenic Alzheimer's disease male mice. J Neurosci Res 2020; 99:927-946. [PMID: 33197957 DOI: 10.1002/jnr.24750] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 10/16/2020] [Accepted: 10/20/2020] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is a serious neurodegenerative disease in people of age 65 or above. The detailed etiology and pathogenesis of AD have not been elucidated yet. In this study, the hippocampi of 2- and 6-month-old triple transgenic Alzheimer's disease male mice and age-sex-matched wild-type (WT) mice were analyzed by using targeted metabolomics approach. Compared with WT mice, 24 and 60 metabolites were found with significant differences in 2- and 6-month-old AD mice. Among these, 14 metabolites were found common while 10 metabolites showed consistent variable trends in both groups. These differential metabolites are found associated with amino acid, lipid, vitamin, nucleotide-related base, neurotransmitter and energy metabolisms, and oxidative stress. The results suggest that these differential metabolites might play a critical role in AD pathophysiology, and may serve as potential biomarkers for AD. Moreover, the results highlight the involvement of abnormal purine, pyrimidine, arginine, and proline metabolism, along with glycerophospholipid metabolism in early pathology of AD. For the first time, several differential metabolites are found to be associated with AD in this study. Targeted metabolomics can be used for rapid and accurate quantitative analysis of specific target metabolites associated with AD.
Collapse
Affiliation(s)
- Yuxi Zhao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P.R. China
| | - Haiquan Chen
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P.R. China
| | - Javed Iqbal
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P.R. China
| | - Xukun Liu
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P.R. China
| | - Huajie Zhang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P.R. China.,Shenzhen Bay Laboratory, Shenzhen, P.R. China
| | - Shifeng Xiao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P.R. China
| | - Na Jin
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P.R. China
| | - Fang Yao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P.R. China.,Shenzhen Key Laboratory of Marine Biotechnology and Ecology, Shenzhen, P.R. China
| | - Liming Shen
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P.R. China
| |
Collapse
|
11
|
cAMP, cGMP and Amyloid β: Three Ideal Partners for Memory Formation. Trends Neurosci 2018; 41:255-266. [PMID: 29501262 DOI: 10.1016/j.tins.2018.02.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/25/2018] [Accepted: 02/01/2018] [Indexed: 02/03/2023]
Abstract
cAMP and cGMP are well established second messengers required for long-term potentiation (LTP) and memory formation/consolidation. By contrast, amyloid β (Aβ), mostly known as one of the main culprits for Alzheimer's disease (AD), has received relatively little attention in the context of plasticity and memory. Of note, however, low physiological concentrations of Aβ seem necessary for LTP induction and for memory formation. This should come as no surprise, since hormesis emerged as a central dogma in biology. Additionally, recent evidence indicates that Aβ is one of the downstream effectors for cAMP and cGMP to trigger synaptic plasticity and memory. We argue that these emerging findings depict a new scenario that should change the general view on the amyloidogenic pathway, and that could have significant implications for the understanding of AD and its pharmacological treatment in the future.
Collapse
|
12
|
Hansen RT, Zhang HT. The Past, Present, and Future of Phosphodiesterase-4 Modulation for Age-Induced Memory Loss. ADVANCES IN NEUROBIOLOGY 2018; 17:169-199. [PMID: 28956333 DOI: 10.1007/978-3-319-58811-7_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The purpose of this chapter is to highlight the state of progress for phosphodiesterase-4 (PDE4) modulation as a potential therapeutic for psychiatric illness, and to draw attention to particular hurdles and obstacles that must be overcome in future studies to develop PDE4-mediated therapeutics. Pathological and non-pathological related memory loss will be the focus of the chapter; however, we will at times also touch upon other psychiatric illnesses like anxiety and depression. First, we will provide a brief background of PDE4, and the rationale for its extensive study in cognition. Second, we will explore fundamental differences in individual PDE4 subtypes, and then begin to address differences between pathological and non-pathological aging. Alterations of cAMP/PDE4 signaling that occur within normal vs. pathological aging, and the potential for PDE4 modulation to combat these alterations within each context will be described. Finally, we will finish the chapter with obstacles that have hindered the field, and future studies and alternative viewpoints that need to be addressed. Overall, we hope this chapter will demonstrate the incredible complexity of PDE4 signaling in the brain, and will be useful in forming a strategy to develop future PDE4-mediated therapeutics for psychiatric illnesses.
Collapse
Affiliation(s)
- Rolf T Hansen
- Departments of Behavioral Medicine & Psychiatry and Physiology & Pharmacology, West Virginia University Health Sciences Center, 1 Medical Center Drive, Morgantown, WV, 26506-9137, USA
| | - Han-Ting Zhang
- Department of Behavioral Medicine and Psychiatry, West Virginia University Health Sciences Center, 1 Medical Center Drive, Morgantown, WV, 26506, USA. .,Department of Physiology and Pharmacology, West Virginia University Health Sciences Center, 1 Medical Center Drive, Morgantown, WV, 26506, USA. .,Institute of Pharmacology, Taishan Medical University, Taian, 271016, China.
| |
Collapse
|
13
|
Abstract
High levels of amyloid-β peptide (Aβ) have been related to Alzheimer's disease pathogenesis. However, in the healthy brain, low physiologically relevant concentrations of Aβ are necessary for long-term potentiation (LTP) and memory. Because cGMP plays a key role in these processes, here we investigated whether the cyclic nucleotide cGMP influences Aβ levels and function during LTP and memory. We demonstrate that the increase of cGMP levels by the phosphodiesterase-5 inhibitors sildenafil and vardenafil induces a parallel release of Aβ due to a change in the approximation of amyloid precursor protein (APP) and the β-site APP cleaving enzyme 1. Moreover, electrophysiological and behavioral studies performed on animals of both sexes showed that blocking Aβ function, by using anti-murine Aβ antibodies or APP knock-out mice, prevents the cGMP-dependent enhancement of LTP and memory. Our data suggest that cGMP positively regulates Aβ levels in the healthy brain which, in turn, boosts synaptic plasticity and memory.SIGNIFICANCE STATEMENT Amyloid-β (Aβ) is a key pathogenetic factor in Alzheimer's disease. However, low concentrations of endogenous Aβ, mimicking levels of the peptide in the healthy brain, enhance hippocampal long-term potentiation (LTP) and memory. Because the second messenger cGMP exerts a central role in LTP mechanisms, here we studied whether cGMP affects Aβ levels and function during LTP. We show that cGMP enhances Aβ production by increasing the APP/BACE-1 convergence in endolysosomal compartments. Moreover, the cGMP-induced enhancement of LTP and memory was disrupted by blockade of Aβ, suggesting that the physiological effect of the cyclic nucleotide on LTP and memory is dependent upon Aβ.
Collapse
|
14
|
Ricciarelli R, Fedele E. The Amyloid Cascade Hypothesis in Alzheimer's Disease: It's Time to Change Our Mind. Curr Neuropharmacol 2017; 15:926-935. [PMID: 28093977 PMCID: PMC5652035 DOI: 10.2174/1570159x15666170116143743] [Citation(s) in RCA: 218] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 01/04/2017] [Accepted: 01/14/2017] [Indexed: 01/18/2023] Open
Abstract
Since its discovery in 1984, the beta amyloid peptide has treaded the boards of neurosciences as the star molecule in Alzheimer's disease pathogenesis. In the last decade, however, this vision has been challenged by evidence-based medicine showing the almost complete failure of clinical trials that experimented anti-amyloid therapies with great hopes. Moreover, data have accumulated which clearly indicate that this small peptide plays a key role in the physiological processes of memory formation. In the present review, we will discuss the different aspects of the amyloid cascade hypothesis, highlighting its pros and cons, and we will analyse the results of the therapeutic approaches attempted to date that should change the direction of Alzheimer's disease research in the future.
Collapse
Affiliation(s)
- Roberta Ricciarelli
- Department of Experimental Medicine, Section of General Pathology, University of Genova, Genova, Italy
| | - Ernesto Fedele
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy
- Center of Excellence for Biomedical Research, University of Genova, Genova, Italy
| |
Collapse
|
15
|
Non-amyloidogenic effects of α2 adrenergic agonists: implications for brimonidine-mediated neuroprotection. Cell Death Dis 2016; 7:e2514. [PMID: 27929541 PMCID: PMC5260990 DOI: 10.1038/cddis.2016.397] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Revised: 10/18/2016] [Accepted: 10/24/2016] [Indexed: 12/18/2022]
Abstract
The amyloid beta (Aβ) pathway is strongly implicated in neurodegenerative conditions such as Alzheimer's disease and more recently, glaucoma. Here, we identify the α2 adrenergic receptor agonists (α2ARA) used to lower intraocular pressure can prevent retinal ganglion cell (RGC) death via the non-amyloidogenic Aβ-pathway. Neuroprotective effects were confirmed in vivo and in vitro in different glaucoma-related models using α2ARAs brimonidine (BMD), clonidine (Clo) and dexmedetomidine. α2ARA treatment significantly reduced RGC apoptosis in experimental-glaucoma models by 97.7% and 92.8% (BMD, P<0.01) and 98% and 92.3% (Clo, P<0.01)) at 3 and 8 weeks, respectively. A reduction was seen in an experimental Aβ-induced neurotoxicity model (67% BMD and 88.6% Clo, both P<0.01, respectively), and in vitro, where α2ARAs significantly (P<0.05) prevented cell death, under both hypoxic (CoCl2) and stress (UV) conditions. In experimental-glaucoma, BMD induced ninefold and 25-fold and 36-fold and fourfold reductions in Aβ and amyloid precursor protein (APP) levels at 3 and 8 weeks, respectively, in the RGC layer, with similar results with Clo, and in vitro with all three α2ARAs. BMD significantly increased soluble APPα (sAPPα) levels at 3 and 8 weeks (2.1 and 1.6-fold) in vivo and in vitro with the CoCl2 and UV-light insults. Furthermore, treatment of UV-insulted cells with an sAPPα antibody significantly reduced cell viability compared with BMD-treated control (52%), co-treatment (33%) and untreated control (27%). Finally, we show that α2ARAs modulate levels of laminin and MMP-9 in RGCs, potentially linked to changes in Aβ through APP processing. Together, these results provide new evidence that α2ARAs are neuroprotective through their effects on the Aβ pathway and sAPPα, which to our knowledge, is the first description. Studies have identified the need for α-secretase activators and sAPPα-mimetics in neurodegeneration; α2ARAs, already clinically available, present a promising therapy, with applications not only to reducing RGC death in glaucoma but also other neurodegenerative processes involving Aβ.
Collapse
|
16
|
Kempf SJ, Metaxas A, Ibáñez-Vea M, Darvesh S, Finsen B, Larsen MR. An integrated proteomics approach shows synaptic plasticity changes in an APP/PS1 Alzheimer's mouse model. Oncotarget 2016; 7:33627-48. [PMID: 27144524 PMCID: PMC5085108 DOI: 10.18632/oncotarget.9092] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 04/19/2016] [Indexed: 02/07/2023] Open
Abstract
The aim of this study was to elucidate the molecular signature of Alzheimer's disease-associated amyloid pathology.We used the double APPswe/PS1ΔE9 mouse, a widely used model of cerebral amyloidosis, to compare changes in proteome, including global phosphorylation and sialylated N-linked glycosylation patterns, pathway-focused transcriptome and neurological disease-associated miRNAome with age-matched controls in neocortex, hippocampus, olfactory bulb and brainstem. We report that signalling pathways related to synaptic functions associated with dendritic spine morphology, neurite outgrowth, long-term potentiation, CREB signalling and cytoskeletal dynamics were altered in 12 month old APPswe/PS1ΔE9 mice, particularly in the neocortex and olfactory bulb. This was associated with cerebral amyloidosis as well as formation of argyrophilic tangle-like structures and microglial clustering in all brain regions, except for brainstem. These responses may be epigenetically modulated by the interaction with a number of miRNAs regulating spine restructuring, Aβ expression and neuroinflammation.We suggest that these changes could be associated with development of cognitive dysfunction in early disease states in patients with Alzheimer's disease.
Collapse
Affiliation(s)
- Stefan J. Kempf
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
| | - Athanasios Metaxas
- Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
| | - María Ibáñez-Vea
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
| | - Sultan Darvesh
- Department of Medical Neuroscience, Dalhousie University, Halifax, NS, Canada
- Department of Medicine (Neurology and Geriatric Medicine), Dalhousie University, Halifax, NS, Canada
| | - Bente Finsen
- Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
| | - Martin R. Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
| |
Collapse
|
17
|
Fedele E, Rivera D, Marengo B, Pronzato MA, Ricciarelli R. Amyloid β: Walking on the dark side of the moon. Mech Ageing Dev 2015; 152:1-4. [DOI: 10.1016/j.mad.2015.09.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 08/31/2015] [Accepted: 09/03/2015] [Indexed: 12/20/2022]
|
18
|
Rivera D, Fedele E, Marinari UM, Pronzato MA, Ricciarelli R. Evaluating the role of hnRNP-C and FMRP in the cAMP-induced APP metabolism. Biofactors 2015; 41:121-6. [PMID: 25809670 DOI: 10.1002/biof.1207] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 02/27/2015] [Indexed: 11/09/2022]
Abstract
Cyclic adenosine monophosphate (cAMP) modulates synaptic plasticity and memory and manipulation of the cAMP/protein kinase A/cAMP responsive element binding protein pathway significantly affects cognitive functions. Notably, cAMP can increase the expression of the amyloid precursor protein (APP), whose proteolytic processing gives rise to amyloid beta (Aβ) peptides. Despite playing a pathogenic role in Alzheimer's disease, physiological concentrations of Aβ are necessary for the cAMP-mediated regulation of long-term potentiation, supporting the existence of a novel cAMP/APP/Aβ cascade with a crucial role in memory formation. However, the molecular mechanisms by which cAMP stimulates APP expression and Aβ production remain unclear. Here, we investigated whether hnRNP-C and FMRP, two RNA-binding proteins largely involved in the expression of APP, are the cAMP effectors inducing the protein synthesis of APP. Using RNA immunoprecipitation and RNA-silencing approaches, we found that neither hnRNP-C nor FMRP is required for cAMP to stimulate APP and Aβ production.
Collapse
Affiliation(s)
- D Rivera
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | | | | | | | | |
Collapse
|
19
|
Bergersen LH. Lactate transport and signaling in the brain: potential therapeutic targets and roles in body-brain interaction. J Cereb Blood Flow Metab 2015; 35:176-85. [PMID: 25425080 PMCID: PMC4426752 DOI: 10.1038/jcbfm.2014.206] [Citation(s) in RCA: 159] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 09/30/2014] [Accepted: 10/24/2014] [Indexed: 12/26/2022]
Abstract
Lactate acts as a 'buffer' between glycolysis and oxidative metabolism. In addition to being exchanged as a fuel by the monocarboxylate transporters (MCTs) between cells and tissues with different glycolytic and oxidative rates, lactate may be a 'volume transmitter' of brain signals. According to some, lactate is a preferred fuel for brain metabolism. Immediately after brain activation, the rate of glycolysis exceeds oxidation, leading to net production of lactate. At physical rest, there is a net efflux of lactate from the brain into the blood stream. But when blood lactate levels rise, such as in physical exercise, there is net influx of lactate from blood to brain, where the lactate is used for energy production and myelin formation. Lactate binds to the lactate receptor GPR81 aka hydroxycarboxylic acid receptor (HCAR1) on brain cells and cerebral blood vessels, and regulates the levels of cAMP. The localization and function of HCAR1 and the three MCTs (MCT1, MCT2, and MCT4) expressed in brain constitute the focus of this review. They are possible targets for new therapeutic drugs and interventions. The author proposes that lactate actions in the brain through MCTs and the lactate receptor underlie part of the favorable effects on the brain resulting from physical exercise.
Collapse
Affiliation(s)
- Linda Hildegard Bergersen
- 1] The Brain and Muscle Energy Group, SN-Lab, Department of Anatomy, Institute of Basic Medical Sciences, Healthy Brain Ageing Centre, University of Oslo, Oslo, Norway [2] Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark [3] Center for Healthy Aging, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark [4] The Brain and Muscle Energy Group, Department of Oral Biology, University of Oslo, Oslo, Norway
| |
Collapse
|
20
|
Ansoleaga B, Jové M, Schlüter A, Garcia-Esparcia P, Moreno J, Pujol A, Pamplona R, Portero-Otín M, Ferrer I. Deregulation of purine metabolism in Alzheimer's disease. Neurobiol Aging 2014; 36:68-80. [PMID: 25311278 DOI: 10.1016/j.neurobiolaging.2014.08.004] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 08/05/2014] [Indexed: 12/30/2022]
Abstract
The neuroprotective role of adenosine and the deregulation of adenosine receptors in Alzheimer's disease (AD) have been extensively studied in recent years. However, little is known about the involvement of purine metabolism in AD. We started by analyzing gene expression in the entorhinal cortex of human controls and AD cases with whole-transcript expression arrays. Once we identified deregulation of the cluster purine metabolism, messenger RNA expression levels of 23 purine metabolism genes were analyzed with qRT-PCR in the entorhinal cortex, frontal cortex area 8, and precuneus at stages I-II, III-IV, and V-VI of Braak and Braak and controls. APRT, DGUOK, POLR3B, ENTPD3, AK5, NME1, NME3, NME5, NME7, and ENTPD2 messenger RNAs were deregulated, with regional variations, in AD cases when compared with controls. In addition, liquid chromatography mass spectrometry based metabolomics in the entorhinal cortex identified altered levels of dGMP, glycine, xanthosine, inosine diphosphate, guanine, and deoxyguanosine, all implicated in this pathway. Our results indicate stage- and region-dependent deregulation of purine metabolism in AD.
Collapse
Affiliation(s)
- Belén Ansoleaga
- Institute of Neuropathology, Bellvitge University Hospital-Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Spain
| | - Mariona Jové
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida, Lleida, Spain
| | - Agatha Schlüter
- Neurometabolic Diseases Laboratory, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Paula Garcia-Esparcia
- Institute of Neuropathology, Bellvitge University Hospital-Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Spain
| | - Jesús Moreno
- Institute of Neuropathology, Bellvitge University Hospital-Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Spain
| | - Aurora Pujol
- Neurometabolic Diseases Laboratory, IDIBELL, L'Hospitalet de Llobregat, Spain; Centre for Biomedical Research on Rare Diseases (CIBERER), Institute Carlos III, Madrid, Spain; Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Reinald Pamplona
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida, Lleida, Spain
| | - Manuel Portero-Otín
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida, Lleida, Spain
| | - Isidre Ferrer
- Institute of Neuropathology, Bellvitge University Hospital-Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Spain; University of Barcelona, Bellvitge Campus, L'Hospitalet de Llobregat, Spain; Centre for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED), Institute Carlos III, Madrid, Spain.
| |
Collapse
|
21
|
Ricciarelli R, Puzzo D, Bruno O, Canepa E, Gardella E, Rivera D, Privitera L, Domenicotti C, Marengo B, Marinari UM, Palmeri A, Pronzato MA, Arancio O, Fedele E. A novel mechanism for cyclic adenosine monophosphate-mediated memory formation: Role of amyloid beta. Ann Neurol 2014; 75:602-7. [PMID: 24591104 DOI: 10.1002/ana.24130] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 02/21/2014] [Accepted: 02/24/2014] [Indexed: 12/22/2022]
Abstract
Cyclic adenosine monophosphate (cAMP) regulates long-term potentiation (LTP) and ameliorates memory in healthy and diseased brain. Increasing evidence shows that, under physiological conditions, low concentrations of amyloid β (Aβ) are necessary for LTP expression and memory formation. Here, we report that cAMP controls amyloid precursor protein (APP) translation and Aβ levels, and that the modulatory effects of cAMP on LTP occur through the stimulation of APP synthesis and Aβ production.
Collapse
Affiliation(s)
- Roberta Ricciarelli
- Department of Experimental Medicine, Section of General Pathology, University of Genoa, Genoa, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Effect of phosphodiesterase-5 inhibition on apoptosis and beta amyloid load in aged mice. Neurobiol Aging 2013; 35:520-31. [PMID: 24112792 DOI: 10.1016/j.neurobiolaging.2013.09.002] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 09/05/2013] [Accepted: 09/06/2013] [Indexed: 01/06/2023]
Abstract
Age-related cognitive decline is accompanied by an increase of neuronal apoptosis and a dysregulation of neuroplasticity-related molecules such as brain-derived neurotrophic factor and neurotoxic factors including beta amyloid (Aβ) peptide. Because it has been previously demonstrated that phosphodiesterase-5 inhibitors (PDE5-Is) protect against hippocampal synaptic dysfunction and memory deficits in mouse models of Alzheimer's disease and physiological aging, we investigated the effect of a treatment with the PDE5-I, sildenafil, on cell death, pro- and antiapoptotic molecules, and Aβ production. We demonstrated that chronic intraperitoneal injection of sildenafil (3 mg/kg for 3 weeks) decreased terminal deoxyuridine triphosphate nick end labeling-positive cells in the CA1 hippocampal area of 26-30-month-old mice, downregulating the proapoptotic proteins, caspase-3 and B-cell lymphoma 2-associated X, and increasing antiapoptotic molecules such as B-cell lymphoma protein-2 and brain-derived neurotrophic factor. Also, sildenafil reverted the shifting of amyloid precursor protein processing toward Aβ42 production and the increase of the Aβ42:Aβ40 ratio in aged mice. Our data suggest that PDE5-I might be beneficial to treat age-related detrimental features in a physiological mouse model of aging.
Collapse
|