1
|
Placzek M, Wilton DK, Weïwer M, Manter MA, Reid SE, Meyer CJ, Campbell AJ, Bajrami B, Bigot A, Bricault S, Fayet A, Frouin A, Gergits F, Gupta M, Jiang W, Melanson M, Romano CD, Riley MM, Wang JM, Wey HY, Wagner FF, Stevens B, Hooker JM. A Fast-Binding, Functionally Reversible, COX-2 Radiotracer for CNS PET Imaging. ACS CENTRAL SCIENCE 2024; 10:1105-1114. [PMID: 38799654 PMCID: PMC11117721 DOI: 10.1021/acscentsci.3c01564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 04/12/2024] [Accepted: 04/18/2024] [Indexed: 05/29/2024]
Abstract
Cyclooxygenase-2 (COX-2) is an enzyme that plays a pivotal role in peripheral inflammation and pain via the prostaglandin pathway. In the central nervous system (CNS), COX-2 is implicated in neurodegenerative and psychiatric disorders as a potential therapeutic target and biomarker. However, clinical studies with COX-2 have yielded inconsistent results, partly due to limited mechanistic understanding of how COX-2 activity relates to CNS pathology. Therefore, developing COX-2 positron emission tomography (PET) radiotracers for human neuroimaging is of interest. This study introduces [11C]BRD1158, which is a potent and uniquely fast-binding, selective COX-2 PET radiotracer. [11C]BRD1158 was developed by prioritizing potency at COX-2, isoform selectivity over COX-1, fast binding kinetics, and free fraction in the brain. Evaluated through in vivo PET neuroimaging in rodent models with human COX-2 overexpression, [11C]BRD1158 demonstrated high brain uptake, fast target-engagement, functional reversibility, and excellent specific binding, which is advantageous for human imaging applications. Lastly, post-mortem samples from Huntington's disease (HD) patients and preclinical HD mouse models showed that COX-2 levels were elevated specifically in disease-affected brain regions, primarily from increased expression in microglia. These findings indicate that COX-2 holds promise as a novel clinical marker of HD onset and progression, one of many potential applications of [11C]BRD1158 human PET.
Collapse
Affiliation(s)
- Michael
S. Placzek
- Athinoula
A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Daniel K. Wilton
- Department
of Neurology and F.M. Kirby Neurobiology Center, Boston Children’s
Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Michel Weïwer
- Center
for the Development of Therapeutics, Broad
Institute of MIT and Harvard, 75 Ames Street, Cambridge, Massachusetts 02142, United States
| | - Mariah A. Manter
- Athinoula
A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, United States
- Lurie
Center for Autism, 1
Maguire Road, Lexington, Massachusetts 02421, United States
- Massachusetts
General Hospital, 55
Fruit St., Boston, Massachusetts 02114, United States
| | - Sarah E. Reid
- Athinoula
A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Christopher J. Meyer
- Center
for the Development of Therapeutics, Broad
Institute of MIT and Harvard, 75 Ames Street, Cambridge, Massachusetts 02142, United States
| | - Arthur J. Campbell
- Center
for the Development of Therapeutics, Broad
Institute of MIT and Harvard, 75 Ames Street, Cambridge, Massachusetts 02142, United States
| | - Besnik Bajrami
- Center
for the Development of Therapeutics, Broad
Institute of MIT and Harvard, 75 Ames Street, Cambridge, Massachusetts 02142, United States
| | - Antoine Bigot
- Center
for the Development of Therapeutics, Broad
Institute of MIT and Harvard, 75 Ames Street, Cambridge, Massachusetts 02142, United States
| | - Sarah Bricault
- Athinoula
A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Agathe Fayet
- Center
for the Development of Therapeutics, Broad
Institute of MIT and Harvard, 75 Ames Street, Cambridge, Massachusetts 02142, United States
| | - Arnaud Frouin
- Department
of Neurology and F.M. Kirby Neurobiology Center, Boston Children’s
Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Frederick Gergits
- Department
of Neurology and F.M. Kirby Neurobiology Center, Boston Children’s
Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Mehak Gupta
- Center
for the Development of Therapeutics, Broad
Institute of MIT and Harvard, 75 Ames Street, Cambridge, Massachusetts 02142, United States
| | - Wei Jiang
- Center
for the Development of Therapeutics, Broad
Institute of MIT and Harvard, 75 Ames Street, Cambridge, Massachusetts 02142, United States
| | - Michelle Melanson
- Center
for the Development of Therapeutics, Broad
Institute of MIT and Harvard, 75 Ames Street, Cambridge, Massachusetts 02142, United States
| | - Chiara D. Romano
- Center
for the Development of Therapeutics, Broad
Institute of MIT and Harvard, 75 Ames Street, Cambridge, Massachusetts 02142, United States
| | - Misha M. Riley
- Athinoula
A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Jessica M. Wang
- Athinoula
A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Hsiao-Ying Wey
- Athinoula
A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Florence F. Wagner
- Center
for the Development of Therapeutics, Broad
Institute of MIT and Harvard, 75 Ames Street, Cambridge, Massachusetts 02142, United States
| | - Beth Stevens
- Department
of Neurology and F.M. Kirby Neurobiology Center, Boston Children’s
Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Stanley
Center for Psychiatric Research, Broad Institute
of MIT and Harvard, 75 Ames Street, Cambridge, Massachusetts 02142, United
- Howard
Hughes Medical Institute, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Jacob M. Hooker
- Athinoula
A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, United States
- Lurie
Center for Autism, 1
Maguire Road, Lexington, Massachusetts 02421, United States
- Massachusetts
General Hospital, 55
Fruit St., Boston, Massachusetts 02114, United States
| |
Collapse
|
2
|
Jiang C, Zhang B, Jiang W, Liu P, Kong Y, Zhang J, Teng W. Metal ion stimulation-related gene signatures correlate with clinical and immunologic characteristics of glioma. Heliyon 2024; 10:e27189. [PMID: 38533032 PMCID: PMC10963200 DOI: 10.1016/j.heliyon.2024.e27189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 03/28/2024] Open
Abstract
Background Environmental factors serve as one of the important pathogenic factors for gliomas. Yet people focus only on the effect of electromagnetic radiation on its pathogenicity, while metals in the environment are neglected. This study aimed to investigate the relationship between metal ion stimulation and the clinical characteristics and immune status of GM patients. Methods Firstly, mRNA expression profiles of GM patients and normal subjects were obtained from Chinese GM Genome Atlas (CGGA) and Gene Expression Omnibus (GEO) to identify differentially expressed metal ion stimulation-related genes(DEMISGs). Secondly, two molecular subtypes were identified and validated based on these DEMISGs using consensus clustering. Diagnostic and prognostic models for GM were constructed after screening these features based on machine learning. Finally, supervised classification and unsupervised clustering were combined to classify and predict the grade of GM based on SHAP values. Results GM patients are divided into two different response states to metal ion stimulation, M1 and M2, which are related to the grade and IDH status of the GM. Six genes with diagnostic value were obtained: SLC30A3, CRHBP, SYT13, DLG2, CDK1, and WNT5A. The AUC in the external validation set was higher than 0.90. The SHAP value improves the performance of classification prediction. Conclusion The gene features associated with metal ion stimulation are related to the clinical and immune characteristics of transgenic patients. XGboost/LightGBM Kmeans has a higher classification prediction accuracy in predicting glioma grades compared to using purely supervised classification techniques.
Collapse
Affiliation(s)
- Chengzhi Jiang
- Shandong Second Medical University, Weifang, Shandong, 261053, People's Republic of China
| | - Binbin Zhang
- Qingdao Municipal Hospital (Group), Qingdao, Shandong, 266000, People's Republic of China
| | - Wenjuan Jiang
- Qingdao Municipal Hospital (Group), Qingdao, Shandong, 266000, People's Republic of China
| | - Pengtao Liu
- Shandong Second Medical University, Weifang, Shandong, 261053, People's Republic of China
| | - Yujia Kong
- Shandong Second Medical University, Weifang, Shandong, 261053, People's Republic of China
| | - Jianhua Zhang
- Jining Medical University, Jining, Shandong, 272067, People's Republic of China
| | - Wenjie Teng
- Shandong Second Medical University, Weifang, Shandong, 261053, People's Republic of China
| |
Collapse
|
3
|
André S, Verteneuil S, Ris L, Kahvecioglu ZC, Nonclercq D, De Winter J, Vander Elst L, Laurent S, Muller RN, Burtea C. Modulation of Cytosolic Phospholipase A2 as a Potential Therapeutic Strategy for Alzheimer's Disease. J Alzheimers Dis Rep 2023; 7:1395-1426. [PMID: 38225969 PMCID: PMC10789292 DOI: 10.3233/adr-230075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 11/17/2023] [Indexed: 01/17/2024] Open
Abstract
Background Alzheimer's disease (AD) is a neurodegenerative disorder lacking any curative treatment up to now. Indeed, actual medication given to the patients alleviates only symptoms. The cytosolic phospholipase A2 (cPLA2-IVA) appears as a pivotal player situated at the center of pathological pathways leading to AD and its inhibition could be a promising therapeutic approach. Objective A cPLA2-IVA inhibiting peptide was identified in the present work, aiming to develop an original therapeutic strategy. Methods We targeted the cPLA2-IVA using the phage display technology. The hit peptide PLP25 was first validated in vitro (arachidonic acid dosage [AA], cPLA2-IVA cellular translocation) before being tested in vivo. We evaluated spatial memory using the Barnes maze, amyloid deposits by MRI and immunohistochemistry (IHC), and other important biomarkers such as the cPLA2-IVA itself, the NMDA receptor, AβPP and tau by IHC after i.v. injection in APP/PS1 mice. Results Showing a high affinity for the C2 domain of this enzyme, the peptide PLP25 exhibited an inhibitory effect on cPLA2-IVA activity by blocking its binding to its substrate, resulting in a decreased release of AA. Coupled to a vector peptide (LRPep2) in order to optimize brain access, we showed an improvement of cognitive abilities of APP/PS1 mice, which also exhibited a decreased number of amyloid plaques, a restored expression of cPLA2-IVA, and a favorable effect on NMDA receptor expression and tau protein phosphorylation. Conclusions cPLA2-IVA inhibition through PLP25 peptide could be a promising therapeutic strategy for AD.
Collapse
Affiliation(s)
- Séverine André
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
| | - Sébastien Verteneuil
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
| | - Laurence Ris
- Department of Neurosciences, University of Mons, Research Institute for Health Science and Technologies, Mons, Belgium
| | - Zehra-Cagla Kahvecioglu
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
| | | | - Julien De Winter
- Organic Synthesis and Mass Spectrometry Laboratory (SMOs), University of Mons-UMONS, Mons, Belgium
| | - Luce Vander Elst
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
| | - Sophie Laurent
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
- Center for Microscopy and Molecular Imaging, Gosselies, Belgium
| | - Robert N. Muller
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
- Center for Microscopy and Molecular Imaging, Gosselies, Belgium
| | - Carmen Burtea
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
| |
Collapse
|
4
|
Moussa N, Dayoub N. Exploring the role of COX-2 in Alzheimer's disease: Potential therapeutic implications of COX-2 inhibitors. Saudi Pharm J 2023; 31:101729. [PMID: 37638222 PMCID: PMC10448476 DOI: 10.1016/j.jsps.2023.101729] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/28/2023] [Indexed: 08/29/2023] Open
Abstract
This review highlights the potential role of cyclooxygenase-2 enzyme (COX-2) in the pathogenesis of Alzheimer's disease (AD) and the potential therapeutic use of non-steroidal anti-inflammatory drugs (NSAIDs) in the management of AD. In addition to COX-2 enzymes role in inflammation, the formation of amyloid plaques and neurofibrillary tangles in the brain, the review emphasizes that COXs-2 have a crucial role in normal synaptic activity and plasticity, and have a relationship with acetylcholine, tau protein, and beta-amyloid (Aβ) which are the main causes of Alzheimer's disease. Furthermore, the review points out that COX-2 enzymes have a relationship with kinase enzymes, including Cyclin Dependent Kinase 5 (CDK5) and Glycogen Synthase Kinase 3β (GSK3β), which are known to play a role in tau phosphorylation and are strongly associated with Alzheimer's disease. Therefore, the use of drugs like NSAIDs may be a hopeful approach for managing AD. However, results from studies examining the effectiveness of NSAIDs in treating AD have been mixed and further research is needed to fully understand the mechanisms by which COX-2 and NSAIDs may be involved in the development and progression of AD and to identify new therapeutic strategies.
Collapse
Affiliation(s)
- Nathalie Moussa
- Department of Pharmaceutical Chemistry and Drug Control, University of Manara, Latakia, Syria
| | - Ninar Dayoub
- Faculty of Pharmacy, University of AL Andalus for Medical Science, Tartus, Syria
| |
Collapse
|
5
|
Kubota H, Kunisawa K, Wulaer B, Hasegawa M, Kurahashi H, Sakata T, Tezuka H, Kugita M, Nagao S, Nagai T, Furuyashiki T, Narumiya S, Saito K, Nabeshima T, Mouri A. High salt induces cognitive impairment via the interaction of the angiotensin II-AT 1 and prostaglandin E2-EP 1 systems. Br J Pharmacol 2023; 180:2393-2411. [PMID: 37076133 DOI: 10.1111/bph.16093] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/05/2023] [Accepted: 04/13/2023] [Indexed: 04/21/2023] Open
Abstract
BACKGROUND AND PURPOSE High salt (HS) intake has been associated with hypertension and cognitive impairment. It is well known that the angiotensin II (Ang II)-AT1 receptor and prostaglandin E2 (PGE2)-EP1 receptor systems are involved in hypertension and neurotoxicity. However, the involvement of these systems in HS-mediated hypertension and emotional and cognitive impairments remains unclear. EXPERIMENTAL APPROACH Mice were loaded with HS solution (2% NaCl drinking water) for 12 weeks, and blood pressure was monitored. Subsequently, effects of HS intake on emotional and cognitive function and tau phosphorylation in the prefrontal cortex (PFC) and hippocampus (HIP) were investigated. The involvement of Ang II-AT1 and PGE2-EP1 systems in HS-induced hypertension and neuronal and behavioural impairments was examined by treatment with losartan, an AT1 receptor blocker (ARB), or EP1 gene knockout. KEY RESULTS We demonstrate that hypertension and impaired social behaviour and object recognition memory following HS intake may be associated with tau hyperphosphorylation, decreased phosphorylation of Ca2+ /calmodulin-dependent protein kinase II (CaMKII), and postsynaptic density protein 95 (PSD95) expression in the PFC and HIP of mice. These changes were blocked by pharmacological treatment with losartan or EP1 receptor gene knockout. CONCLUSIONS AND IMPLICATIONS Our findings suggest that the interaction of Ang II-AT1 receptor and PGE2-EP1 receptor systems could be novel therapeutic targets for hypertension-induced cognitive impairment.
Collapse
Affiliation(s)
- Hisayoshi Kubota
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Toyoake, Aichi, Japan
| | - Kazuo Kunisawa
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Toyoake, Aichi, Japan
| | - Bolati Wulaer
- Laboratory of Health and Medical Science Innovation (HMSI), Fujita Health University Graduate School of Health Science, Toyoake, Aichi, Japan
| | - Masaya Hasegawa
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Toyoake, Aichi, Japan
| | - Hitomi Kurahashi
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Toyoake, Aichi, Japan
| | - Takatoshi Sakata
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Toyoake, Aichi, Japan
| | - Hiroyuki Tezuka
- Department of Cellular Function Analysis, Research Promotion and Support Headquarters, Fujita Health University, Toyoake, Aichi, Japan
| | - Masanori Kugita
- Education and Research Facility of Animal Models for Human Diseases, Center for Research Promotion and Support, Fujita Health University, Toyoake, Aichi, Japan
| | - Shizuko Nagao
- Education and Research Facility of Animal Models for Human Diseases, Center for Research Promotion and Support, Fujita Health University, Toyoake, Aichi, Japan
| | - Taku Nagai
- Division of Behavioral Neuropharmacology International Center for Brain Science (ICBS), Fujita Health University, Toyoake, Aichi, Japan
| | - Tomoyuki Furuyashiki
- Division of Pharmacology, Graduate School of Medicine, Kobe University, Kobe, Hyogo, Japan
| | - Shuh Narumiya
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto, Kyoto, Japan
| | - Kuniaki Saito
- Laboratory of Health and Medical Science Innovation (HMSI), Fujita Health University Graduate School of Health Science, Toyoake, Aichi, Japan
- Department of Disease Control and Prevention, Fujita Health University Graduate School of Health Science, Toyoake, Aichi, Japan
- Japanese Drug Organization of Appropriate Use and Research, Nagoya, Aichi, Japan
| | - Toshitaka Nabeshima
- Laboratory of Health and Medical Science Innovation (HMSI), Fujita Health University Graduate School of Health Science, Toyoake, Aichi, Japan
- Japanese Drug Organization of Appropriate Use and Research, Nagoya, Aichi, Japan
| | - Akihiro Mouri
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Toyoake, Aichi, Japan
- Japanese Drug Organization of Appropriate Use and Research, Nagoya, Aichi, Japan
| |
Collapse
|
6
|
Mushtaq U. EP1 receptor: Devil in emperors coat. J Cell Biochem 2023; 124:1105-1114. [PMID: 37450673 DOI: 10.1002/jcb.30436] [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: 01/23/2023] [Revised: 05/20/2023] [Accepted: 06/06/2023] [Indexed: 07/18/2023]
Abstract
EP1 receptor belongs to prostanoid receptors and is activated by prostaglandin E2. The receptor performs contrasting functions in central nervous system (CNS) and other tissues. Although the receptor is neurotoxic and proapoptotic in CNS, it has also been reported to act in an antiapoptotic manner by modulating cell survival, proliferation, invasion, and migration in different types of cancers. The receptor mediates its neurotoxic effects by increasing cytosolic Ca2+ levels, leading to the activation of its downstream target, protein kinase C, in different neurological disorders including Alzheimer's disease, Parkinson's disease, stroke, amyotrophic lateral sclerosis, and epilepsy. Antagonists ONO-8713, SC51089, and SC51322 against EP1 receptor ameliorate the neurotoxic effect by attenuating the neuroinflammation. The receptor also shows increased expression in different types of cancers and has been found to activate different signaling pathways, which lead to the development, progression, and metastasis of different cancers. The receptor stimulates the cell survival pathway by phosphorylating the AKT and PTEN (phosphatase and tensin homolog deleted on chromosome 10) signaling pathways. Although there are limited studies about this receptor and not a single clinical trial has been targeting the EP1 receptor for different neurological disorders or cancer, the receptor is appearing as a potential candidate for therapeutic targets. The aim of this article is to review the recent progress in understanding the pathogenic roles of EP1 receptors in different pathological conditions.
Collapse
Affiliation(s)
- Umar Mushtaq
- Department of Biotechnology, Central University of Kashmir, Ganderbal, India
| |
Collapse
|
7
|
Montero-Cosme TG, Pascual-Mathey LI, Hernández-Aguilar ME, Herrera-Covarrubias D, Rojas-Durán F, Aranda-Abreu GE. Potential drugs for the treatment of Alzheimer's disease. Pharmacol Rep 2023; 75:544-559. [PMID: 37005970 DOI: 10.1007/s43440-023-00481-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/24/2023] [Accepted: 03/26/2023] [Indexed: 04/04/2023]
Abstract
It is well known that amyloid precursor protein (APP), the enzyme β-secretase 1 (BACE1), cyclooxygenase 2 (COX-2), nicastrin (NCT), and hyperphosphorylated tau protein (p-tau) are closely related to the development of Alzheimer's disease (AD). In addition, recent evidence shows that neuroinflammation also contributes to the pathogenesis of AD. Although the mechanism is not clearly known, such inflammation could alter the activity of the aforementioned molecules. Therefore, the use of anti-inflammatory agents could slow the progression of the disease. Nimesulide, resveratrol, and citalopram are three anti-inflammatory agents that could contribute to a decrease in neuroinflammation and consequently to a decrease in the overexpression of APP, BACE1, COX-2, NCT, and p-Tau, as they possess anti-inflammatory effects that could regulate the expression of APP, BACE1, COX-2, NCT, and p-Tau of potent pro-inflammatory markers indirectly involved in the expression of APP, BACE1, NCT, COX-2, and p-Tau; therefore, their use could be beneficial as preventive treatment as well as in the early stages of AD.
Collapse
Affiliation(s)
| | | | | | | | - Fausto Rojas-Durán
- Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa, Veracruz, México
| | | |
Collapse
|
8
|
Sahbani K, Cardozo CP, Bauman WA, Tawfeek HA. Inhibition of TGF-β Signaling Attenuates Disuse-induced Trabecular Bone Loss After Spinal Cord Injury in Male Mice. Endocrinology 2022; 163:bqab230. [PMID: 34791098 DOI: 10.1210/endocr/bqab230] [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: 09/17/2021] [Indexed: 11/19/2022]
Abstract
Bone loss is one of the most common complications of immobilization after spinal cord injury (SCI). Whether transforming growth factor (TGF)-β signaling plays a role in SCI-induced disuse bone loss has not been determined. Thus, 16-week-old male mice underwent sham or spinal cord contusion injury to cause complete hindlimb paralysis. Five days later, 10 mg/kg/day control (IgG) or anti-TGF-β1,2,3 neutralizing antibody (1D11) was administered twice weekly for 4 weeks. Femurs were examined by micro-computed tomography (micro-CT) scanning and histology. Bone marrow (BM) supernatants were analyzed by enzyme-linked immunosorbent assay for levels of procollagen type 1 intact N-terminal propeptide (P1NP), tartrate-resistant acid phosphatase (TRAcP-5b), receptor activator of nuclear factor-kappa B ligand (RANKL), osteoprotegerin (OPG), and prostaglandin E2 (PGE2). Distal femoral micro-CT analysis showed that SCI-1D11 mice had significantly (P < .05) attenuated loss of trabecular fractional bone volume (123% SCI-1D11 vs 69% SCI-IgG), thickness (98% vs 81%), and connectivity (112% vs 69%) and improved the structure model index (2.1 vs 2.7). Histomorphometry analysis revealed that osteoclast numbers were lower in the SCI-IgG mice than in sham-IgG control. Biochemically, SCI-IgG mice had higher levels of P1NP and PGE2 but similar TRAcP-5b and RANKL/OPG ratio to the sham-IgG group. The SCI-1D11 group exhibited higher levels of P1NP but similar TRAcP-5b, RANKL/OPG ratio, and PGE2 to the sham-1D11 group. Furthermore, 1D11 treatment prevented SCI-induced hyperphosphorylation of tau protein in osteocytes, an event that destabilizes the cytoskeleton. Together, inhibition of TGF-β signaling after SCI protects trabecular bone integrity, likely by balancing bone remodeling, inhibiting PGE2 elevation, and preserving the osteocyte cytoskeleton.
Collapse
Affiliation(s)
- Karim Sahbani
- National Center for the Medical Consequences of Spinal Cord Injury, James J Peters Veterans Affairs Medical Center, Bronx, NY 10468, USA
- Bronx Veterans Medical Research Foundation, Bronx, NY 10468, USA
| | - Christopher P Cardozo
- National Center for the Medical Consequences of Spinal Cord Injury, James J Peters Veterans Affairs Medical Center, Bronx, NY 10468, USA
- Bronx Veterans Medical Research Foundation, Bronx, NY 10468, USA
- Department of Medicine, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Rehabilitation Medicine and Human Performance, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mount Sinai Institute for Systems Biomedicine, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - William A Bauman
- National Center for the Medical Consequences of Spinal Cord Injury, James J Peters Veterans Affairs Medical Center, Bronx, NY 10468, USA
- Bronx Veterans Medical Research Foundation, Bronx, NY 10468, USA
- Department of Medicine, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Rehabilitation Medicine and Human Performance, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mount Sinai Institute for Systems Biomedicine, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Hesham A Tawfeek
- National Center for the Medical Consequences of Spinal Cord Injury, James J Peters Veterans Affairs Medical Center, Bronx, NY 10468, USA
- Bronx Veterans Medical Research Foundation, Bronx, NY 10468, USA
- Department of Medicine, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
9
|
Cao LL, Guan PP, Zhang SQ, Yang Y, Huang XS, Wang P. Downregulating expression of OPTN elevates neuroinflammation via AIM2 inflammasome- and RIPK1-activating mechanisms in APP/PS1 transgenic mice. J Neuroinflammation 2021; 18:281. [PMID: 34861878 PMCID: PMC8641240 DOI: 10.1186/s12974-021-02327-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 11/17/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Neuroinflammation is thought to be a cause of Alzheimer's disease (AD), which is partly caused by inadequate mitophagy. As a receptor of mitophagy, we aimed to reveal the regulatory roles of optineurin (OPTN) on neuroinflammation in the pathogenesis of AD. METHODS BV2 cells and APP/PS1 transgenic (Tg) mice were used as in vitro and in vivo experimental models to determine the regulatory roles of OPTN in neuroinflammation of AD. Sophisticated molecular technologies including quantitative (q) RT-PCR, western blot, enzyme linked immunosorbent assay (ELISA), co-immunoprecipitation (Co-IP) and immunofluorescence (IF) were employed to reveal the inherent mechanisms. RESULTS As a consequence, key roles of OPTN in regulating neuroinflammation were identified by depressing the activity of absent in melanoma 2 (AIM2) inflammasomes and receptor interacting serine/threonine kinase 1 (RIPK1)-mediated NF-κB inflammatory mechanisms. In detail, we found that expression of OPTN was downregulated, which resulted in activation of AIM2 inflammasomes due to a deficiency in mitophagy in APP/PS1 Tg mice. By ectopic expression, OPTN blocks the effects of Aβ oligomer (Aβo) on activating AIM2 inflammasomes by inhibiting mRNA expression of AIM2 and apoptosis-associated speck-like protein containing a C-terminal caspase recruitment domain (ASC), leading to a reduction in the active form of caspase-1 and interleukin (IL)-1β in microglial cells. Moreover, RIPK1 was also found to be negatively regulated by OPTN via ubiquitin protease hydrolysis, resulting in the synthesis of IL-1β by activating the transcriptional activity of NF-κB in BV2 cells. As an E3 ligase, the UBAN domain of OPTN binds to the death domain (DD) of RIPK1 to facilitate its ubiquitination. Based on these observations, ectopically expressed OPTN in APP/PS1 Tg mice deactivated microglial cells and astrocytes via the AIM2 inflammasome and RIPK-dependent NF-κB pathways, leading to reduce neuroinflammation. CONCLUSIONS These results suggest that OPTN can alleviate neuroinflammation through AIM2 and RIPK1 pathways, suggesting that OPTN deficiency may be a potential factor leading to the occurrence of AD.
Collapse
Affiliation(s)
- Long-Long Cao
- College of Life and Health Sciences, Northeastern University, No. 3-11. Wenhua Road, Shenyang, 110819, People's Republic of China
| | - Pei-Pei Guan
- College of Life and Health Sciences, Northeastern University, No. 3-11. Wenhua Road, Shenyang, 110819, People's Republic of China
| | - Shen-Qing Zhang
- College of Life and Health Sciences, Northeastern University, No. 3-11. Wenhua Road, Shenyang, 110819, People's Republic of China
| | - Yi Yang
- College of Life and Health Sciences, Northeastern University, No. 3-11. Wenhua Road, Shenyang, 110819, People's Republic of China
| | - Xue-Shi Huang
- College of Life and Health Sciences, Northeastern University, No. 3-11. Wenhua Road, Shenyang, 110819, People's Republic of China.
| | - Pu Wang
- College of Life and Health Sciences, Northeastern University, No. 3-11. Wenhua Road, Shenyang, 110819, People's Republic of China.
| |
Collapse
|
10
|
Elevating the Levels of Calcium Ions Exacerbate Alzheimer's Disease via Inducing the Production and Aggregation of β-Amyloid Protein and Phosphorylated Tau. Int J Mol Sci 2021; 22:ijms22115900. [PMID: 34072743 PMCID: PMC8198078 DOI: 10.3390/ijms22115900] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/08/2021] [Accepted: 05/08/2021] [Indexed: 01/03/2023] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease with a high incidence rate. The main pathological features of AD are β-amyloid plaques (APs), which are formed by β-amyloid protein (Aβ) deposition, and neurofibrillary tangles (NFTs), which are formed by the excessive phosphorylation of the tau protein. Although a series of studies have shown that the accumulation of metal ions, including calcium ions (Ca2+), can promote the formation of APs and NFTs, there is no systematic review of the mechanisms by which Ca2+ affects the development and progression of AD. In view of this, the current review summarizes the mechanisms by which Ca2+ is transported into and out of cells and organelles, such as the cell, endoplasmic reticulum, mitochondrial and lysosomal membranes to affect the balance of intracellular Ca2+ levels. In addition, dyshomeostasis of Ca2+ plays an important role in modulating the pathogenesis of AD by influencing the production and aggregation of Aβ peptides and tau protein phosphorylation and the ways that disrupting the metabolic balance of Ca2+ can affect the learning ability and memory of people with AD. In addition, the effects of these mechanisms on the synaptic plasticity are also discussed. Finally, the molecular network through which Ca2+ regulates the pathogenesis of AD is introduced, providing a theoretical basis for improving the clinical treatment of AD.
Collapse
|
11
|
Ye T, Gao HW, Xuan WT, Ye S, Zhou P, Li XQ, Wang Y, Song H, Liu YY, Cai B. The Regulating Mechanism of Chrysophanol on Protein Level of CaM-CaMKIV to Protect PC12 Cells Against Aβ 25-35-Induced Damage. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:2715-2723. [PMID: 32764873 PMCID: PMC7381772 DOI: 10.2147/dddt.s245128] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 06/30/2020] [Indexed: 12/14/2022]
Abstract
Objective To investigate the neuroprotective effect of chrysophanol (CHR) on PC12 treated with Aβ25-35, and the involved mechanism. Methods After the establishment of an AD cell model induced by Aβ25-35, the cell survival rate was detected by MTT, cell apoptosis was assayed by Hoechst 33342 staining, mRNA expressions of calmodulin (CaM), calcium/calmodulin-dependent protein kinase kinase (CaMKK), calcium/calmodulin-dependent protein kinase IV (CaMKIV) and tau (MAPT; commonly known as tau) were determined by qRT-PCR, and protein levels of CaM, CaMKK, CaMKIV, phospho-CaMKIV (p-CaMKIV), tau and phospho-tau (p-tau) were detected by Western blot analysis. Results When pretreated with CHR before exposure to Aβ25-35, PC12 cells showed that increased cell viability and reduced apoptosis. The qRT-PCR results indicated that the deposition of Aβ25-35 triggers a decrease in levels of CaM, CaMKK, CaMKIV, and tau in PC12 cells. In addition, Western blot results also suggested that Aβ25-35 decreases the protein expression of CaM, CaMKK, CaMKIV, p-CaMKIV, and the ratio of p-tau to tau in PC12 cells. However, the above effects were significantly alleviated after the treatment of CHR. Conclusion CHR plays a neuroprotective role in AD though decreasing the protein level of CaM-CaMKK-CaMKIV and the expression of p-tau downstream.
Collapse
Affiliation(s)
- Ting Ye
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, People's Republic of China
| | - Hua-Wu Gao
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, People's Republic of China
| | - Wei-Ting Xuan
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, People's Republic of China
| | - Shu Ye
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, People's Republic of China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei 230012, People's Republic of China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, People's Republic of China
| | - Peng Zhou
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, People's Republic of China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei 230012, People's Republic of China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, People's Republic of China
| | - Xin-Quan Li
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, People's Republic of China
| | - Yan Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, People's Republic of China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei 230012, People's Republic of China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, People's Republic of China
| | - Hang Song
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, People's Republic of China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei 230012, People's Republic of China
| | - Yan-Yan Liu
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, People's Republic of China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei 230012, People's Republic of China
| | - Biao Cai
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, People's Republic of China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei 230012, People's Republic of China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, People's Republic of China
| |
Collapse
|
12
|
Dioguardi M, Crincoli V, Laino L, Alovisi M, Sovereto D, Mastrangelo F, Lo Russo L, Lo Muzio L. The Role of Periodontitis and Periodontal Bacteria in the Onset and Progression of Alzheimer's Disease: A Systematic Review. J Clin Med 2020; 9:E495. [PMID: 32054121 PMCID: PMC7074205 DOI: 10.3390/jcm9020495] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/28/2020] [Accepted: 02/09/2020] [Indexed: 12/28/2022] Open
Abstract
The evidence of a connection between the peripheral inflammatory processes and neurodegenerative diseases of the central nervous system is becoming more apparent. This review of the related literature highlights the most recent clinical, epidemiological, and in vitro studies trying to investigate possible connections between periodontal bacteria and the onset and progression of Alzheimer's disease. This review was conducted by searching databases such as PubMed and Scopus using keywords or combinations such as Alzheimer's Disease AND periodontal or dementia AND periodontitis OR periodontal. After eliminating overlaps and screening the articles not related to these issues, we identified 1088 records and proceeded to the selection of articles for an evaluation of the associative assumptions. The hypothesis suggested by the authors and confirmed by the literature is that the bacterial load and the inflammatory process linked to periodontal disease can intensify inflammation at the level of the central nervous system, favoring the occurrence of the disease. The analysis of the literature highlights how periodontal disease can directly contribute to the peripheral inflammatory environment by the introduction of periodontal or indirect pathogenic bacteria and proinflammatory cytokines locally produced at the periodontal level following bacterial colonization of periodontal defects.
Collapse
Affiliation(s)
- Mario Dioguardi
- Department of Clinical and Experimental Medicine, University of Foggia, Via Rovelli 50, 71122 Foggia, Italy; (D.S.); (F.M.); (L.L.R.)
| | - Vito Crincoli
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Division of Complex Operating Unit of Dentistry, “Aldo Moro” University of Bari, Piazza G. Cesare 11, 70124 Bari, Italy;
| | - Luigi Laino
- Multidisciplinary Department of Medical-Surgical and Odontostomatological Specialties, University of Campania “Luigi Vanvitelli”, 80121 Naples, Italy;
| | - Mario Alovisi
- Department of Surgical Sciences, Dental School, University of Turin, 10126 Turin, Italy
| | - Diego Sovereto
- Department of Clinical and Experimental Medicine, University of Foggia, Via Rovelli 50, 71122 Foggia, Italy; (D.S.); (F.M.); (L.L.R.)
| | - Filiberto Mastrangelo
- Department of Clinical and Experimental Medicine, University of Foggia, Via Rovelli 50, 71122 Foggia, Italy; (D.S.); (F.M.); (L.L.R.)
| | - Lucio Lo Russo
- Department of Clinical and Experimental Medicine, University of Foggia, Via Rovelli 50, 71122 Foggia, Italy; (D.S.); (F.M.); (L.L.R.)
| | - Lorenzo Lo Muzio
- Department of Clinical and Experimental Medicine, University of Foggia, Via Rovelli 50, 71122 Foggia, Italy; (D.S.); (F.M.); (L.L.R.)
| |
Collapse
|
13
|
Hu S, Hu M, Liu J, Zhang B, Zhang Z, Zhou FH, Wang L, Dong J. Phosphorylation of Tau and α-Synuclein Induced Neurodegeneration in MPTP Mouse Model of Parkinson's Disease. Neuropsychiatr Dis Treat 2020; 16:651-663. [PMID: 32184604 PMCID: PMC7061418 DOI: 10.2147/ndt.s235562] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 02/09/2020] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Parkinson's disease (PD) is the second most common neurodegenerative disease. The α-Synuclein is a major component of Lewy bodies and Lewy neurites, the pathologic hallmark of PD. It is known that α-Synuclein is phosphorylated (p-α-Synuclein) in PD and tau-hyperphosphorylation (p-Tau) is also a pathologic feature of PD. However, the relationship between p-Synuclein and p-Tau in PD is not clear, in particular in the MPTP model of PD. The purpose of this study was to reveal their relationship in the mouse MPTP model. METHODS Firstly, the p-α-Synuclein, α-Synuclein, p-Tau and Tau protein levels were analyzed. Then, GSK3β activation was determined using immunoblot and immunohistochemical staining. Finally, the dopaminergic neurodegeneration was assessed using Tyrosine Hydroxylase (TH) staining and retrograde labeling and microglial marker were labeled. Microglial activation and nigrostriatal pathway degeneration were observed. RESULTS The results showed that p-α-Synuclein, α-Synuclein, p-Tau and Tau were upregulated in both hippocampus and substantia nigra of the PD mouse model. Furthermore, p-α-Synuclein and p-Tau were localized in the same regions of substantial nigra (SN) and dentate gyrus (DG) of hippocampus (Hippo). The activated form of GSK3β (phosphor GSK3β Y216) was increased in multiple brain areas. The GSK3β inhibitor AZD1080 injected in MPTP mice suppressed the expression of p-Tau and p-GSK3β and improved motor functions. CONCLUSION These findings revealed that p-α-Synuclein and p-Tau proteins are key pathological events leading to neurodegeneration and motor dysfunctions in the mouse MPTP model of PD. Our data suggest that the interference with the GSK3β activity may be an effective approach for the treatment of PD.
Collapse
Affiliation(s)
- Shanshan Hu
- Good Clinical Practice Center, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, Guizhou, People's Republic of China
| | - Meigui Hu
- The Second School of Clinical Medicine, Zhuhai Campus of Zunyi Medical University, Zhuhai 519041, Guangdong, People's Republic of China
| | - Jian Liu
- Department of Anatomy, Zunyi Medical University, Zunyi 563000, Guizhou, People's Republic of China
| | - Bei Zhang
- Department of Stomatology, The First People's Hospital of Zunyi, Zunyi 563099, Guizhou, People's Republic of China
| | - Zhen Zhang
- Department of Hand Surgery, Department of Plastic Reconstructive Surgery, Ningbo No. 6 Hospital, Ningbo 315040, People's Republic of China
| | - Fiona H Zhou
- School of Pharmacy and Medical Sciences, and UniSA Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia
| | - Liping Wang
- Department of Hand Surgery, Department of Plastic Reconstructive Surgery, Ningbo No. 6 Hospital, Ningbo 315040, People's Republic of China.,School of Pharmacy and Medical Sciences, and UniSA Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia
| | - Jianghui Dong
- Department of Hand Surgery, Department of Plastic Reconstructive Surgery, Ningbo No. 6 Hospital, Ningbo 315040, People's Republic of China.,School of Pharmacy and Medical Sciences, and UniSA Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia
| |
Collapse
|
14
|
Cyclooxygenase-2 is critical for the propagation of β-amyloid protein and reducing the glycosylation of tau in Alzheimer's disease. Cell Mol Immunol 2019; 16:892-894. [PMID: 31551514 DOI: 10.1038/s41423-019-0294-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 08/29/2019] [Indexed: 12/24/2022] Open
|
15
|
Biringer RG. The Role of Eicosanoids in Alzheimer's Disease. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16142560. [PMID: 31323750 PMCID: PMC6678666 DOI: 10.3390/ijerph16142560] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/09/2019] [Accepted: 07/13/2019] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative disorders known. Estimates from the Alzheimer's Association suggest that there are currently 5.8 million Americans living with the disease and that this will rise to 14 million by 2050. Research over the decades has revealed that AD pathology is complex and involves a number of cellular processes. In addition to the well-studied amyloid-β and tau pathology, oxidative damage to lipids and inflammation are also intimately involved. One aspect all these processes share is eicosanoid signaling. Eicosanoids are derived from polyunsaturated fatty acids by enzymatic or non-enzymatic means and serve as short-lived autocrine or paracrine agents. Some of these eicosanoids serve to exacerbate AD pathology while others serve to remediate AD pathology. A thorough understanding of eicosanoid signaling is paramount for understanding the underlying mechanisms and developing potential treatments for AD. In this review, eicosanoid metabolism is examined in terms of in vivo production, sites of production, receptor signaling, non-AD biological functions, and known participation in AD pathology.
Collapse
Affiliation(s)
- Roger G Biringer
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, 5000 Lakewood Ranch Blvd., Bradenton, FL 34211, USA.
| |
Collapse
|