1
|
Lindberg M, Hu J, Sparr E, Linse S. Reduced protein solubility - cause or consequence in amyloid disease? QRB DISCOVERY 2025; 6:e8. [PMID: 40070848 PMCID: PMC11894405 DOI: 10.1017/qrd.2024.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 03/14/2025] Open
Abstract
In this perspective, we ask the question whether the apparently lower solubility of specific proteins in amyloid disease is a cause or consequence of the protein deposition seen in such diseases. We focus on Alzheimer's disease and start by reviewing the experimental evidence of disease-associated reduction in the measured concentration of amyloid β peptide, Aβ42, in cerebrospinal fluid. We propose a series of possible physicochemical explanations for these observations. These include a reduced solubility, a reduced apparent solubility, as well as a long-lived metastable state manifested in healthy individuals as a free concentration of Aβ42 in the solution phase above the solubility limit. For each scenario, we discuss whether it is most likely a cause or a consequence of the observed protein deposition in the disease.
Collapse
Affiliation(s)
- Max Lindberg
- Biochemistry and Structural Biology, Lund University, Lund, Sweden
| | - Jing Hu
- Division for Physical Chemistry, Lund University, Lund, Sweden
| | - Emma Sparr
- Division for Physical Chemistry, Lund University, Lund, Sweden
| | - Sara Linse
- Biochemistry and Structural Biology, Lund University, Lund, Sweden
| |
Collapse
|
2
|
Jang YJ, Kang SJ, Park HS, Lee DH, Kim JH, Kim JE, Kim DI, Chung CH, Yoon JK, Bhang SH. Drug delivery strategies with lipid-based nanoparticles for Alzheimer's disease treatment. J Nanobiotechnology 2025; 23:99. [PMID: 39930497 PMCID: PMC11809104 DOI: 10.1186/s12951-025-03109-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 01/11/2025] [Indexed: 02/14/2025] Open
Abstract
Alzheimer's disease (AD) is a distinctive form of dementia characterized by age-related cognitive decline and memory impairment. A key hallmark of AD is the irreversible overaccumulation of beta-amyloid (Aβ) in the brain, associated with neuroinflammation and neuronal death. Although Aβ clearance and immunoregulation have been the major therapeutic strategies for AD, highly selective transport across the blood-brain barrier (BBB) negatively affects the delivery efficacy of the drugs without the ability to cross the BBB. In this review, we discuss the potential of lipid-based nanoparticles (LBNs) as promising vehicles for drug delivery in AD treatment. LBNs, composed of phospholipid mono- or bilayer, have attracted attention due to their exceptional cellular penetration capabilities and drug loading capabilities, which also facilitate cargo transcytosis across the BBB. Recent advances in the development and engineering of LBNs overcome the existing limitations of the current clinical approaches for AD treatment by addressing off-target effects and low therapeutic efficacy. Here, we review the transport pathways across the BBB, as well as various types of LBNs for AD therapy, including exosomes, liposomes, solid lipid nanoparticles (SLNs), and nanostructured lipid carriers (NLCs), to elucidate their distinctive properties, preparation methodologies, and therapeutic efficacy, thereby offering innovative avenues for novel drug development for clinical translation in AD therapy.
Collapse
Affiliation(s)
- Young-Ju Jang
- School of Chemical Engineering, Sungkyunkwan University, Suwon-si, 16419, Gyeonggi-do, Republic of Korea
| | - Seong-Jun Kang
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si, 17546, Gyeonggi-do, Republic of Korea
| | - Hyun Su Park
- School of Chemical Engineering, Sungkyunkwan University, Suwon-si, 16419, Gyeonggi-do, Republic of Korea
| | - Dong-Hyun Lee
- School of Chemical Engineering, Sungkyunkwan University, Suwon-si, 16419, Gyeonggi-do, Republic of Korea
| | - Jae Hoon Kim
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si, 17546, Gyeonggi-do, Republic of Korea
| | - Ju-El Kim
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si, 17546, Gyeonggi-do, Republic of Korea
| | - Dong-Ik Kim
- Division of Vascular Surgery, Samsung Medical Center, School of Medicine, Sungkyunkwan University, Seoul, 06351, Republic of Korea
| | - Chan-Hwa Chung
- School of Chemical Engineering, Sungkyunkwan University, Suwon-si, 16419, Gyeonggi-do, Republic of Korea
| | - Jeong-Kee Yoon
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si, 17546, Gyeonggi-do, Republic of Korea.
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon-si, 16419, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
3
|
Engelhardt E, Resende EDPF, Gomes KB. Physiopathological mechanisms underlying Alzheimer's disease: a narrative review. Dement Neuropsychol 2024; 18:e2024VR01. [PMID: 39697643 PMCID: PMC11654088 DOI: 10.1590/1980-5764-dn-2024-vr01] [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: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 12/20/2024] Open
Abstract
The neuropathological signature of Alzheimer's disease (AD) comprises mainly amyloid plaques, and neurofibrillary tangles, resulting in synaptic and neuronal loss. These pathological structures stem from amyloid dysfunctional metabolism according to the amyloid cascade hypothesis, leading to the formation of plaques, and apparently inducing the initiation of the abnormal tau pathway, with phosphorylation and aggregation of these proteins, ultimately causing the formation of tangles. In this narrative review, the existing hypothesis related to the pathophysiology of AD were compiled, and biological pathways were highlighted in order to identify the molecules that could represent biological markers of the disease, necessary to establish early diagnosis, as well as the selection of patients for therapeutical interventional strategies.
Collapse
Affiliation(s)
- Eliasz Engelhardt
- Universidade Federal do Rio de Janeiro, Instituto de Neurologia Deolindo Couto, Rio de Janeiro RJ, Brazil
| | - Elisa de Paula França Resende
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Belo Horizonte MG, Brazil
- Faculdade Ciências Médicas de Minas Gerais, Belo Horizonte MG, Brazil
| | - Karina Braga Gomes
- Universidade Federal de Minas Gerais, Faculdade de Farmácia, Belo Horizonte MG, Brazil
| |
Collapse
|
4
|
Choudhary G, Prajapat M, Kaur G, Singh H, Mahendiratta S, Prakash A, Medhi B. Integrated in-silico and in-vitro assessments of HDAC6 inhibitor efficacy in mitigating amyloid beta pathology in Alzheimer's disease. J Biomol Struct Dyn 2024; 42:9720-9730. [PMID: 37878051 DOI: 10.1080/07391102.2023.2274518] [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: 02/22/2023] [Accepted: 08/21/2023] [Indexed: 10/26/2023]
Abstract
Alzheimer's disease, marked by memory loss and cognitive decline, is associated with amyloid-beta (Aβ) peptide accumulation in the brain. The enzyme neprilysin (NEP), crucial for Aβ degradation, decreases with age and in sporadic Alzheimer's disease, leading to increased Aβ build-up. This study hypothesized the targeting of enzyme HDAC6, believed to influence NEP activity. An in-silico study was conducted using an FDA-approved drug database, with the focus on their interaction with the HDAC6 structure. Among tested ligands, Panobinostat showed the most favourable interaction with HDAC6. In-vitro experiments on the SH-SY5Y neuronal cell line confirmed these findings, with Panobinostat inhibiting HDAC6, enhancing NEP levels, and reducing Aβ load. The study suggests Panobinostat as a potential Alzheimer's therapeutic agent, mitigating Aβ accumulation via NEP upregulation. Further research is required for comprehensive understanding and validation.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | | | - Gurjeet Kaur
- Department of Pharmacology, PGIMER, Chandigarh, India
| | | | | | - Ajay Prakash
- Department of Pharmacology, PGIMER, Chandigarh, India
| | - Bikash Medhi
- Department of Pharmacology, PGIMER, Chandigarh, India
| |
Collapse
|
5
|
Chiarini A, Armato U, Gui L, Yin M, Chang S, Dal Prà I. Early divergent modulation of NLRP2's and NLRP3's inflammasome sensors vs. AIM2's one by signals from Aβ·Calcium-sensing receptor complexes in human astrocytes. Brain Res 2024; 1846:149283. [PMID: 39426463 DOI: 10.1016/j.brainres.2024.149283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 10/04/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024]
Abstract
Alzheimer's disease (AD), the most prevalent human dementia, is driven by accruals of extracellular Aβ42 senile patches and intracellular neurofibrillary tangles of hyperphosphorylated Tau (p-Tau) proteins. AD's concurrent neuroinflammation is prompted by innate immunity-related cytosolic protein oligomers named inflammasomes. Upon proper "first" (priming) and "second" (activating) signals, inflammasomes overproduce proinflammatory Interleukin (IL)-1β, and IL-18 while cleaving pyroptosis-promoting Gasdermin D's N-terminal fragments. Our earlier studies highlighted that in pure monocultures, exogenous Aβ25-35-treated nonproliferating human cortical astrocytes (HCAs) made and released surpluses of endogenous Aβ42-oligomers (-os) and p-Tau-os, just as alike-treated human cortical neurons did. Aβ25-35-exposed HCAs also over-released NO, VEGFA, and IL-6. Aβ•CaSR (Aβ·Calcium-Sensing Receptor) complexes generated intracellular signals mediating all such neurotoxic effects since CaSR's negative allosteric modulators (aka NAMs or calcilytics, e.g., NPS2143) fully suppressed them. However, it had hitherto remained unexplored whether signals from Aβ·CaSR complexes also induced the early expression and/or activation of NOD-like 2 (NLRP2) and 3 (NLRP3) and of PYHIN absent in melanoma 2 (AIM2) inflammasomes in monocultured HCAs. To clarify this topic, we used in-situ-Proximity Ligation, qRT-PCR, double antibody arrays, immunoblots, and Caspase 1/4 enzymatic assays. Aβ·CaSR complexes quickly assembled on HCAs surface and issued intracellular signals activating Akt and JAK/STAT axes. In turn, the latter upregulated NLRP2 and NLRP3 PRRs (pattern recognition receptors) yet downregulated AIM2. These effects were specific, being significantly hindered by NPS2143 and inhibitors of PI3K (LY294002), AMPKα (Dorsomorphin), mTOR (Torin1), and JAK/TYK (Brepoticinib). A wide-spectrum inhibitor, Bay11-7082, intensified the Aβ·CaSR/Akt/JAK/STAT axis-driven opposite control of NLRP3's and AIM2's PRR proteins without affecting NLRP2 PRR upregulation. However, the said effects on the PRRs proteins vanished within 24-h. Moreover, Aβ·CaSR signals neither concurrently changed ASC, pro-IL-1β, and Gasdermin-D (holo- and fragments) protein levels and Caspases 1 and 4 enzymatic activities nor induced pyroptosis. Therefore, Aβ·CaSR cues acted as "first (priming) signals" temporarily increasing NLRP2 and NLRP3 PRRs expression without activating the corresponding inflammasomes. The neatly divergent modulation of NLRP3's vs. AIM2's PRR proteins by Aβ·CaSR cues and by Bay11-7082 suggests that, when bacterial or viral DNA fragments are absent, AIM2 might play "anti-inflammasomal" or other roles in HCAs. However, Bay11-7082's no effect on NLRP2 PRR overexpression also reveals that CaSR's downstream mechanisms controlling inflammasomes' sensors are quite complex in HCAs, and hence, given AD's impact on human health, well worth further studies.
Collapse
Affiliation(s)
- Anna Chiarini
- Department of Surgery, Dentistry, Pediatrics, and Gynecology, University of Verona, 8 Strada Le Grazie, 37134 Verona, Italy.
| | - Ubaldo Armato
- Department of Surgery, Dentistry, Pediatrics, and Gynecology, University of Verona, 8 Strada Le Grazie, 37134 Verona, Italy.
| | - Li Gui
- Department of Neurology, Southwest Hospital, Army Medical University, 29 Gaotanyan Street, Chongqing, 400038, China.
| | - Meifang Yin
- Department of Surgery, Dentistry, Pediatrics, and Gynecology, University of Verona, 8 Strada Le Grazie, 37134 Verona, Italy.
| | - Shusen Chang
- Department of Surgery, Dentistry, Pediatrics, and Gynecology, University of Verona, 8 Strada Le Grazie, 37134 Verona, Italy.
| | - Ilaria Dal Prà
- Department of Surgery, Dentistry, Pediatrics, and Gynecology, University of Verona, 8 Strada Le Grazie, 37134 Verona, Italy.
| |
Collapse
|
6
|
Sandoval KE, Witt KA. Somatostatin: Linking Cognition and Alzheimer Disease to Therapeutic Targeting. Pharmacol Rev 2024; 76:1291-1325. [PMID: 39013601 PMCID: PMC11549939 DOI: 10.1124/pharmrev.124.001117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/18/2024] Open
Abstract
Over 4 decades of research support the link between Alzheimer disease (AD) and somatostatin [somatotropin-releasing inhibitory factor (SRIF)]. SRIF and SRIF-expressing neurons play an essential role in brain function, modulating hippocampal activity and memory formation. Loss of SRIF and SRIF-expressing neurons in the brain rests at the center of a series of interdependent pathological events driven by amyloid-β peptide (Aβ), culminating in cognitive decline and dementia. The connection between the SRIF and AD further extends to the neuropsychiatric symptoms, seizure activity, and inflammation, whereas preclinical AD investigations show SRIF or SRIF receptor agonist administration capable of enhancing cognition. SRIF receptor subtype-4 activation in particular presents unique attributes, with the potential to mitigate learning and memory decline, reduce comorbid symptoms, and enhance enzymatic degradation of Aβ in the brain. Here, we review the links between SRIF and AD along with the therapeutic implications. SIGNIFICANCE STATEMENT: Somatostatin and somatostatin-expressing neurons in the brain are extensively involved in cognition. Loss of somatostatin and somatostatin-expressing neurons in Alzheimer disease rests at the center of a series of interdependent pathological events contributing to cognitive decline and dementia. Targeting somatostatin-mediated processes has significant therapeutic potential for the treatment of Alzheimer disease.
Collapse
Affiliation(s)
- Karin E Sandoval
- Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, Illinois
| | - Ken A Witt
- Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, Illinois
| |
Collapse
|
7
|
Azargoonjahromi A. Immunotherapy in Alzheimer's disease: focusing on the efficacy of gantenerumab on amyloid-β clearance and cognitive decline. J Pharm Pharmacol 2024; 76:1115-1131. [PMID: 38767981 DOI: 10.1093/jpp/rgae066] [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: 01/18/2024] [Accepted: 05/08/2024] [Indexed: 05/22/2024]
Abstract
Gantenerumab, a human monoclonal antibody (mAb), has been thought of as a potential agent to treat Alzheimer's disease (AD) by specifically targeting regions of the amyloid-β (Aβ) peptide sequence. Aβ protein accumulation in the brain leads to amyloid plaques, causing neuroinflammation, oxidative stress, neuronal damage, and neurotransmitter dysfunction, thereby causing cognitive decline in AD. Gantenerumab involves disrupting Aβ aggregation and promoting the breakdown of larger Aβ aggregates into smaller fragments, which facilitates the action of Aβ-degrading enzymes in the brain, thus slowing down the progression of AD. Moreover, Gantenerumab acts as an opsonin, coating Aβ plaques and enhancing their recognition by immune cells, which, combined with its ability to improve the activity of microglia, makes it an intriguing candidate for promoting Aβ plaque clearance. Indeed, the multifaceted effects of Gantenerumab, including Aβ disaggregation, enhanced immune recognition, and improved microglia activity, may position it as a promising therapeutic approach for AD. Of note, reports suggest that Gantenerumab, albeit its capacity to reduce or eliminate Aβ, has not demonstrated effectiveness in reducing cognitive decline. This review, after providing an overview of immunotherapy approaches that target Aβ in AD, explores the efficacy of Gantenerumab in reducing Aβ levels and cognitive decline.
Collapse
|
8
|
Watamura N, Kakiya N, Fujioka R, Kamano N, Takahashi M, Nilsson P, Saito T, Iwata N, Fujisawa S, Saido TC. The dopaminergic system promotes neprilysin-mediated degradation of amyloid-β in the brain. Sci Signal 2024; 17:eadk1822. [PMID: 39106321 DOI: 10.1126/scisignal.adk1822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 04/21/2024] [Accepted: 07/01/2024] [Indexed: 08/09/2024]
Abstract
Deposition of amyloid-β (Aβ) in the brain can impair neuronal function and contribute to cognitive decline in Alzheimer's disease (AD). Here, we found that dopamine and the dopamine precursor levodopa (also called l-DOPA) induced Aβ degradation in the brain. Chemogenetic approaches in mice revealed that the activation of dopamine release from ventral tegmental area (VTA) neurons increased the abundance and activity of the Aβ-degrading enzyme neprilysin and reduced the amount of Aβ deposits in the prefrontal cortex in a neprilysin-dependent manner. Aged mice had less dopamine and neprilysin in the anterior cortex, a decrease that was accentuated in AD model mice. Treating AD model mice with levodopa reduced Aβ deposition and improved cognitive function. These observations demonstrate that dopamine promotes brain region-specific, neprilysin-dependent degradation of Aβ, suggesting that dopamine-associated strategies have the potential to treat this aspect of AD pathology.
Collapse
Affiliation(s)
- Naoto Watamura
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Naomasa Kakiya
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Ryo Fujioka
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Naoko Kamano
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Mika Takahashi
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division for Neurogeriatrics, Karolinska Institutet, 171 64, Solna, Sweden
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi 464-8601, Japan
| | - Nobuhisa Iwata
- Department of Genome-based Drug Discovery & Leading Medical Research Core Unit, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8521, Japan
| | - Shigeyoshi Fujisawa
- Laboratory for Systems Neurophysiology, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| |
Collapse
|
9
|
Fotuhi SN, Khalaj-Kondori M. Imbalanced clearance of Aβ peptide cause presynaptic plaque formation. Int J Neurosci 2024; 134:66-70. [PMID: 35639020 DOI: 10.1080/00207454.2022.2085099] [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: 08/03/2020] [Accepted: 05/26/2022] [Indexed: 10/18/2022]
Abstract
Alzheimer's disease is characterized by abnormal increase of Aβ peptide which is likely as the result of imbalanced homeostasis of its production and clearance mechanisms. Here, we briefly review that the uncleaned extracellular Aβ peptides are loaded into presynaptic neurons. The Aβ oligomers desperately affect pre- and post-synapse neuron activity and turn into plaques inside the presynaptic neurons over the time passes.
Collapse
Affiliation(s)
- Seyedeh Nahid Fotuhi
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Mohammad Khalaj-Kondori
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| |
Collapse
|
10
|
Almeida VN. Somatostatin and the pathophysiology of Alzheimer's disease. Ageing Res Rev 2024; 96:102270. [PMID: 38484981 DOI: 10.1016/j.arr.2024.102270] [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: 07/18/2023] [Revised: 03/09/2024] [Accepted: 03/09/2024] [Indexed: 03/28/2024]
Abstract
Among the central features of Alzheimer's disease (AD) progression are altered levels of the neuropeptide somatostatin (SST), and the colocalisation of SST-positive interneurons (SST-INs) with amyloid-β plaques, leading to cell death. In this theoretical review, I propose a molecular model for the pathogenesis of AD based on SST-IN hypofunction and hyperactivity. Namely, hypofunctional and hyperactive SST-INs struggle to control hyperactivity in medial regions in early stages, leading to axonal Aβ production through excessive presynaptic GABAB inhibition, GABAB1a/APP complex downregulation and internalisation. Concomitantly, excessive SST-14 release accumulates near SST-INs in the form of amyloids, which bind to Aβ to form toxic mixed oligomers. This leads to differential SST-IN death through excitotoxicity, further disinhibition, SST deficits, and increased Aβ release, fibrillation and plaque formation. Aβ plaques, hyperactive networks and SST-IN distributions thereby tightly overlap in the brain. Conversely, chronic stimulation of postsynaptic SST2/4 on gulutamatergic neurons by hyperactive SST-INs promotes intense Mitogen-Activated Protein Kinase (MAPK) p38 activity, leading to somatodendritic p-tau staining and apoptosis/neurodegeneration - in agreement with a near complete overlap between p38 and neurofibrillary tangles. This model is suitable to explain some of the principal risk factors and markers of AD progression, including mitochondrial dysfunction, APOE4 genotype, sex-dependent vulnerability, overactive glial cells, dystrophic neurites, synaptic/spine losses, inter alia. Finally, the model can also shed light on qualitative aspects of AD neuropsychology, especially within the domains of spatial and declarative (episodic, semantic) memory, under an overlying pattern of contextual indiscrimination, ensemble instability, interference and generalisation.
Collapse
Affiliation(s)
- Victor N Almeida
- Institute of Psychiatry, Faculty of Medicine, University of São Paulo (USP), Brazil; Faculty of Languages, Federal University of Minas Gerais (UFMG), Brazil.
| |
Collapse
|
11
|
Pang KS, Peng HB, Li BP, Wen B, Noh K, Xia R, Toscan A, Serson S, Fraser PE, Tirona RG, de Lannoy IAM. Aging and brain free cholesterol concentration on amyloid-β peptide accumulation in guinea pigs. Biopharm Drug Dispos 2024; 45:93-106. [PMID: 38488691 DOI: 10.1002/bdd.2386] [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: 04/22/2023] [Revised: 01/21/2024] [Accepted: 02/08/2024] [Indexed: 04/19/2024]
Abstract
Alzheimer's disease is a complex multifactorial neurodegenerative disorder wherein age is a major risk factor. The appropriateness of the Hartley guinea pig (GP), which displays high sequence homologies of its amyloid-β (Aβ40 and Aβ42) peptides, Mdr1 and APP (amyloid precursor protein) and similarity in lipid handling to humans, was appraised among 9-40 weeks old guinea pigs. Protein expression levels of P-gp (Abcb1) and Cyp46a1 (24(S)-hydroxylase) for Aβ40, and Aβ42 efflux and cholesterol metabolism, respectively, were decreased with age, whereas those for Lrp1 (low-density lipoprotein receptor related protein 1), Rage (receptor for advanced glycation endproducts) for Aβ efflux and influx, respectively, and Abca1 (the ATP binding cassette subfamily A member 1) for cholesterol efflux, were unchanged among the ages examined. There was a strong, negative correlation of the brain Aβ peptide concentrations and Abca1 protein expression levels with free cholesterol. The correlation of Aβ peptide concentrations with Cyp46a1 was, however, not significant, and concentrations of the 24(S)-hydroxycholesterol metabolite revealed a decreasing trend from 20 weeks old toward 40 weeks old guinea pigs. The composite data suggest a role for free cholesterol on brain Aβ accumulation. The decreases in P-gp and Lrp1 protein levels should further exacerbate the accumulation of Aβ peptides in guinea pig brain.
Collapse
Affiliation(s)
- K Sandy Pang
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - H Benson Peng
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Betty P Li
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Binyu Wen
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Keumhan Noh
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Runyu Xia
- Department of Pharmacology and Toxicology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Anja Toscan
- Transpharmation Canada, Mississauga, Ontario, Canada
| | - Sylvia Serson
- Transpharmation Canada, Mississauga, Ontario, Canada
| | - Paul E Fraser
- Tanz Centre for Research in Neurodegenerative Diseases and Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Rommel G Tirona
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | | |
Collapse
|
12
|
Alruwad MI, Salah El Dine R, Gendy AM, Sabry MM, El Hefnawy HM. Exploring the Biological and Phytochemical Potential of Jordan's Flora: A Review and Update of Eight Selected Genera from Mediterranean Region. Molecules 2024; 29:1160. [PMID: 38474670 DOI: 10.3390/molecules29051160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Jordan's flora is known for its rich diversity, with a grand sum of 2978 plant species that span 142 families and 868 genera across four different zones. Eight genera belonging to four different plant families have been recognized for their potential natural medicinal properties within the Mediterranean region. These genera include Chrysanthemum L., Onopordum Vaill. Ex. L., Phagnalon Cass., and Senecio L. from the Asteraceae family, in addition to Clematis L. and Ranunculus L. from the Ranunculaceae family, Anchusa L. from the Boraginaceae family, and Eryngium L. from the Apiaceae family. The selected genera show a wide variety of secondary metabolites with encouraging pharmacological characteristics including antioxidant, antibacterial, cytotoxic, anti-inflammatory, antidiabetic, anti-ulcer, and neuroprotective actions. Further research on these genera and their extracts will potentially result in the formulation of novel and potent natural pharmaceuticals. Overall, Jordan's rich flora provides a valuable resource for exploring and discovering new plant-based medicines.
Collapse
Affiliation(s)
- Manal I Alruwad
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt
| | - Riham Salah El Dine
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt
| | - Abdallah M Gendy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October 6 University, Giza 12585, Egypt
| | - Manal M Sabry
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt
| | - Hala M El Hefnawy
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt
| |
Collapse
|
13
|
Oliveira FFD, Almeida SSD, Chen ES, Smith MC, Bertolucci PHF. Pharmacogenetics of angiotensin modulators according to APOE-ϵ4 alleles and the ACE insertion/deletion polymorphism in Alzheimer's disease. Acta Neuropsychiatr 2023; 35:346-361. [PMID: 37605989 DOI: 10.1017/neu.2023.38] [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] [Indexed: 08/23/2023]
Abstract
OBJECTIVE In Alzheimer's disease (AD), angiotensin II receptor blockers (ARBs) could reduce cerebrovascular dysfunction, while angiotensin-converting enzyme inhibitors (ACEis) might increase brain amyloid-β by suppressing effects of the angiotensin-converting enzyme 1, an amyloid-β-degrading enzyme. However, ACEis could benefit patients with AD by reducing the amyloidogenic processing of the amyloid precursor protein, by central cholinergic and anti-inflammatory mechanisms, and by peripheral modulation of glucose homeostasis. We aimed to investigate whether the ACE insertion/deletion polymorphism is associated with clinical changes in patients with AD, while considering apolipoprotein E (APOE)-ϵ4 carrier status and blood pressure response to angiotensin modulators. METHODS Consecutive outpatients with late-onset AD were screened with cognitive tests and anthropometric measurements, while their caregivers were queried for functional and caregiver burden scores. Prospective pharmacogenetic associations were estimated for 1 year, taking APOE-ϵ4 carrier status and genotypes of the ACE insertion/deletion polymorphism into account, along with treatment with ACEis or ARBs. RESULTS For 193 patients (67.4% women, 53.4% APOE-ϵ4 carriers), the ACE insertion/deletion polymorphism was in Hardy-Weinberg equilibrium (p = 0.281), while arterial hypertension was prevalent in 80.3% (n = 124 used an ACEi, n = 21 used an ARB). ARBs benefitted mostly APOE-ϵ4 carriers concerning caregiver burden variations, cognitive and functional decline. ACEis benefitted APOE-ϵ4 non-carriers concerning cognitive and functional decline due to improved blood pressure control in addition to possible central mechanisms. The ACE insertion/deletion polymorphism led to variable response to angiotensin modulators concerning neurological outcomes and blood pressure variations. CONCLUSION Angiotensin modulators may be disease-modifiers in AD, while genetic stratification of samples is recommended in clinical studies.
Collapse
Affiliation(s)
- Fabricio Ferreira de Oliveira
- Department of Neurology and Neurosurgery, Escola Paulista de Medicina, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Sandro Soares de Almeida
- Department of Biophysics, Escola Paulista de Medicina, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Elizabeth Suchi Chen
- Department of Morphology and Genetics, Escola Paulista de Medicina, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Marilia Cardoso Smith
- Department of Morphology and Genetics, Escola Paulista de Medicina, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | | |
Collapse
|
14
|
Amano A, Sanjo N, Araki W, Anraku Y, Nakakido M, Matsubara E, Tomiyama T, Nagata T, Tsumoto K, Kataoka K, Yokota T. Peripheral administration of nanomicelle-encapsulated anti-Aβ oligomer fragment antibody reduces various toxic Aβ species in the brain. J Nanobiotechnology 2023; 21:36. [PMID: 36721182 PMCID: PMC9888736 DOI: 10.1186/s12951-023-01772-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 01/07/2023] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Although a large amount of evidence has revealed that amyloid β (Aβ), especially Aβ oligomers, protofibrils, and pyroglutamated Aβs, participate primarily in the pathophysiological processes of Alzheimer's disease, most clinical trials of anti-Aβ antibody therapy have never acquired successful efficacy in human clinical trials, partly because peripheral administration of antibody medications was unable to deliver sufficient amounts of the molecules to the brain. Recently, we developed polymeric nanomicelles capable of passing through the blood-brain barrier that function as chaperones to deliver larger amounts of heavy molecules to the brain. Herein, we aimed to evaluate the efficacy of newly developed antibody 6H4 fragments specific to Aβ oligomers encapsulated in polymeric nanomicelles on the development of Alzheimer's disease pathology in Alzheimer's disease model mice at the age of emergence of early Alzheimer's disease pathology. RESULTS During the 10-week administration of 6H4 antibody fragments in polymeric nanomicelles, a significant reduction in the amounts of various toxic Aβ species, such as Aβ oligomers, toxic Aβ conformers, and pyroglutamated Aβs in the brain was observed. In addition, immunohistochemistry indicated inhibition of diameters of Aβ plaques, Aβ-antibody immunoreactive areas, and also plaque core formation. Behavioral analysis of the mice model revealed that the 6H4 fragments-polymeric nanomicelle group was significantly better at maintaining long-term spatial reference memory in the probe and platform tests of the water maze, thereby indicating inhibition of the pathophysiological process of Alzheimer's disease. CONCLUSIONS The results indicated that the strategy of reducing toxic Aβ species in early dementia owing to Alzheimer's disease by providing sufficient antibodies in the brain may modify Alzheimer's disease progression.
Collapse
Affiliation(s)
- Akiko Amano
- grid.265073.50000 0001 1014 9130Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima Bunkyo-ku, Tokyo, 113-8510 Japan
| | - Nobuo Sanjo
- grid.265073.50000 0001 1014 9130Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima Bunkyo-ku, Tokyo, 113-8510 Japan
| | - Wataru Araki
- grid.265073.50000 0001 1014 9130Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima Bunkyo-ku, Tokyo, 113-8510 Japan
| | - Yasutaka Anraku
- grid.26999.3d0000 0001 2151 536XDepartment of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan ,grid.493442.c0000 0004 5936 3316Innovation Center of Nano Medicine, Kawasaki Institute of Industrial Promotion, Kanagawa, Japan
| | - Makoto Nakakido
- grid.26999.3d0000 0001 2151 536XDepartment of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan ,grid.26999.3d0000 0001 2151 536XDepartment of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Etsuro Matsubara
- grid.412334.30000 0001 0665 3553Department of Neurology, Oita University, Oita, Japan
| | - Takami Tomiyama
- grid.258799.80000 0004 0372 2033Department of Translational Neuroscience, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Tetsuya Nagata
- grid.265073.50000 0001 1014 9130Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima Bunkyo-ku, Tokyo, 113-8510 Japan
| | - Kouhei Tsumoto
- grid.26999.3d0000 0001 2151 536XDepartment of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan ,grid.26999.3d0000 0001 2151 536XDepartment of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, Japan ,grid.26999.3d0000 0001 2151 536XThe Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kazunori Kataoka
- grid.493442.c0000 0004 5936 3316Innovation Center of Nano Medicine, Kawasaki Institute of Industrial Promotion, Kanagawa, Japan
| | - Takanori Yokota
- grid.265073.50000 0001 1014 9130Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima Bunkyo-ku, Tokyo, 113-8510 Japan
| |
Collapse
|
15
|
El-Din Hussein AS, Abou-El Nour RKED, Khorshid OA, Osman AS. Study of the possible effect of sacubitril/valsartan combination versus valsartan on the cognitive function in Alzheimer's disease model in rats. Int J Immunopathol Pharmacol 2023; 37:3946320231161469. [PMID: 36877667 PMCID: PMC9996744 DOI: 10.1177/03946320231161469] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023] Open
Abstract
Objectives: Alzheimer's disease (AD) is an irreversible, progressive neurodegenerative disorder. The proportion of elderly individuals at risk for AD and cardiovascular problems increases by raising life expectancy. The present study was designed to investigate the effect of the sacubitril/valsartan combination compared to that of valsartan alone in a rat model of AD. Methods: 72 male adult Wistar rats were divided into seven groups; control untreated rats received saline, control valsartan-treated rats received valsartan orally, control sacubitril/valsartan treated rats received sacubitril/valsartan orally, model rats received aluminum chloride i.p., model valsartan treated rats received aluminum chloride i.p. and valsartan orally and model sacubitril/valsartan treated rats received aluminum chloride i.p. and sacubitril/valsartan combination orally. All previous treatments continued on a daily basis for 6 weeks. At the second, fourth, and sixth weeks of the experiment, behavioral changes were evaluated using the Morris water maze and novel object recognition tests, and systolic blood pressure was measured. In the end, rat brain malondialdehyde and amyloid-beta 1-42 levels were measured, and the isolated hippocampus was evaluated histopathologically. Results: Valsartan improved AD symptoms in the aluminum-induced rat model, while the sacubitril/valsartan combination significantly worsened all tested parameters in both control and model rats compared with untreated and valsartan-treated animals. Conclusion: Based on the current study's findings, valsartan did not increase the risk for AD development in control rats and improved AD symptoms in a rat model, while sacubitril/valsartan combination increased the risk of AD in control rats and worsened the condition in a rat model.
Collapse
Affiliation(s)
| | | | - Omayma A Khorshid
- Department of Medical Pharmacology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Afaf S Osman
- Department of Medical Pharmacology, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
16
|
Shah AJ, Mohi-Ud-Din R, Sabreen S, Wani TU, Jan R, Javed MN, Mir PA, Mir RH, Masoodi MH. Clinical Biomarkers and Novel Drug Targets to Cut Gordian Knots of Alzheimer's Disease. Curr Mol Pharmacol 2023; 16:254-279. [PMID: 36056834 DOI: 10.2174/1874467215666220903095837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 12/03/2021] [Accepted: 12/16/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Alzheimer's disease (AD), the primary cause of dementia, escalating worldwide, has no proper diagnosis or effective treatment. Neuronal cell death and impairment of cognitive abilities, possibly triggered by several brain mechanisms, are the most significant characteristic of this disorder. METHODS A multitude of pharmacological targets have been identified for potential drug design against AD. Although many advances in treatment strategies have been made to correct various abnormalities, these often exhibit limited clinical significance because this disease aggressively progresses into different regions of the brain, causing severe deterioration. RESULTS These biomarkers can be game-changers for early detection and timely monitoring of such disorders. CONCLUSION This review covers clinically significant biomarkers of AD for precise and early monitoring of risk factors and stages of this disease, the potential site of action and novel targets for drugs, and pharmacological approaches to clinical management.
Collapse
Affiliation(s)
- Abdul Jalil Shah
- Department of Pharmaceutical Sciences, Pharmaceutical Chemistry Division, University of Kashmir, Hazratbal, Srinagar- 190006, Kashmir, India
| | - Roohi Mohi-Ud-Din
- Department of General Medicine, Sher-I-Kashmir Institute of Medical Sciences (SKIMS), Srinagar- 190011, Jammu and Kashmir, India
| | - Saba Sabreen
- Department of Pharmaceutical Sciences, Pharmaceutical Chemistry Division, University of Kashmir, Hazratbal, Srinagar- 190006, Kashmir, India
| | - Taha Umair Wani
- Department of Pharmaceutical Sciences, Pharmaceutics Lab, School of Applied Sciences and Technology, University of Kashmir, Hazratbal, Srinagar-190006, Kashmir India
| | - Rafia Jan
- Defence Research and Development Organization (DRDO), Hospital, Khonmoh, Srinagar 190001, Jammu & Kashmir, India
| | - Md Noushad Javed
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
- Department of Pharmaceutics, KR Mangalam University, Gurugram, India
| | - Prince Ahad Mir
- Khalsa College of Pharmacy, G.T. Road, Amritsar-143002, Punjab, India
| | - Reyaz Hassan Mir
- Department of Pharmaceutical Sciences, Pharmaceutical Chemistry Division, University of Kashmir, Hazratbal, Srinagar- 190006, Kashmir, India
- Pharmaceutical Chemistry Division, Chandigarh College of Pharmacy, Mohali, Punjab 140307, India
| | - Mubashir Hussain Masoodi
- Department of Pharmaceutical Sciences, Pharmaceutical Chemistry Division, University of Kashmir, Hazratbal, Srinagar- 190006, Kashmir, India
| |
Collapse
|
17
|
Zhou X, Huang K, Wang Y, Zhang Z, Liu Y, Hou Q, Yang X, Hoi MPM. Evaluation of therapeutic effects of tetramethylpyrazine nitrone in Alzheimer's disease mouse model and proteomics analysis. Front Pharmacol 2023; 14:1082602. [PMID: 36950017 PMCID: PMC10025301 DOI: 10.3389/fphar.2023.1082602] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 02/22/2023] [Indexed: 03/08/2023] Open
Abstract
The pathophysiology of Alzheimer's disease (AD) is multifactorial with characteristic extracellular accumulation of amyloid-beta (Aβ) and intraneuronal aggregation of hyperphosphorylated tau in the brain. Development of disease-modifying treatment for AD has been challenging. Recent studies suggest that deleterious alterations in neurovascular cells happens in parallel with Aβ accumulation, inducing tau pathology and necroptosis. Therefore, therapies targeting cellular Aβ and tau pathologies may provide a more effective strategy of disease intervention. Tetramethylpyrazine nitrone (TBN) is a nitrone derivative of tetramethylpyrazine, an active ingredient from Ligusticum wallichii Franchat (Chuanxiong). We previously showed that TBN is a potent scavenger of free radicals with multi-targeted neuroprotective effects in rat and monkey models of ischemic stroke. The present study aimed to investigate the anti-AD properties of TBN. We employed AD-related cellular model (N2a/APPswe) and transgenic mouse model (3×Tg-AD mouse) for mechanistic and behavioral studies. Our results showed that TBN markedly improved cognitive functions and reduced Aβ and hyperphosphorylated tau levels in mouse model. Further investigation of the underlying mechanisms revealed that TBN promoted non-amyloidogenic processing pathway of amyloid precursor protein (APP) in N2a/APPswe in vitro. Moreover, TBN preserved synapses from dendritic spine loss and upregulated synaptic protein expressions in 3×Tg-AD mice. Proteomic analysis of 3×Tg-AD mouse hippocampal and cortical tissues showed that TBN induced neuroprotective effects through modulating mitophagy, MAPK and mTOR pathways. In particular, TBN significantly upregulated PINK1, a key protein for mitochondrial homeostasis, implicating PINK1 as a potential therapeutic target for AD. In summary, TBN improved cognitive functions in AD-related mouse model, inhibited Aβ production and tau hyperphosphorylation, and rescued synaptic loss and neuronal damage. Multiple mechanisms underlie the anti-AD effects of TBN including the modulation of APP processing, mTOR signaling and PINK1-related mitophagy.
Collapse
Affiliation(s)
- Xinhua Zhou
- Department of Neurology and Stroke Center, Jinan University College of Pharmacy, The First Affiliated Hospital of Jinan University and Institute of New Drug Research, Guangzhou, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinse Medical Sciences, University of Macau, Macau, China
- Institute of GCP, Guangzhou Eighth People’s Hospital Guangzhou Medical University, Guangzhou, China
| | - Kaipeng Huang
- Institute of GCP, Guangzhou Eighth People’s Hospital Guangzhou Medical University, Guangzhou, China
| | - Yuqiang Wang
- Department of Neurology and Stroke Center, Jinan University College of Pharmacy, The First Affiliated Hospital of Jinan University and Institute of New Drug Research, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic, College of Pharmacy, Institute of New Drug Research, Constituents of Traditional Chinese Medicine & New Drug Research, Jinan University, Guangdong, China
| | - Zaijun Zhang
- Guangdong Province Key Laboratory of Pharmacodynamic, College of Pharmacy, Institute of New Drug Research, Constituents of Traditional Chinese Medicine & New Drug Research, Jinan University, Guangdong, China
| | - Yingying Liu
- Department of Neurology, Daqing People’s Hospital, Daqing, China
| | - Qinghua Hou
- Department of Neurology, Clinical Neuroscience Center, the 7th Affiliated Hospital, Sun-Yat-sen University. Shenzhen, China
- *Correspondence: Maggie Pui Man Hoi, ; Xifei Yang, ; Qinghua Hou,
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
- *Correspondence: Maggie Pui Man Hoi, ; Xifei Yang, ; Qinghua Hou,
| | - Maggie Pui Man Hoi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinse Medical Sciences, University of Macau, Macau, China
- DPS, Faculty of Health Sciences, University of Macau, Macau, China
- *Correspondence: Maggie Pui Man Hoi, ; Xifei Yang, ; Qinghua Hou,
| |
Collapse
|
18
|
Sasaguri H, Hashimoto S, Watamura N, Sato K, Takamura R, Nagata K, Tsubuki S, Ohshima T, Yoshiki A, Sato K, Kumita W, Sasaki E, Kitazume S, Nilsson P, Winblad B, Saito T, Iwata N, Saido TC. Recent Advances in the Modeling of Alzheimer's Disease. Front Neurosci 2022; 16:807473. [PMID: 35431779 PMCID: PMC9009508 DOI: 10.3389/fnins.2022.807473] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 02/22/2022] [Indexed: 12/13/2022] Open
Abstract
Since 1995, more than 100 transgenic (Tg) mouse models of Alzheimer's disease (AD) have been generated in which mutant amyloid precursor protein (APP) or APP/presenilin 1 (PS1) cDNA is overexpressed ( 1st generation models ). Although many of these models successfully recapitulate major pathological hallmarks of the disease such as amyloid β peptide (Aβ) deposition and neuroinflammation, they have suffered from artificial phenotypes in the form of overproduced or mislocalized APP/PS1 and their functional fragments, as well as calpastatin deficiency-induced early lethality, calpain activation, neuronal cell death without tau pathology, endoplasmic reticulum stresses, and inflammasome involvement. Such artifacts bring two important uncertainties into play, these being (1) why the artifacts arise, and (2) how they affect the interpretation of experimental results. In addition, destruction of endogenous gene loci in some Tg lines by transgenes has been reported. To overcome these concerns, single App knock-in mouse models harboring the Swedish and Beyreuther/Iberian mutations with or without the Arctic mutation (AppNL-G-F and AppNL-F mice) were developed ( 2nd generation models ). While these models are interesting given that they exhibit Aβ pathology, neuroinflammation, and cognitive impairment in an age-dependent manner, the model with the Artic mutation, which exhibits an extensive pathology as early as 6 months of age, is not suitable for investigating Aβ metabolism and clearance because the Aβ in this model is resistant to proteolytic degradation and is therefore prone to aggregation. Moreover, it cannot be used for preclinical immunotherapy studies owing to the discrete affinity it shows for anti-Aβ antibodies. The weakness of the latter model (without the Arctic mutation) is that the pathology may require up to 18 months before it becomes sufficiently apparent for experimental investigation. Nevertheless, this model was successfully applied to modulating Aβ pathology by genome editing, to revealing the differential roles of neprilysin and insulin-degrading enzyme in Aβ metabolism, and to identifying somatostatin receptor subtypes involved in Aβ degradation by neprilysin. In addition to discussing these issues, we also provide here a technical guide for the application of App knock-in mice to AD research. Subsequently, a new double knock-in line carrying the AppNL-F and Psen1 P117L/WT mutations was generated, the pathogenic effect of which was found to be synergistic. A characteristic of this 3rd generation model is that it exhibits more cored plaque pathology and neuroinflammation than the AppNL-G-F line, and thus is more suitable for preclinical studies of disease-modifying medications targeting Aβ. Furthermore, a derivative AppG-F line devoid of Swedish mutations which can be utilized for preclinical studies of β-secretase modifier(s) was recently created. In addition, we introduce a new model of cerebral amyloid angiopathy that may be useful for analyzing amyloid-related imaging abnormalities that can be caused by anti-Aβ immunotherapy. Use of the App knock-in mice also led to identification of the α-endosulfine-K ATP channel pathway as components of the somatostatin-evoked physiological mechanisms that reduce Aβ deposition via the activation of neprilysin. Such advances have provided new insights for the prevention and treatment of preclinical AD. Because tau pathology plays an essential role in AD pathogenesis, knock-in mice with human tau wherein the entire murine Mapt gene has been humanized were generated. Using these mice, the carboxy-terminal PDZ ligand of neuronal nitric oxide synthase (CAPON) was discovered as a mediator linking tau pathology to neurodegeneration and showed that tau humanization promoted pathological tau propagation. Finally, we describe and discuss the current status of mutant human tau knock-in mice and a non-human primate model of AD that we have successfully created.
Collapse
Affiliation(s)
- Hiroki Sasaguri
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
| | - Shoko Hashimoto
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
| | - Naoto Watamura
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
| | - Kaori Sato
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
- Laboratory for Molecular Brain Science, Department of Life Science and Medical Bioscience, Waseda University, Shinjuku City, Japan
| | - Risa Takamura
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
- Laboratory for Molecular Brain Science, Department of Life Science and Medical Bioscience, Waseda University, Shinjuku City, Japan
| | - Kenichi Nagata
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Satoshi Tsubuki
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
| | - Toshio Ohshima
- Laboratory for Molecular Brain Science, Department of Life Science and Medical Bioscience, Waseda University, Shinjuku City, Japan
| | - Atsushi Yoshiki
- Experimental Animal Division, RIKEN BioResource Research Center, Tsukuba, Japan
| | - Kenya Sato
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
- Department of Marmoset Biology and Medicine, Central Institute for Experimental Animals, Kawasaki, Japan
| | - Wakako Kumita
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
- Department of Marmoset Biology and Medicine, Central Institute for Experimental Animals, Kawasaki, Japan
| | - Erika Sasaki
- Department of Marmoset Biology and Medicine, Central Institute for Experimental Animals, Kawasaki, Japan
- Laboratory for Marmoset Neural Architecture, RIKEN Center for Brain Science, Wako, Japan
| | - Shinobu Kitazume
- Department of Clinical Laboratory Sciences, School of Health Sciences, Fukushima Medical University, Fukushima, Japan
| | - Per Nilsson
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Bioclinicum, Karolinska Institutet, Stockholm, Sweden
| | - Bengt Winblad
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Bioclinicum, Karolinska Institutet, Stockholm, Sweden
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Nobuhisa Iwata
- Department of Genome-Based Drug Discovery and Leading Medical Research Core Unit, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Takaomi C. Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
| |
Collapse
|
19
|
Watamura N, Kakiya N, Nilsson P, Tsubuki S, Kamano N, Takahashi M, Hashimoto S, Sasaguri H, Saito T, Saido TC. Somatostatin-evoked Aβ catabolism in the brain: Mechanistic involvement of α-endosulfine-K ATP channel pathway. Mol Psychiatry 2022; 27:1816-1828. [PMID: 34737456 PMCID: PMC9095489 DOI: 10.1038/s41380-021-01368-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is characterized by the deposition of amyloid β peptide (Aβ) in the brain. The neuropeptide somatostatin (SST) regulates Aβ catabolism by enhancing neprilysin (NEP)-catalyzed proteolytic degradation. However, the mechanism by which SST regulates NEP activity remains unclear. Here, we identified α-endosulfine (ENSA), an endogenous ligand of the ATP-sensitive potassium (KATP) channel, as a negative regulator of NEP downstream of SST signaling. The expression of ENSA is significantly increased in AD mouse models and in patients with AD. In addition, NEP directly contributes to the degradation of ENSA, suggesting a substrate-dependent feedback loop regulating NEP activity. We also discovered the specific KATP channel subtype that modulates NEP activity, resulting in the Aβ levels altered in the brain. Pharmacological intervention targeting the particular KATP channel attenuated Aβ deposition, with impaired memory function rescued via the NEP activation in our AD mouse model. Our findings provide a mechanism explaining the molecular link between KATP channel and NEP activation, and give new insights into alternative strategies to prevent AD.
Collapse
Affiliation(s)
- Naoto Watamura
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan.
| | - Naomasa Kakiya
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Per Nilsson
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
- Karolinska Institutet, Center for Alzheimer Research, Dept. of Neurobiology, Care Science and Society, Division for Neurogeriatrics, Visionsgatan 4, Solna, 171-64, Sweden
| | - Satoshi Tsubuki
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Naoko Kamano
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Mika Takahashi
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Shoko Hashimoto
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Hiroki Sasaguri
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, 467-8601, Japan
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan.
| |
Collapse
|
20
|
Eldem E, Barve A, Sallin O, Foucras S, Annoni JM, Schmid AW, Alberi Auber L. Salivary Proteomics Identifies Transthyretin as a Biomarker of Early Dementia Conversion. J Alzheimers Dis Rep 2022; 6:31-41. [PMID: 35360272 PMCID: PMC8925122 DOI: 10.3233/adr-210056] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 01/06/2022] [Indexed: 01/18/2023] Open
Abstract
Background: Alzheimer’s disease (AD) remains to date an incurable disease with a long asymptomatic phase. Early diagnosis in peripheral biofluids has emerged as key for identifying subjects at risk and developing therapeutics and preventative approaches. Objective: We apply proteomics discovery to identify salivary diagnostic biomarkers for AD, which are suitable for self-sampling and longitudinal biomonitoring during aging. Methods: 57 participants were recruited for the study and were categorized into Cognitively normal (CNh) (n = 19), mild cognitive impaired (MCI) (n = 21), and Alzheimer’s disease (AD) (n = 17). On a subset of subjects, 3 CNh and 3 mild AD, shot-gun filter aided sample preparation (FASP) proteomics and liquid chromatography mass spectroscopy (LC-MS/MS) was employed in saliva and cerebrospinal fluid (CSF) to identify neural-derived proteins. The protein level of salivary Transthyretin (TTR) was validated using western blot analysis across groups. Results: We found that 19.8% of the proteins in saliva are shared with CSF. When we compared the saliva and CSF proteome, 24 hits were decreased with only one protein expressed more. Among the differentially expressed proteins, TTR with reported function in amyloid misfolding, shows a significant drop in AD samples, confirmed by western blot showing a 0.5-fold reduction in MCI and AD compared to CNh. Conclusion: A reduction in salivary TTR appears with the onset of cognitive symptoms. More in general, the proteomic profiling of saliva shows a plethora of biomarkers worth pursuing as non-invasive hallmarks of dementia in the preclinical stage.
Collapse
Affiliation(s)
- Ece Eldem
- Department of Medicine, Faculty of Science, University of Fribourg, Fribourg, Switzerland
- Swiss Integrative Center for Human Health, Fribourg, Switzerland
| | - Aatmika Barve
- Department of Medicine, Faculty of Science, University of Fribourg, Fribourg, Switzerland
- Swiss Integrative Center for Human Health, Fribourg, Switzerland
| | - Olivier Sallin
- Swiss Integrative Center for Human Health, Fribourg, Switzerland
| | | | - Jean-Marie Annoni
- Department of Medicine, Faculty of Science, University of Fribourg, Fribourg, Switzerland
- Hôpital Cantonal Fribourgeois, Fribourg, Switzerland
| | | | - Lavinia Alberi Auber
- Department of Medicine, Faculty of Science, University of Fribourg, Fribourg, Switzerland
- Swiss Integrative Center for Human Health, Fribourg, Switzerland
| |
Collapse
|
21
|
Catarina Duarte A, Raquel Costa A, Gonçalves I, Quintela T, Preissner R, R A Santos C. The druggability of bitter taste receptors for the treatment of neurodegenerative disorders. Biochem Pharmacol 2022; 197:114915. [PMID: 35051386 DOI: 10.1016/j.bcp.2022.114915] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/05/2022] [Accepted: 01/10/2022] [Indexed: 12/14/2022]
Abstract
The delivery of therapeutic drugs to the brain remains a major pharmacology challenge. A complex system of chemical surveillance to protect the brain from endogenous and exogenous toxicants at brain barriers hinders the uptake of many compounds with significant in vitro and ex vivo therapeutic properties. Despite the advances in the field in recent years, the components of this system are not completely understood. Recently, a large group of chemo-sensing receptors, have been identified in the blood-cerebrospinal fluid barrier. Among these chemo-sensing receptors, bitter taste receptors (TAS2R) hold promise as potential drug targets, as many TAS2R bind compounds with recognized neuroprotective activity (quercetin, resveratrol, among others). Whether activation of TAS2R by their ligands contributes to their diverse biological actions described in other cells and tissues is still debatable. In this review, we discuss the potential role of TAS2R gene family as the mediators of the biological activity of their ligands for the treatment of central nervous system disorders and discuss their potential to counteract drug resistance by improving drug delivery to the brain.
Collapse
Affiliation(s)
- Ana Catarina Duarte
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Covilhã, Portugal; CPIRN-IPG- Centro de Potencial e Inovação de Recursos Naturais- Instituto Politécnico da Guarda, Av. Dr. Francisco de Sá Carneiro, 6300-559, Guarda, Portugal
| | - Ana Raquel Costa
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Covilhã, Portugal
| | - Isabel Gonçalves
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Covilhã, Portugal
| | - Telma Quintela
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Covilhã, Portugal
| | - Robert Preissner
- Institute of Physiology and Science-IT, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Philippstrasse 12, 10115, Berlin, Germany
| | - Cecília R A Santos
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Covilhã, Portugal.
| |
Collapse
|
22
|
Klonarakis M, De Vos M, Woo E, Ralph L, Thacker JS, Gil-Mohapel J. The three sisters of fate: Genetics, pathophysiology and outcomes of animal models of neurodegenerative diseases. Neurosci Biobehav Rev 2022; 135:104541. [DOI: 10.1016/j.neubiorev.2022.104541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 11/28/2021] [Accepted: 01/13/2022] [Indexed: 02/07/2023]
|
23
|
Neumann WL, Sandoval KE, Mobayen S, Minaeian M, Kukielski SG, Srabony KN, Frare R, Slater O, Farr SA, Niehoff ML, Hospital A, Kontoyianni M, Crider AM, Witt KA. Synthesis and structure-activity relationships of 3,4,5-trisubstituted-1,2,4-triazoles: high affinity and selective somatostatin receptor-4 agonists for Alzheimer's disease treatment. RSC Med Chem 2021; 12:1352-1365. [PMID: 34458738 DOI: 10.1039/d1md00044f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 04/14/2021] [Indexed: 12/23/2022] Open
Abstract
Somatostatin receptor-4 (SST4) is highly expressed in brain regions affiliated with learning and memory. SST4 agonist treatment may act to mitigate Alzheimer's disease (AD) pathology. An integrated approach to SST4 agonist lead optimization is presented herein. High affinity and selective agonists with biological efficacy were identified through iterative cycles of a structure-based design strategy encompassing computational methods, chemistry, and preclinical pharmacology. 1,2,4-Triazole derivatives of our previously reported hit (4) showed enhanced SST4 binding affinity, activity, and selectivity. Thirty-five compounds showed low nanomolar range SST4 binding affinity, 12 having a K i < 1 nM. These compounds showed >500-fold affinity for SST4 as compared to SST2A. SST4 activities were consistent with the respective SST4 binding affinities (EC50 < 10 nM for 34 compounds). Compound 208 (SST4 K i = 0.7 nM; EC50 = 2.5 nM; >600-fold selectivity over SST2A) display a favorable physiochemical profile, and was advanced to learning and memory behavior evaluations in the senescence accelerated mouse-prone 8 model of AD-related cognitive decline. Chronic administration enhanced learning with i.p. dosing (1 mg kg-1) compared to vehicle. Chronic administration enhanced memory with both i.p. (0.01, 0.1, 1 mg kg-1) and oral (0.01, 10 mg kg-1) dosing compared to vehicle. This study identified a novel series of SST4 agonists with high affinity, selectivity, and biological activity that may be useful in the treatment of AD.
Collapse
Affiliation(s)
- William L Neumann
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville Edwardsville IL 62026 USA
| | - Karin E Sandoval
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville Edwardsville IL 62026 USA
| | - Shirin Mobayen
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville Edwardsville IL 62026 USA
| | - Mahsa Minaeian
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville Edwardsville IL 62026 USA
| | - Stephen G Kukielski
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville Edwardsville IL 62026 USA
| | - Khush N Srabony
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville Edwardsville IL 62026 USA
| | - Rafael Frare
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville Edwardsville IL 62026 USA
| | - Olivia Slater
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville Edwardsville IL 62026 USA
| | - Susan A Farr
- Research and Development Service, VA Medical Center, Division of Geriatric Medicine, Saint Louis University School of Medicine 1402 South Grand Boulevard, M238 St Louis MO 63104 USA
| | - Michael L Niehoff
- Research and Development Service, VA Medical Center, Division of Geriatric Medicine, Saint Louis University School of Medicine 1402 South Grand Boulevard, M238 St Louis MO 63104 USA
| | - Audrey Hospital
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville Edwardsville IL 62026 USA
| | - Maria Kontoyianni
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville Edwardsville IL 62026 USA
| | - A Michael Crider
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville Edwardsville IL 62026 USA
| | - Ken A Witt
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville Edwardsville IL 62026 USA
| |
Collapse
|
24
|
Wojtunik-Kulesza K, Rudkowska M, Kasprzak-Drozd K, Oniszczuk A, Borowicz-Reutt K. Activity of Selected Group of Monoterpenes in Alzheimer's Disease Symptoms in Experimental Model Studies-A Non-Systematic Review. Int J Mol Sci 2021; 22:7366. [PMID: 34298986 PMCID: PMC8306454 DOI: 10.3390/ijms22147366] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia and cognitive function impairment. The multi-faced character of AD requires new drug solutions based on substances that incorporate a wide range of activities. Antioxidants, AChE/BChE inhibitors, BACE1, or anti-amyloid platelet aggregation substances are most desirable because they improve cognition with minimal side effects. Plant secondary metabolites, used in traditional medicine and pharmacy, are promising. Among these are the monoterpenes-low-molecular compounds with anti-inflammatory, antioxidant, enzyme inhibitory, analgesic, sedative, as well as other biological properties. The presented review focuses on the pathophysiology of AD and a selected group of anti-neurodegenerative monoterpenes and monoterpenoids for which possible mechanisms of action have been explained. The main body of the article focuses on monoterpenes that have shown improved memory and learning, anxiolytic and sleep-regulating effects as determined by in vitro and in silico tests-followed by validation in in vivo models.
Collapse
Affiliation(s)
| | - Monika Rudkowska
- Independent Experimental Neuropathophysiology Unit, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland; (M.R.); (K.B.-R.)
| | - Kamila Kasprzak-Drozd
- Department of Inorganic Chemistry, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland;
| | - Anna Oniszczuk
- Department of Inorganic Chemistry, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland;
| | - Kinga Borowicz-Reutt
- Independent Experimental Neuropathophysiology Unit, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland; (M.R.); (K.B.-R.)
| |
Collapse
|
25
|
Welcome MO, Mastorakis NE. The taste of neuroinflammation: Molecular mechanisms linking taste sensing to neuroinflammatory responses. Pharmacol Res 2021; 167:105557. [PMID: 33737243 DOI: 10.1016/j.phrs.2021.105557] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 03/10/2021] [Accepted: 03/10/2021] [Indexed: 02/07/2023]
Abstract
Evidence indicates a critical role of neuroinflammatory response as an underlying pathophysiological process in several central nervous system disorders, including neurodegenerative diseases. However, the molecular mechanisms that trigger neuroinflammatory processes are not fully known. The discovery of bitter taste receptors in regions other than the oral cavity substantially increased research interests on their functional roles in extra-oral tissues. It is now widely accepted that bitter taste receptors, for instance, in the respiratory, intestinal, reproductive and urinary tracts, are crucial not only for sensing poisonous substances, but also, act as immune sentinels, mobilizing defense mechanisms against pathogenic aggression. The relatively recent discovery of bitter taste receptors in the brain has intensified research investigation on the functional implication of cerebral bitter taste receptor expression. Very recent data suggest that responses of bitter taste receptors to neurotoxins and microbial molecules, under normal condition, are necessary to prevent neuroinflammatory reactions. Furthermore, emerging data have revealed that downregulation of key components of the taste receptor signaling cascade leads to increased oxidative stress and inflammasome signaling in neurons that ultimately culminate in neuroinflammation. Nevertheless, the mechanisms that link taste receptor mediated surveillance of the extracellular milieu to neuroinflammatory responses are not completely understood. This review integrates new data on the molecular mechanisms that link bitter taste receptor sensing to neuroinflammatory responses. The role of bitter taste receptor-mediated sensing of toxigenic substances in brain disorders is also discussed. The therapeutic significance of targeting these receptors for potential treatment of neurodegenerative diseases is also highlighted.
Collapse
Affiliation(s)
- Menizibeya O Welcome
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Nile University of Nigeria, Abuja, Nigeria.
| | | |
Collapse
|
26
|
Tamano H, Togo J, Sato Y, Shioya A, Tempaku M, Takeda A. Retention Period of Amyloid β 1-42 in the Brain Extracellular Fluid as the Toxicological Determinant in Freely Moving Rats. Biol Pharm Bull 2020; 43:1975-1978. [PMID: 33268719 DOI: 10.1248/bpb.b20-00565] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The pathological significance of amyloid-β1-42 (Aβ1-42) dynamics is poorly understood in the brain extracellular compartment. Here we test which of the concentration or the retention is critical for Aβ1-42 toxicity after injection of equal dose into dentate granule cell layer of freely moving rats. The toxicity of Aβ1-42 (25 µM) was compared between injections at the rate of 0.25 µL/min for 4 min (fast injection) and 0.025 µL/min for 40 min (slow injection). Dentate gyrus long-term potentiation (LTP) was affected 1 and 2 h after the fast injection, but not 4 h. In contrast, LTP was affected even 72 h after the slow injection. Aβ1-42 staining 5 min after finish of the slow injection was more intense in the dentate granule cell layer than of the fast injection. The present study indicates that the retention of Aβ1-42 in the extracellular fluid is correlated with neuronal Aβ1-42 uptake and plays a key role in Aβ1-42 neurotoxicity. In the extracellular fluid of the dentate gyrus, the retention period of Aβ1-42 is much more critical for Aβ1-42 toxicity than Aβ1-42 concentration. It is likely that Aβ1-42 toxicity is accelerated by the disturbance of Aβ1-42 metabolism in the dentate gyrus.
Collapse
Affiliation(s)
- Haruna Tamano
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka
| | - Junichi Togo
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka
| | - Yuichi Sato
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka
| | - Aoi Shioya
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka
| | - Munekazu Tempaku
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka
| | - Atsushi Takeda
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka
| |
Collapse
|
27
|
Peng HB, Noh K, Pan SR, Saldivia V, Serson S, Toscan A, de Lannoy IAM, Pang KS. Human Amyloid-β 40 Kinetics after Intravenous and Intracerebroventricular Injections and Calcitriol Treatment in Rats In Vivo. Drug Metab Dispos 2020; 48:944-955. [PMID: 32759365 DOI: 10.1124/dmd.120.090886] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 07/08/2020] [Indexed: 02/13/2025] Open
Abstract
Amyloid-β peptides of 40 and 42 amino acid lengths, which are synthesized in neurons and degraded in the brain and liver, have the potential to aggregate and form neuritic plaques in Alzheimer disease. The kinetics of human amyloid-β (hAβ) 40 were examined in the rat pursuant to intravenous and intracerebroventricular administration after pretreatment with calcitriol, the active vitamin D receptor ligand (6.4 nmol·kg-1 in 0.3 ml corn oil every other day for four intraperitoneal doses) to induce P-glycoprotein (P-gp) and enhance hAβ40 brain efflux. The interference of hAβ40 by media matrix that suppressed absorbance readings in the ELISA assay was circumvented with use of different calibration curves prepared in Standard Dilution Buffer, undiluted, 10-10,000 or 5-fold diluted plasma, or artificial cerebrospinal fluid. Simultaneous fitting of hAβ40 plasma and cerebrospinal fluid (CSF) data after intravenous and intracerebroventricular administration were described by catenary-mammillary models comprising of a central and two peripheral compartments, the brain, and one to four CSF compartments. The model with only one CSF compartment (model I) best fitted the intravenous data that showed a faster plasma decay t1/2 and slower equilibration between plasma and brain/CSF. Calcitriol induction increased the brain efflux rate constant, k41 (1.8-fold), at the blood-brain barrier when compared with the control group, as confirmed by the 2-fold (P < 0.05) increase in brain P-gp relative protein expression. SIGNIFICANCE STATEMENT: An accurate description of the kinetic behavior of human amyloid-β (hAβ) 40 is needed in defining the toxic peptide as a biomarker of Alzheimer disease. Modeling of hAβ40 data after intravenous and intracerebroventricular administration to the rat revealed an initially faster plasma half-life that reflected faster peripheral distribution but slower equilibration between plasma and brain/cerebrospinal fluid even with calcitriol pretreatment that increased P-glycoprotein protein expression and enhanced efflux clearance from brain.
Collapse
Affiliation(s)
- H Benson Peng
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada (H.B.P., K.N., K.S.P.) and InterVivo Solutions Inc., Mississauga, Ontario, Canada (S.R.P., V.S., S.S., A.T., I.A.M.d.L.)
| | - Keumhan Noh
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada (H.B.P., K.N., K.S.P.) and InterVivo Solutions Inc., Mississauga, Ontario, Canada (S.R.P., V.S., S.S., A.T., I.A.M.d.L.)
| | - Sophie R Pan
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada (H.B.P., K.N., K.S.P.) and InterVivo Solutions Inc., Mississauga, Ontario, Canada (S.R.P., V.S., S.S., A.T., I.A.M.d.L.)
| | - Victor Saldivia
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada (H.B.P., K.N., K.S.P.) and InterVivo Solutions Inc., Mississauga, Ontario, Canada (S.R.P., V.S., S.S., A.T., I.A.M.d.L.)
| | - Sylvia Serson
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada (H.B.P., K.N., K.S.P.) and InterVivo Solutions Inc., Mississauga, Ontario, Canada (S.R.P., V.S., S.S., A.T., I.A.M.d.L.)
| | - Anja Toscan
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada (H.B.P., K.N., K.S.P.) and InterVivo Solutions Inc., Mississauga, Ontario, Canada (S.R.P., V.S., S.S., A.T., I.A.M.d.L.)
| | - Inés A M de Lannoy
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada (H.B.P., K.N., K.S.P.) and InterVivo Solutions Inc., Mississauga, Ontario, Canada (S.R.P., V.S., S.S., A.T., I.A.M.d.L.)
| | - K Sandy Pang
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada (H.B.P., K.N., K.S.P.) and InterVivo Solutions Inc., Mississauga, Ontario, Canada (S.R.P., V.S., S.S., A.T., I.A.M.d.L.)
| |
Collapse
|
28
|
Irmak DK, Darıcı H, Karaöz E. Stem Cell Based Therapy Option in COVID-19: Is It Really Promising? Aging Dis 2020; 11:1174-1191. [PMID: 33014531 PMCID: PMC7505270 DOI: 10.14336/ad.2020.0608] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 06/08/2020] [Indexed: 01/08/2023] Open
Abstract
The COVID-19 patients were first detected in China, in December 2019, then the novel virus with associated pneumonia and other diseases spread quickly to worldwide becoming a serious public health intimidation. Despite all the efforts, the pharmacological agents used for controlling or treating the disease, especially respiratory problems, have not been accomplished so far. Among various treatment options, mesenchymal stem cell-based cellular therapies are being investigated, because of their regeneration ability and multipotency along with other features like immunomodulation, antifibrosis and anti-inflammatory effects. This paper intends to analyze the current clinical trials on stem cell treatment of novel virus, searching and reviewing the available information and the International Clinical Trials Registry Platform (ICTRP) of World Health Organization (WHO). We concluded that the stem cell treatment of COVID-19 is found promising with pilot studies' results, but still in the early development phase. There is an urgent need for large-scale investigations to confirm and validate the safety and efficacy profile of these therapies with reliable scientific evidence.
Collapse
Affiliation(s)
- Duygu Koyuncu Irmak
- Istinye University, Faculty of Medicine, Department of Histology & Embryology, Istanbul, Turkey
- Istinye University, Stem Cell and Tissue Engineering R&D Center, Istanbul, Turkey
| | - Hakan Darıcı
- Istinye University, Faculty of Medicine, Department of Histology & Embryology, Istanbul, Turkey
- Istinye University, Stem Cell and Tissue Engineering R&D Center, Istanbul, Turkey
- Istinye University, 3D Bioprinting Design & Prototyping R&D Center, Istanbul, Turkey
| | - Erdal Karaöz
- Istinye University, Faculty of Medicine, Department of Histology & Embryology, Istanbul, Turkey
- Istinye University, Stem Cell and Tissue Engineering R&D Center, Istanbul, Turkey
- Istinye University, 3D Bioprinting Design & Prototyping R&D Center, Istanbul, Turkey
- Liv Hospital, Stem Cell and Regenerative Therapies Center (LivMedCell), Istanbul, Turkey
| |
Collapse
|
29
|
Kumar V, De P, Ojha PK, Saha A, Roy K. A Multi-layered Variable Selection Strategy for QSAR Modeling of Butyrylcholinesterase Inhibitors. Curr Top Med Chem 2020; 20:1601-1627. [DOI: 10.2174/1568026620666200616142753] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 07/23/2019] [Accepted: 10/28/2019] [Indexed: 02/08/2023]
Abstract
Background:
Alzheimer’s disease (AD), a neurological disorder, is the most common cause
of senile dementia. Butyrylcholinesterase (BuChE) enzyme plays a vital role in regulating the brain acetylcholine
(ACh) neurotransmitter, but in the case of Alzheimer’s disease (AD), BuChE activity gradually
increases in patients with a decrease in the acetylcholine (ACh) concentration via hydrolysis. ACh
plays an essential role in regulating learning and memory as the cortex originates from the basal forebrain,
and thus, is involved in memory consolidation in these sites.
Methods:
In this work, we have developed a partial least squares (PLS)-regression based two dimensional
quantitative structure-activity relationship (2D-QSAR) model using 1130 diverse chemical classes
of compounds with defined activity against the BuChE enzyme. Keeping in mind the strict Organization
for Economic Co-operation and Development (OECD) guidelines, we have tried to select significant
descriptors from the large initial pool of descriptors using multi-layered variable selection strategy using
stepwise regression followed by genetic algorithm (GA) followed by again stepwise regression technique
and at the end best subset selection prior to development of final model thus reducing noise in the
input. Partial least squares (PLS) regression technique was employed for the development of the final
model while model validation was performed using various stringent validation criteria.
Results:
The results obtained from the QSAR model suggested that the quality of the model is acceptable
in terms of both internal (R2= 0.664, Q2= 0.650) and external (R2
Pred= 0.657) validation parameters.
The QSAR studies were analyzed, and the structural features (hydrophobic, ring aromatic and hydrogen
bond acceptor/donor) responsible for enhancement of the activity were identified. The developed model
further suggests that the presence of hydrophobic features like long carbon chain would increase the
BuChE inhibitory activity and presence of amino group and hydrazine fragment promoting the hydrogen
bond interactions would be important for increasing the inhibitory activity against BuChE enzyme.
Conclusion:
Furthermore, molecular docking studies have been carried out to understand the molecular
interactions between the ligand and receptor, and the results are then correlated with the structural features
obtained from the QSAR models. The information obtained from the QSAR models are well corroborated
with the results of the docking study.
Collapse
Affiliation(s)
- Vinay Kumar
- Drug Theoretics and Cheminformatics Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Priyanka De
- Drug Theoretics and Cheminformatics Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Probir Kumar Ojha
- Drug Theoretics and Cheminformatics Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Achintya Saha
- Department of Chemical Technology, University of Calcutta, 92 APC Road, Kolkata 700 032, India
| | - Kunal Roy
- Drug Theoretics and Cheminformatics Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| |
Collapse
|
30
|
|
31
|
Wu Y, Gong Y, Luan Y, Li Y, Liu J, Yue Z, Yuan B, Sun J, Xie C, Li L, Zhen J, Jin X, Zheng Y, Wang X, Xie L, Wang W. BHBA treatment improves cognitive function by targeting pleiotropic mechanisms in transgenic mouse model of Alzheimer's disease. FASEB J 2019; 34:1412-1429. [PMID: 31914599 DOI: 10.1096/fj.201901984r] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 09/17/2019] [Accepted: 11/14/2019] [Indexed: 12/19/2022]
Abstract
Accumulation of amyloid β (Aβ) peptide, inflammation, and oxidative stress contribute to Alzheimer's disease (AD) and trigger complex pathogenesis. The ketone body β-hydroxybutyrate (BHBA) is an endogenous metabolic intermediate that protects against stroke and neurodegenerative diseases, but the underlying mechanisms are unclear. The present study aims to elucidate the protective effects of BHBA in the early stage of AD model and investigate the underlying molecular mechanisms. Three-and-half-month-old double-transgenic mice (5XFAD) overexpressing β-amyloid precursor protein (APP) and presenilin-1 (PS1) were used as the AD model. The 5XFAD mice received 1.5 mmol/kg/d BHBA subcutaneously for 28 days. Morris water maze test, nest construction, and passive avoidance experiments were performed to assess the therapeutic effects on AD prevention in vivo, and brain pathology of 5XFAD mice including amyloid plaque deposition and microglia activation were assessed. Gene expression profiles in the cortexes of 5XFAD- and BHBA-treated 5XFAD mice were performed with high-throughput sequencing and bioinformatic analysis. Mouse HT22 cells were treated with 2 mM BHBA to explore its in vitro protective effects of BHBA on hippocampal neurons against Aβ oligomer toxicity, ATP production, ROS generation, and mitochondrial aerobic respiratory function. APP, BACE1, and neprilysin (NEP) expression levels were evaluated in HT22 cells following treatment with BHBA by measuring the presence or absence of G protein-coupled receptor 109A (GPR109A). BHBA improved cognitive function of 5XFAD mice in Morris water maze test, nesting construction and passive avoidance experiments, and attenuated Aβ accumulation and microglia overactivation in the brain. BHBA also enhanced mitochondrial respiratory function of hippocampal neurons and protected it from Aβ toxicity. The enzymes, APP and NEP were regulated by BHBA via G-protein-coupled receptor 109A (GPR109A). Furthermore, RNA sequencing revealed that BHBA-regulated genes mainly annotated in aging, immune system, nervous system, and neurodegenerative diseases. Our data suggested that BHBA confers protection against the AD-like pathological events in the AD mouse model by targeting multiple aspects of AD and it may become a promising candidate for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Yancheng Wu
- Innovative Institute of Animal Healthy Breeding, Key Laboratory of Waterfowl Healthy Breeding of Guangdong Province, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, P.R. China.,College of Veterinary Medicine, Jilin University, Changchun, P.R. China
| | - Yuhong Gong
- College of Veterinary Medicine, Jilin University, Changchun, P.R. China
| | - Yongxin Luan
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P.R. China
| | - Yang Li
- Department of Neurobiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, P.R. China
| | - Juxiong Liu
- College of Veterinary Medicine, Jilin University, Changchun, P.R. China
| | - Zitong Yue
- Changchun Jida Middle School Experimental School, Changchun, P.R. China
| | - Boyu Yuan
- College of Veterinary Medicine, Jilin University, Changchun, P.R. China
| | - Jingxuan Sun
- College of Veterinary Medicine, Jilin University, Changchun, P.R. China
| | - Changxin Xie
- College of Veterinary Medicine, Jilin University, Changchun, P.R. China
| | - Lijuan Li
- Department of Neurobiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, P.R. China.,The Second Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Junli Zhen
- Department of Neurobiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, P.R. China
| | - Xinxin Jin
- College of Veterinary Medicine, Jilin University, Changchun, P.R. China
| | - Yan Zheng
- Department of Neurobiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, P.R. China
| | - Xiaomin Wang
- Department of Neurobiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, P.R. China
| | - Liwei Xie
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangzhou, P.R. China
| | - Wei Wang
- Innovative Institute of Animal Healthy Breeding, Key Laboratory of Waterfowl Healthy Breeding of Guangdong Province, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, P.R. China.,College of Veterinary Medicine, Jilin University, Changchun, P.R. China.,Department of Neurobiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, P.R. China
| |
Collapse
|
32
|
Yaghoubi Y, Movassaghpour A, Zamani M, Talebi M, Mehdizadeh A, Yousefi M. Human umbilical cord mesenchymal stem cells derived-exosomes in diseases treatment. Life Sci 2019; 233:116733. [PMID: 31394127 DOI: 10.1016/j.lfs.2019.116733] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Accepted: 08/04/2019] [Indexed: 02/08/2023]
Abstract
Exosomes are extracellular vesicles with the size of 40-100 nm in diameter and a density of 1.13-1.19 g/mL, containing proteins, mRNAs, miRNAs, and DNAs. Exosomes change the recipient cells biochemical features through biomolecules delivery and play a role in cellular communication. These vesicles are produced from body fluids and different cell types like mesenchymal stem cells (MSCs). Evidence suggests that mesenchymal stem cells-derived exosome (MSC-EXO) exhibit functions similar to MSCs with low immunogenicity and no tumorization. MSCs can also be isolated from a variety of sources including human umbilical cord (HUC). Because of the non-invasive collection method, higher proliferation and lower immunogenicity, HUCMSC-EXO has been frequently used in regenerative medicine and various diseases treatment compared to the other MSC-EXO resources. This review aimed to investigate the applications of HUCMSC-EXO in different diseases.
Collapse
Affiliation(s)
- Yoda Yaghoubi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - AliAkbar Movassaghpour
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Majid Zamani
- Department of Immunology, Tabriz university of Medical Sciences, Tabriz, Iran
| | - Mehdi Talebi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Endocrine Research Center, Tabriz university of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Endocrine Research Center, Tabriz university of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
33
|
Potential Benefits of Nobiletin, A Citrus Flavonoid, against Alzheimer's Disease and Parkinson's Disease. Int J Mol Sci 2019; 20:ijms20143380. [PMID: 31295812 PMCID: PMC6678479 DOI: 10.3390/ijms20143380] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/05/2019] [Accepted: 07/05/2019] [Indexed: 12/17/2022] Open
Abstract
Alzheimer’s disease (AD), which is characterized by the presence of amyloid-β (Aβ) plaques and neurofibrillary tangles, accompanied by neurodegeneration, is the most common form of age-related neurodegenerative disease. Parkinson’s disease (PD) is the second most common neurodegenerative disease after AD, and is characterized by early prominent loss of dopaminergic neurons in the substantia nigra pars compacta. As currently available treatments are not able to significantly alter the progression of these diseases, successful therapeutic and preventive interventions are strongly needed. In the course of our survey of substances from natural resources having anti-dementia and neuroprotective activity, we found nobiletin, a polymethoxylated flavone from the peel of Citrus depressa. Nobiletin improved cognitive deficits and the pathological features of AD, such as Aβ pathology, hyperphosphorylation of tau, and oxidative stress, in animal models of AD. In addition, nobiletin improved motor and cognitive deficits in PD animal models. These observations suggest that nobiletin has the potential to become a novel drug for the treatment and prevention of neurodegenerative diseases such as AD and PD.
Collapse
|
34
|
Mohajeri M, Behnam B, Barreto GE, Sahebkar A. Carbon nanomaterials and amyloid-beta interactions: potentials for the detection and treatment of Alzheimer's disease? Pharmacol Res 2019; 143:186-203. [DOI: 10.1016/j.phrs.2019.03.023] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 03/26/2019] [Accepted: 03/26/2019] [Indexed: 01/24/2023]
|
35
|
Wooster J, Cook EA, Shipman D. Psychiatric Manifestations With Sacubitril/Valsartan: A Case Report. J Pharm Pract 2019; 33:553-557. [PMID: 30991886 DOI: 10.1177/0897190019842700] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Sacubitril/valsartan is an angiotensin receptor-neprilysin inhibitor approved for the treatment of heart failure with reduced ejection fraction (HFrEF). Valsartan is well studied, but sacubitril has much left to understand. This report describes a 31-year-old African American female diagnosed with HFrEF who presented with a 7-day history of psychiatric symptoms following a dose increase in sacubitril/valsartan. Prior to the dose increase, the patient had no history of psychiatric diagnoses, but upon hospital presentation, family described instances of confabulation, paranoia, delusions, hallucinations, and sleep disturbances. Laboratory tests were unremarkable, ruling out infectious processes and illicit substance use. However, cranial computed tomography scans depicted intracranial volume loss abnormal for age with commensurate mild ventricular enlargement. Sacubitril/valsartan was discontinued inpatient, symptoms resolved, and the medication intolerance was documented. Clinical trials involving sacubitril/valsartan lack systematic documentation of cognitive symptoms, but active studies exploring the role of neprilysin inhibition may expand knowledge of possible psychiatric adverse effects.
Collapse
Affiliation(s)
- Jessica Wooster
- University of Texas at Tyler, Ben and Maytee Fisch College of Pharmacy, Tyler, TX, USA
| | - Elizabeth A Cook
- University of Texas at Tyler, Ben and Maytee Fisch College of Pharmacy, Tyler, TX, USA
| | - Denver Shipman
- University of Texas at Tyler, Ben and Maytee Fisch College of Pharmacy, Tyler, TX, USA
| |
Collapse
|
36
|
Medeiros ADM, Silva RH. Sex Differences in Alzheimer’s Disease: Where Do We Stand? J Alzheimers Dis 2019; 67:35-60. [DOI: 10.3233/jad-180213] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- André de Macêdo Medeiros
- Behavioral Neuroscience Laboratory, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
- Center of Health and Biological Sciences, Universidade Federal Rural do Semiárido, Mossoró, Brazil
| | - Regina Helena Silva
- Behavioral Neuroscience Laboratory, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
37
|
Abstract
Mechanisms for elimination of metabolites from ISF include metabolism, blood-brain barrier transport and non-selective, perivascular efflux, this last being assessed by measuring the clearance of markers like inulin. Clearance describes elimination. Clearance of a metabolite generated within the brain is determined as its elimination rate divided by its concentration in interstitial fluid (ISF). However, the more frequently measured parameter is the rate constant for elimination determined as elimination rate divided by amount present, which thus depends on both the elimination processes and the distribution of the metabolite in the brain. The relative importance of the various elimination mechanisms depends on the particular metabolite. Little is known about the effects of sleep on clearance via metabolism or blood-brain barrier transport, but studies with inulin in mice comparing perivascular effluxes during sleep and wakefulness reveal a 4.2-fold increase in clearance. Amongst the important brain metabolites considered, CO2 is eliminated so rapidly across the blood-brain barrier that clearance is blood flow limited and elimination quickly balances production. Glutamate is removed from ISF primarily by uptake into astrocytes and conversion to glutamine, but also by transport across the blood-brain barrier. Both lactate and amyloid-β are eliminated by metabolism, blood-brain barrier transport and perivascular efflux and both show decreased production, decreased ISF concentration and increased perivascular clearance during sleep. Taken altogether available data indicate that sleep increases perivascular and non-perivascular clearances for amyloid-β which reduces its concentration and may have long-term consequences for the formation of plaques and cerebral arterial deposits.
Collapse
Affiliation(s)
- Stephen B Hladky
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD, UK.
| | - Margery A Barrand
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD, UK
| |
Collapse
|
38
|
Sharma P, Srivastava P, Seth A, Tripathi PN, Banerjee AG, Shrivastava SK. Comprehensive review of mechanisms of pathogenesis involved in Alzheimer's disease and potential therapeutic strategies. Prog Neurobiol 2018; 174:53-89. [PMID: 30599179 DOI: 10.1016/j.pneurobio.2018.12.006] [Citation(s) in RCA: 224] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 12/04/2018] [Accepted: 12/28/2018] [Indexed: 12/14/2022]
Abstract
AD is a progressive neurodegenerative disorder and a leading cause of dementia in an aging population worldwide. The enormous challenge which AD possesses to global healthcare makes it as urgent as ever for the researchers to develop innovative treatment strategies to fight this disease. An in-depth analysis of the extensive available data associated with the AD is needed for a more comprehensive understanding of underlying molecular mechanisms and pathophysiological pathways associated with the onset and progression of the AD. The currently understood pathological and biochemical manifestations include cholinergic, Aβ, tau, excitotoxicity, oxidative stress, ApoE, CREB signaling pathways, insulin resistance, etc. However, these hypotheses have been criticized with several conflicting reports for their involvement in the disease progression. Several issues need to be addressed such as benefits to cost ratio with cholinesterase therapy, the dilemma of AChE selectivity over BChE, BBB permeability of peptidic BACE-1 inhibitors, hurdles related to the implementation of vaccination and immunization therapy, and clinical failure of candidates related to newly available targets. The present review provides an insight to the different molecular mechanisms involved in the development and progression of the AD and potential therapeutic strategies, enlightening perceptions into structural information of conventional and novel targets along with the successful applications of computational approaches for the design of target-specific inhibitors.
Collapse
Affiliation(s)
- Piyoosh Sharma
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Pavan Srivastava
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Ankit Seth
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Prabhash Nath Tripathi
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Anupam G Banerjee
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Sushant K Shrivastava
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India.
| |
Collapse
|
39
|
Hladky SB, Barrand MA. Elimination of substances from the brain parenchyma: efflux via perivascular pathways and via the blood-brain barrier. Fluids Barriers CNS 2018; 15:30. [PMID: 30340614 PMCID: PMC6194691 DOI: 10.1186/s12987-018-0113-6] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 08/30/2018] [Indexed: 02/06/2023] Open
Abstract
This review considers efflux of substances from brain parenchyma quantified as values of clearances (CL, stated in µL g-1 min-1). Total clearance of a substance is the sum of clearance values for all available routes including perivascular pathways and the blood-brain barrier. Perivascular efflux contributes to the clearance of all water-soluble substances. Substances leaving via the perivascular routes may enter cerebrospinal fluid (CSF) or lymph. These routes are also involved in entry to the parenchyma from CSF. However, evidence demonstrating net fluid flow inwards along arteries and then outwards along veins (the glymphatic hypothesis) is still lacking. CLperivascular, that via perivascular routes, has been measured by following the fate of exogenously applied labelled tracer amounts of sucrose, inulin or serum albumin, which are not metabolized or eliminated across the blood-brain barrier. With these substances values of total CL ≅ 1 have been measured. Substances that are eliminated at least partly by other routes, i.e. across the blood-brain barrier, have higher total CL values. Substances crossing the blood-brain barrier may do so by passive, non-specific means with CLblood-brain barrier values ranging from < 0.01 for inulin to > 1000 for water and CO2. CLblood-brain barrier values for many small solutes are predictable from their oil/water partition and molecular weight. Transporters specific for glucose, lactate and many polar substrates facilitate efflux across the blood-brain barrier producing CLblood-brain barrier values > 50. The principal route for movement of Na+ and Cl- ions across the blood-brain barrier is probably paracellular through tight junctions between the brain endothelial cells producing CLblood-brain barrier values ~ 1. There are large fluxes of amino acids into and out of the brain across the blood-brain barrier but only small net fluxes have been observed suggesting substantial reuse of essential amino acids and α-ketoacids within the brain. Amyloid-β efflux, which is measurably faster than efflux of inulin, is primarily across the blood-brain barrier. Amyloid-β also leaves the brain parenchyma via perivascular efflux and this may be important as the route by which amyloid-β reaches arterial walls resulting in cerebral amyloid angiopathy.
Collapse
Affiliation(s)
- Stephen B. Hladky
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD UK
| | - Margery A. Barrand
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD UK
| |
Collapse
|
40
|
Ding M, Shen Y, Wang P, Xie Z, Xu S, Zhu Z, Wang Y, Lyu Y, Wang D, Xu L, Bi J, Yang H. Exosomes Isolated From Human Umbilical Cord Mesenchymal Stem Cells Alleviate Neuroinflammation and Reduce Amyloid-Beta Deposition by Modulating Microglial Activation in Alzheimer's Disease. Neurochem Res 2018; 43:2165-2177. [PMID: 30259257 DOI: 10.1007/s11064-018-2641-5] [Citation(s) in RCA: 180] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 09/09/2018] [Accepted: 09/17/2018] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease characterized by excessive accumulation of the amyloid-β peptide (Aβ) in the brain, which has been considered to mediate the neuroinflammation process. Microglial activation is the main component of neuroimmunoregulation. In recent years, exosomes isolated from human umbilical cord mesenchymal stem cells (hucMSC-exosomes) have been demonstrated to mimic the therapeutic effects of hucMSCs in many inflammation-related diseases. In this study, exosomes from the supernatant of hucMSCs were injected into AD mouse models. We observed that hucMSC-exosomes injection could repair cognitive disfunctions and help to clear Aβ deposition in these mice. Moreover, we found that hucMSC-exosomes injection could modulate the activation of microglia in brains of the mice to alleviated neuroinflammation. The levels of pro-inflammatory cytokines in peripheral blood and brains of mice were increased and the levels of anti-inflammatory cytokines were decreased. We also treated BV2 cells with hucMSC-exosomes in culture medium. HucMSC-exosomes also had inflammatory regulating effects to alternatively activate microglia and modulate the levels of inflammatory cytokines in vitro.
Collapse
Affiliation(s)
- Mao Ding
- Medicine School, Shandong University, Jinan, 250012, China
| | - Yang Shen
- Medicine School, Shandong University, Jinan, 250012, China
| | - Ping Wang
- Department of Neurology Medicine, Second Hospital of Shandong University, Jinan, 250033, China
| | - Zhaohong Xie
- Department of Neurology Medicine, Second Hospital of Shandong University, Jinan, 250033, China
| | - Shunliang Xu
- Department of Neurology Medicine, Second Hospital of Shandong University, Jinan, 250033, China
| | - ZhengYu Zhu
- Department of Neurology Medicine, Second Hospital of Shandong University, Jinan, 250033, China
| | - Yun Wang
- Department of Neurology Medicine, Second Hospital of Shandong University, Jinan, 250033, China
| | - Yongtao Lyu
- Department of Neurology Medicine, Shandong Provincial Third Hospital, Jinan, 250031, China
| | - Dewei Wang
- Department of Neurology Medicine, Second Hospital of Shandong University, Jinan, 250033, China
| | - Linlin Xu
- Department of Neurology Medicine, Second Hospital of Shandong University, Jinan, 250033, China
| | - JianZhong Bi
- Department of Neurology Medicine, Second Hospital of Shandong University, Jinan, 250033, China.
| | - Hui Yang
- Department of Neurology Medicine, Second Hospital of Shandong University, Jinan, 250033, China.
| |
Collapse
|
41
|
Haque ME, Kim IS, Jakaria M, Akther M, Choi DK. Importance of GPCR-Mediated Microglial Activation in Alzheimer's Disease. Front Cell Neurosci 2018; 12:258. [PMID: 30186116 PMCID: PMC6110855 DOI: 10.3389/fncel.2018.00258] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/30/2018] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder associated with impairment of cognition, memory deficits and behavioral abnormalities. Accumulation of amyloid beta (Aβ) is a characteristic hallmark of AD. Microglia express several GPCRs, which, upon activation by modulators, mediate microglial activation and polarization phenotype. This GPCR-mediated microglial activation has both protective and detrimental effects. Microglial GPCRs are involved in amyloid precursor protein (APP) cleavage and Aβ generation. In addition, microglial GPCRs are featured in the regulation of Aβ degradation and clearance through microglial phagocytosis and chemotaxis. Moreover, in response to Aβ binding on microglial Aβ receptors, they can trigger multiple inflammatory pathways. However, there is still a lack of insight into the mechanistic link between GPCR-mediated microglial activation and its pathological consequences in AD. Currently, the available drugs for the treatment of AD are mostly symptomatic and dominated by acetylcholinesterase inhibitors (AchEI). The selection of a specific microglial GPCR that is highly expressed in the AD brain and capable of modulating AD progression through Aβ generation, degradation and clearance will be a potential source of therapeutic intervention. Here, we have highlighted the expression and distribution of various GPCRs connected to microglial activation in the AD brain and their potential to serve as therapeutic targets of AD.
Collapse
Affiliation(s)
- Md Ezazul Haque
- Department of Applied Life Science, Graduate School, Konkuk University, Chungju, South Korea
| | - In-Su Kim
- Department of Integrated Bioscience and Biotechnology, College of Biomedical and Health Science, Research Institute of Inflammatory Disease, Konkuk University, Chungju, South Korea
| | - Md Jakaria
- Department of Applied Life Science, Graduate School, Konkuk University, Chungju, South Korea
| | - Mahbuba Akther
- Department of Applied Life Science, Graduate School, Konkuk University, Chungju, South Korea
| | - Dong-Kug Choi
- Department of Applied Life Science, Graduate School, Konkuk University, Chungju, South Korea.,Department of Integrated Bioscience and Biotechnology, College of Biomedical and Health Science, Research Institute of Inflammatory Disease, Konkuk University, Chungju, South Korea
| |
Collapse
|
42
|
Agrawal M, Saraf S, Saraf S, Antimisiaris SG, Chougule MB, Shoyele SA, Alexander A. Nose-to-brain drug delivery: An update on clinical challenges and progress towards approval of anti-Alzheimer drugs. J Control Release 2018; 281:139-177. [DOI: 10.1016/j.jconrel.2018.05.011] [Citation(s) in RCA: 203] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 05/10/2018] [Accepted: 05/10/2018] [Indexed: 01/02/2023]
|
43
|
Insulin-signaling Pathway Regulates the Degradation of Amyloid β-protein via Astrocytes. Neuroscience 2018; 385:227-236. [PMID: 29932983 DOI: 10.1016/j.neuroscience.2018.06.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 06/09/2018] [Accepted: 06/11/2018] [Indexed: 01/06/2023]
Abstract
Alzheimer's disease (AD) has been considered as a metabolic dysfunction disease associated with impaired insulin signaling. Determining the mechanisms underlying insulin signaling dysfunction and resistance in AD will be important for its treatment. Impaired clearance of amyloid-β peptide (Aβ) significantly contributes to amyloid accumulation, which is typically observed in the brain of AD patients. Reduced expression of important Aβ-degrading enzymes in the brain, such as neprilysin (NEP) and insulin-degrading enzyme (IDE), can promote Aβ deposition in sporadic late-onset AD patients. Here, we investigated whether insulin regulates the degradation of Aβ by inducing expression of NEP and IDE in cultured astrocytes. Treatment of astrocytes with insulin significantly reduced cellular NEP levels, but increased IDE expression. The effects of insulin on the expression of NEP and IDE involved activation of an extracellular signal-regulated kinase (ERK)-mediated pathway. The reduction in cellular NEP levels was associated with NEP secretion into the culture medium, whereas IDE was increased in the cell membranes. Moreover, insulin-treated astrocytes significantly facilitated the degradation of exogenous Aβ within the culture medium. Interestingly, pretreatment of astrocytes with an ERK inhibitor prior to insulin exposure markedly inhibited insulin-induced degradation of Aβ. These results suggest that insulin exposure enhanced Aβ degradation via an increase in NEP secretion and IDE expression in astrocytes, via activation of the ERK-mediated pathway. The inhibition of insulin signaling pathways delayed Aβ degradation by attenuating alterations in NEP and IDE levels and competition with insulin and Aβ. Our results provide further insight into the pathological relevance of insulin resistance in AD development.
Collapse
|
44
|
Zhou MM, Ding L, Wen M, Che HX, Huang JQ, Zhang TT, Xue CH, Mao XZ, Wang YM. Mechanisms of DHA-enriched phospholipids in improving cognitive deficits in aged SAMP8 mice with high-fat diet. J Nutr Biochem 2018; 59:64-75. [PMID: 29986309 DOI: 10.1016/j.jnutbio.2018.05.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 04/15/2018] [Accepted: 05/16/2018] [Indexed: 01/29/2023]
Abstract
Recent studies have shown that a high-fat diet (HFD) is involved in both metabolic dysfunction and cognitive deficiency and that docosahexaenoic-acid-enriched phospholipids (DHA-PLs) have beneficial effects on obesity and cognitive impairment. However, there are only a few studies comparing differences between DHA-PC and DHA-PS in HFD-induced Alzheimer's disease (AD) models. After 8 weeks feeding with HFD, 10-month-old SAMP8 mice were fed with 1% (w/w) DHA-PC or 1% DHA-PS (biosynthesized from DHA-PC) for 8 weeks; we then tested the behavioral performances in the Barnes maze test and Morris maze test. The changes of the generation and accumulation of Aβ, oxidative stress, apoptosis, neuroinflammation and neurotrophic factors were also measured. The results indicated that both DHA-PC and DHA-PS significantly improved the metabolic disorders and cognitive deficits. Both DHA-PC and DHA-PS could ameliorate oxidative stress, and DHA-PS presented more notable benefits than DHA-PC on Aβ pathology, mitochondrial damage, neuroinflammation and neurotrophic factors; DHA-PS was for the first time found to increase the production of insoluble Aβ (less pathogenic) in this AD model. These data suggest that DHA-PLs can significantly improve cognitive deficiency, and the molecular mechanisms for this closely relate to the phospholipid polar groups.
Collapse
Affiliation(s)
- Miao-Miao Zhou
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao, Shandong Province 266003, PR China
| | - Lin Ding
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao, Shandong Province 266003, PR China
| | - Min Wen
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao, Shandong Province 266003, PR China
| | - Hong-Xia Che
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao, Shandong Province 266003, PR China
| | - Jia-Qi Huang
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao, Shandong Province 266003, PR China
| | - Tian-Tian Zhang
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao, Shandong Province 266003, PR China
| | - Chang-Hu Xue
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao, Shandong Province 266003, PR China; Qingdao National Laboratory for Marine Science and Technology, Laboratory of Marine Drugs & Biological products, Qingdao 266237, PR China
| | - Xiang-Zhao Mao
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao, Shandong Province 266003, PR China; Qingdao National Laboratory for Marine Science and Technology, Laboratory of Marine Drugs & Biological products, Qingdao 266237, PR China.
| | - Yu-Ming Wang
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao, Shandong Province 266003, PR China; Qingdao National Laboratory for Marine Science and Technology, Laboratory of Marine Drugs & Biological products, Qingdao 266237, PR China.
| |
Collapse
|
45
|
Perlman A, Hirsh Raccah B, Matok I, Muszkat M. Cognition- and Dementia-Related Adverse Effects With Sacubitril-Valsartan: Analysis of the FDA Adverse Event Report System Database. J Card Fail 2018; 24:533-536. [PMID: 29746915 DOI: 10.1016/j.cardfail.2018.04.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 04/24/2018] [Accepted: 04/29/2018] [Indexed: 10/17/2022]
Abstract
BACKGROUND Because neprilysin is involved in the degradation of amyloid-beta, there is concern that the angiotensin-neprilysin inhibitor sacubitril-valsartan could increase the risk for dementia. METHODS We analyzed adverse event cases submitted to the Food and Drug Administration Adverse Event Report System from July 2015 to March 2017. Cognition- and dementia-related adverse event cases were defined with the use of broad and narrow structured medical queries. RESULTS During the period evaluated, 9,004 adverse event reports (out of a total of 2,249,479) involved the use of sacubitril-valsartan. Based on the broad definition, sacubitril-valsartan was associated with cognition- and dementia-related adverse events in 459 reports (5.1%), but this was lower than the proportion of these reports among other medications (6.6%, reporting odds ratio [ROR] 0.72, 95% confidence interval [CI] 0.65-0.79). Restricting the comparison to cases with age >60 years and with the use of a comparator group with heart failure resulted in no association between sacubitril-valsartan and dementia-related adverse events, with the use of both the broad and the narrow definitions (ROR 0.87, 95% CI 0.76-1.02, and ROR 1.06, 95% CI 0.4-3.16, respectively). CONCLUSION Sacubitril-valsartan is not associated with a disproportionately high rate of short-term dementia-related adverse effect reports. Long-term studies assessing cognitive outcomes are required to better establish the medication's cognition effects.
Collapse
Affiliation(s)
- Amichai Perlman
- Department of Internal Medicine, Hadassah University Hospital, Jerusalem, Israel; Division of Clinical Pharmacy, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Bruria Hirsh Raccah
- Division of Clinical Pharmacy, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel; Department of Cardiology, Hadassah University Hospital, Jerusalem, Israel
| | - Ilan Matok
- Division of Clinical Pharmacy, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Mordechai Muszkat
- Department of Internal Medicine, Hadassah University Hospital, Jerusalem, Israel
| |
Collapse
|
46
|
Perlman A, Raccah BH, Matok I, Muszkat M. WITHDRAWN: Cognition and Dementia Related Adverse Effects with Sacubitril-Valsartan: Analysis of the FDA Adverse Event Report System Database. J Card Fail 2018:S1071-9164(18)30169-6. [PMID: 29746914 DOI: 10.1016/j.cardfail.2018.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 04/24/2018] [Accepted: 04/29/2018] [Indexed: 11/29/2022]
Abstract
The Publisher regrets that this article is an accidental duplication of an article that has already been published, http://dx.doi.org/10.1016/j.cardfail.2018.04.009. The duplicate article has therefore been withdrawn. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.
Collapse
Affiliation(s)
- Amichai Perlman
- Department of Internal Medicine, Hadassah University Hospital, Jerusalem, Israel; Division of Clinical Pharmacy, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Bruria Hirsh Raccah
- Department of Cardiology, Hadassah University Hospital, Jerusalem, Israel; Division of Clinical Pharmacy, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Ilan Matok
- Division of Clinical Pharmacy, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Mordechai Muszkat
- Department of Internal Medicine, Hadassah University Hospital, Jerusalem, Israel
| |
Collapse
|
47
|
Shah NN, Dogar MU, Shah PN, Ishtiaq S, Mathew S, Shah P, Ishtiaq A, Vittorio TJ. Impact of prolonged utilization of neprilysin inhibition on the cognitive function of heart failure patients. Ther Adv Cardiovasc Dis 2018. [PMID: 29529959 DOI: 10.1177/1753944718756563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Niel N Shah
- Smt. N.H.L. Municipal Medical College, Ahmedabad, Gujarat, India
| | | | - Parin N Shah
- B.J. Medical College, Civil Hospital, Ahmedabad, Gujarat, India
| | | | - Shawn Mathew
- New York Institute of Technology, Old Westbury, NY, USA
| | | | - Alia Ishtiaq
- SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Timothy J Vittorio
- Bronx-Lebanon Hospital Center, Department of Medicine/Division of Cardiology, 1650 Grand Concourse, Bronx, NY 10457, USA
| |
Collapse
|
48
|
Harris SA, Harris EA. Molecular Mechanisms for Herpes Simplex Virus Type 1 Pathogenesis in Alzheimer's Disease. Front Aging Neurosci 2018; 10:48. [PMID: 29559905 PMCID: PMC5845560 DOI: 10.3389/fnagi.2018.00048] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 02/12/2018] [Indexed: 12/12/2022] Open
Abstract
This review focuses on research in the areas of epidemiology, neuropathology, molecular biology and genetics that implicates herpes simplex virus type 1 (HSV-1) as a causative agent in the pathogenesis of sporadic Alzheimer’s disease (AD). Molecular mechanisms whereby HSV-1 induces AD-related pathophysiology and pathology, including neuronal production and accumulation of amyloid beta (Aβ), hyperphosphorylation of tau proteins, dysregulation of calcium homeostasis, and impaired autophagy, are discussed. HSV-1 causes additional AD pathologies through mechanisms that promote neuroinflammation, oxidative stress, mitochondrial damage, synaptic dysfunction, and neuronal apoptosis. The AD susceptibility genes apolipoprotein E (APOE), phosphatidylinositol binding clathrin assembly protein (PICALM), complement receptor 1 (CR1) and clusterin (CLU) are involved in the HSV lifecycle. Polymorphisms in these genes may affect brain susceptibility to HSV-1 infection. APOE, for example, influences susceptibility to certain viral infections, HSV-1 viral load in the brain, and the innate immune response. The AD susceptibility gene cholesterol 25-hydroxylase (CH25H) is upregulated in the AD brain and is involved in the antiviral immune response. HSV-1 interacts with additional genes to affect cognition-related pathways and key enzymes involved in Aβ production, Aβ clearance, and hyperphosphorylation of tau proteins. Aβ itself functions as an antimicrobial peptide (AMP) against various pathogens including HSV-1. Evidence is presented supporting the hypothesis that Aβ is produced as an AMP in response to HSV-1 and other brain infections, leading to Aβ deposition and plaque formation in AD. Epidemiologic studies associating HSV-1 infection with AD and cognitive impairment are discussed. Studies are reviewed supporting subclinical chronic reactivation of latent HSV-1 in the brain as significant in the pathogenesis of AD. Finally, the rationale for and importance of clinical trials treating HSV-1-infected MCI and AD patients with antiviral medication is discussed.
Collapse
Affiliation(s)
- Steven A Harris
- St. Vincent Medical Group, Northside Internal Medicine, Indianapolis, IN, United States
| | - Elizabeth A Harris
- Department of Neurology, University of Chicago Medical Center, Chicago, IL, United States
| |
Collapse
|
49
|
Lin CY, Cheng YS, Liao TY, Lin C, Chen ZT, Twu WI, Chang CW, Tan DTW, Liu RS, Tu PH, Chen RPY. Intranasal Administration of a Polyethylenimine-Conjugated Scavenger Peptide Reduces Amyloid-β Accumulation in a Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2018; 53:1053-67. [PMID: 27340844 DOI: 10.3233/jad-151024] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Amyloid-β (Aβ) aggregation in the brain plays a central and initiatory role in pathogenesis and/or progression of Alzheimer's disease (AD). Inhibiting Aβ aggregation is a potential strategy in the prevention of AD. A scavenger peptide, V24P(10-40), designed to decrease Aβ accumulation in the brain, was conjugated to polyethylenimine (PEI) and tested as a preventive/therapeutic strategy for AD in this study. This PEI-conjugated V24P(10-40) peptide was delivered intranasally, as nasal drops, to four-month-old APP/PS1 double transgenic mice for four or eight months. Compared with control values, peptide treatment for four months significantly reduced the amount of GdnHCl-extracted Aβ40 and Aβ42 in the mice's hippocampus and cortex. After treatment for eight months, amyloid load, as quantified by Pittsburgh compound B microPET imaging, was significantly decreased in the mice's hippocampus, cortex, amygdala, and olfactory bulb. Our data suggest that this intranasally delivered scavenger peptide is effective in decreasing Aβ accumulation in the brain of AD transgenic mice. Nasal application of peptide drops is easy to use and could be further developed to prevent and treat AD.
Collapse
Affiliation(s)
- Chih-Yun Lin
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan.,Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Yu-Sung Cheng
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Tai-Yan Liao
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Chen Lin
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Zih-Ten Chen
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Woan-Ing Twu
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Chi-Wei Chang
- Biomedical Imaging Research Center, Department of Nuclear Medicine, National Yang Ming University and Taipei Veterans General Hospital, Taipei, Taiwan
| | - David Tat-Wei Tan
- Biomedical Imaging Research Center, Department of Nuclear Medicine, National Yang Ming University and Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ren-Shyan Liu
- Biomedical Imaging Research Center, Department of Nuclear Medicine, National Yang Ming University and Taipei Veterans General Hospital, Taipei, Taiwan.,Molecular and Genetic Imaging Core, Taiwan Mouse Clinic, Academia Sinica, Taipei, Taiwan
| | - Pang-Hsien Tu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Rita P-Y Chen
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan.,Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
50
|
Kimura J, Shimizu K, Kajima K, Yokosuka A, Mimaki Y, Oku N, Ohizumi Y. Nobiletin Reduces Intracellular and Extracellular β-Amyloid in iPS Cell-Derived Alzheimer’s Disease Model Neurons. Biol Pharm Bull 2018; 41:451-457. [DOI: 10.1248/bpb.b17-00364] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Junko Kimura
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka
| | - Kosuke Shimizu
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka
| | | | - Akihito Yokosuka
- Department of Medicinal Pharmacognosy, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | - Yoshihiro Mimaki
- Department of Medicinal Pharmacognosy, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | - Naoto Oku
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka
| | - Yasushi Ohizumi
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka
- Kansei Fukushi Research Institute, Tohoku Fukushi University
| |
Collapse
|