1
|
Mirehei M, Motamedi F, Maghsoudi N, Mansouri Z, Naderi S, Khodagholi F, Abbaszadeh F. Effects of Bufexamac, a class IIb HDAC inhibitor, on behavior and neuropathological features in an Aβ-induced rat model of Alzheimer's disease. Exp Gerontol 2025; 204:112746. [PMID: 40185252 DOI: 10.1016/j.exger.2025.112746] [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: 01/26/2025] [Revised: 03/24/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
It has been suggested that Alzheimer's disease (AD), a progressive neurological condition, can potentially be treated through epigenetic means by targeting histone deacetylases (HDACs), enzymes that regulate gene expression. In this study, we investigated the molecular mechanisms of Bufexamac, in an animal model of AD. Bufexamac specifically targets Class IIb HDACs, which are particularly relevant in the context of neuroinflammation and neurodegeneration. This selectivity may reduce off-target effects commonly associated with broader-spectrum HDAC inhibitors, such as pan-HDAC inhibitors, which can affect multiple HDAC classes and potentially lead to undesirable side effects. Male rats injected with Aβ25-35 for AD-like symptoms were treated with 20 μg/rat Bufexamac for 8 days. Cognitive function, depression, and anxiety were assessed through behavioral tests, while Western blotting, H&E staining, and ELISA were used to detect protein expression, morphological changes, and enzyme activity. Bufexamac treatment markedly improved cognitive and behavioral impairments in Aβ-injected rats and regulated the key proteins related to neuroinflammation (GFAP, Iba1), histone, and α-tubulin acetylation. Simultaneously, it decreased the expression of proteins in the stromal interaction molecule (STIM) pathway. Furthermore, Bufexamac lowered the activity of monoamine oxidase enzymes, elevated the count of healthy neurons, and ameliorated neuronal structure in the hippocampus. Overall, these findings suggest that Bufexamac could be a more targeted therapy for AD than other non-selective HDAC inhibitors, which often have diverse functions and potential side effects. Bufexamac enhances cognitive function and alleviates depression and anxiety by regulating proteins related to neuroinflammation, histone, and α-tubulin acetylation, as well as modulating STIM levels and MAO activity.
Collapse
Affiliation(s)
- Monireh Mirehei
- Neuroscience Research Center, Institute of Neuroscience and Cognition, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fereshteh Motamedi
- Neuroscience Research Center, Institute of Neuroscience and Cognition, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nader Maghsoudi
- Neuroscience Research Center, Institute of Neuroscience and Cognition, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Mansouri
- Neuroscience Research Center, Institute of Neuroscience and Cognition, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudabeh Naderi
- Neuroscience Research Center, Institute of Neuroscience and Cognition, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Institute of Neuroscience and Cognition, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Fatemeh Abbaszadeh
- Neurobiology Research Center, Institute of Neuroscience and Cognition, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Timofeeva AM, Aulova KS, Nevinsky GA. Modeling Alzheimer's Disease: A Review of Gene-Modified and Induced Animal Models, Complex Cell Culture Models, and Computational Modeling. Brain Sci 2025; 15:486. [PMID: 40426657 PMCID: PMC12109626 DOI: 10.3390/brainsci15050486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Revised: 04/30/2025] [Accepted: 05/03/2025] [Indexed: 05/29/2025] Open
Abstract
Alzheimer's disease, a complex neurodegenerative disease, is characterized by the pathological aggregation of insoluble amyloid β and hyperphosphorylated tau. Multiple models of this disease have been employed to investigate the etiology, pathogenesis, and multifactorial aspects of Alzheimer's disease and facilitate therapeutic development. Mammals, especially mice, are the most common models for studying the pathogenesis of this disease in vivo. To date, the scientific literature has documented more than 280 mouse models exhibiting diverse aspects of Alzheimer's disease pathogenesis. Other mammalian species, including rats, pigs, and primates, have also been utilized as models. Selected aspects of Alzheimer's disease have also been modeled in simpler model organisms, such as Drosophila melanogaster, Caenorhabditis elegans, and Danio rerio. It is possible to model Alzheimer's disease not only by creating genetically modified animal lines but also by inducing symptoms of this neurodegenerative disease. This review discusses the main methods of creating induced models, with a particular focus on modeling Alzheimer's disease on cell cultures. Induced pluripotent stem cell (iPSC) technology has facilitated novel investigations into the mechanistic underpinnings of diverse diseases, including Alzheimer's. Progress in culturing brain tissue allows for more personalized studies on how drugs affect the brain. Recent years have witnessed substantial advancements in intricate cellular system development, including spheroids, three-dimensional scaffolds, and microfluidic cultures. Microfluidic technologies have emerged as cutting-edge tools for studying intercellular interactions, the tissue microenvironment, and the role of the blood-brain barrier (BBB). Modern biology is experiencing a significant paradigm shift towards utilizing big data and omics technologies. Computational modeling represents a powerful methodology for researching a wide array of human diseases, including Alzheimer's. Bioinformatic methodologies facilitate the analysis of extensive datasets generated via high-throughput experimentation. It is imperative to underscore the significance of integrating diverse modeling techniques in elucidating pathogenic mechanisms in their entirety.
Collapse
Affiliation(s)
- Anna M. Timofeeva
- SB RAS Institute of Chemical Biology and Fundamental Medicine, Novosibirsk 630090, Russia
| | | | | |
Collapse
|
3
|
Canet G, Zussy C, Vitalis M, Morin F, Chevallier N, Hunt H, Claeysen S, Blaquière M, Marchi N, Planel E, Meijer OC, Desrumaux C, Givalois L. Advancing Alzheimer's disease pharmacotherapy: efficacy of glucocorticoid modulation with dazucorilant (CORT113176) in preclinical mouse models. Br J Pharmacol 2025; 182:1930-1956. [PMID: 39891319 DOI: 10.1111/bph.17457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/08/2024] [Accepted: 11/24/2024] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND AND PURPOSE Exposure to chronic stress and high levels of glucocorticoid hormones in adulthood has been associated with cognitive deficits and increased risk of Alzheimer's disease (AD). Dazucorilant has recently emerged as a selective glucocorticoid receptor (NR3C1) modulator, exhibiting efficacy in counteracting amyloid-β toxicity in an acute model of AD. We aim to assess the therapeutic potential of dazucorilant in reversing amyloid and tau pathologies through the inhibition of glucocorticoid receptor pathological activity, and providing additional evidence for its consideration in AD treatment. EXPERIMENTAL APPROACH The efficacy of dazucorilant was evaluated in two transgenic mouse models of amyloid pathology. The slowly progressing J20 and the aggressively pathological 5xFAD mice. Behavioural analysis was conducted to evaluate welfare, cognitive performances and anxiety levels. The activity of the glucocorticoid receptor system, neuroinflammation, amyloid burden and tau phosphorylation were examined in hippocampi. KEY RESULTS In both AD models, chronic treatment with dazucorilant improved working and long-term spatial memories along with the inhibition of glucocorticoid receptor-dependent pathogenic processes and the normalization of plasma glucocorticoid levels. Dazucorilant treatment also resulted in a reduction in tau hyperphosphorylation and amyloid production and aggregation. Additionally, dazucorilant seemed to mediate a specific re-localization of activated glial cells onto amyloid plaques in J20 mice, suggesting a restoration of physiological neuroinflammatory processes. CONCLUSION AND IMPLICATIONS Dazucorilant exhibited sustained disease-modifying effects in two AD models. Given that this compound has demonstrated safety and tolerability in human subjects, our results provide pre-clinical support for conducting clinical trials to evaluate its potential in AD.
Collapse
Affiliation(s)
- Geoffrey Canet
- MMDN, Univ Montpellier, EPHE-PSL, INSERM, Montpellier, France
- Faculty of Medicine, Department of Psychiatry and Neurosciences, CR-CHUQ, Laval University, Québec City, Quebec, Canada
| | - Charleine Zussy
- MMDN, Univ Montpellier, EPHE-PSL, INSERM, Montpellier, France
| | - Mathieu Vitalis
- MMDN, Univ Montpellier, EPHE-PSL, INSERM, Montpellier, France
| | - Françoise Morin
- Faculty of Medicine, Department of Psychiatry and Neurosciences, CR-CHUQ, Laval University, Québec City, Quebec, Canada
| | | | - Hazel Hunt
- Corcept Therapeutics, Menlo Park, California, USA
| | | | | | - Nicola Marchi
- IGF, Univ Montpellier, CNRS, INSERM, Montpellier, France
| | - Emmanuel Planel
- Faculty of Medicine, Department of Psychiatry and Neurosciences, CR-CHUQ, Laval University, Québec City, Quebec, Canada
| | - Onno C Meijer
- Einthoven Laboratory, Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
| | - Catherine Desrumaux
- MMDN, Univ Montpellier, EPHE-PSL, INSERM, Montpellier, France
- LIPSTIC LabEx, Dijon, France
| | - Laurent Givalois
- MMDN, Univ Montpellier, EPHE-PSL, INSERM, Montpellier, France
- Faculty of Medicine, Department of Psychiatry and Neurosciences, CR-CHUQ, Laval University, Québec City, Quebec, Canada
- CNRS, Paris, France
| |
Collapse
|
4
|
Yenkoyan KB, Kotova MM, Apukhtin KV, Galstyan DS, Amstislavskaya TG, Strekalova T, de Abreu MS, Chavushyan VA, Lim LW, Yang L, Rosemberg DD, Kalueff AV. Experimental modeling of Alzheimer's disease: Translational lessons from cross-taxon analyses. Alzheimers Dement 2025; 21:e70273. [PMID: 40420360 PMCID: PMC12106051 DOI: 10.1002/alz.70273] [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: 12/05/2024] [Revised: 04/02/2025] [Accepted: 04/21/2025] [Indexed: 05/28/2025]
Abstract
Alzheimer's disease (AD) is a severely debilitating neurodegenerative disease with a rapidly increasing global prevalence, poorly understood causes, and no efficient treatments. Experimental models are valuable for studying AD pathogenesis, including amyloid beta and tau accumulation, synaptic dysfunction, and neuroinflammation. While no model fully reproduces the disease, we take an evolutionary biology approach to discuss available models across taxa, from mammals (rodents, primates) to zebrafish, Drosophila melanogaster, and Caenorhabditis elegans. Evaluating their strengths and limitations provides insight into disease mechanisms and may refine research strategies for improved diagnostics and therapeutic screening. Traditional models have significantly contributed to AD research, yet their translational limitations highlight the need for physiologically relevant alternatives. Integrating humanized rodent models, zebrafish, organoids, and induced pluripotent stem cell-based systems-along with advances in bioengineering and genetic editing-may offer a more comprehensive framework to bridge the gap between preclinical research and clinical application. HIGHLIGHTS: Experimental models across rodents, primates, zebrafish, fruit flies, and worms provide key insights into Alzheimer's disease (AD). Cross-taxon comparisons assess strengths and weaknesses in AD models. Evolutionary biology approaches refine experimental strategies for AD research. Diverse animal models improve understanding of AD pathogenesis. Cross-species models enhance diagnostics and therapeutic strategy development.
Collapse
Affiliation(s)
- Konstantin B. Yenkoyan
- Neuroscience Laboratory, COBRAIN CenterYerevan State Medical University after M. HeratsiYerevanArmenia
| | - Maria M. Kotova
- Neuroscience ProgramSirius University of Science and TechnologySochiRussia
| | - Kirill V. Apukhtin
- Neuroscience ProgramSirius University of Science and TechnologySochiRussia
| | - David S. Galstyan
- Institute of Translational BiomedicineSt. Petersburg State UniversitySt. PetersburgRussia
- Institute of Experimental Medicine, Almazov National Medical Research CentreMinistry of Healthcare of Russian FederationSt. PetersburgRussia
| | | | | | - Murilo S. de Abreu
- Graduate Program in Health SciencesFederal University of Health Sciences of Porto AlegrePorto AlegreBrazil
| | - Vergine A. Chavushyan
- Neuroscience Laboratory, COBRAIN CenterYerevan State Medical University after M. HeratsiYerevanArmenia
| | - Lee Wei Lim
- Department of Biosciences and Bioinformatics, School of ScienceXi'an Jiaotong‐Liverpool UniversitySuzhouChina
- Suzhou Municipal Key Laboratory of Neurobiology and Cell Signaling, School of ScienceXi'an Jiaotong‐Liverpool UniversitySuzhouChina
| | - Longen Yang
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences CenterFederal University of Santa MariaSanta MariaBrazil
- Graduate Program in Biological Sciences: Toxicological BiochemistryFederal University of Santa MariaSanta MariaBrazil
| | - Denis D. Rosemberg
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences CenterFederal University of Santa MariaSanta MariaBrazil
- Graduate Program in Biological Sciences: Toxicological BiochemistryFederal University of Santa MariaSanta MariaBrazil
- The International Zebrafish Neuroscience Research Consortium (ZNRC)SlidellLouisianaUSA
| | - Allan V. Kalueff
- Suzhou Municipal Key Laboratory of Neurobiology and Cell Signaling, School of ScienceXi'an Jiaotong‐Liverpool UniversitySuzhouChina
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences CenterFederal University of Santa MariaSanta MariaBrazil
- The International Zebrafish Neuroscience Research Consortium (ZNRC)SlidellLouisianaUSA
| |
Collapse
|
5
|
Niyonzima YB, Asato Y, Murakami T, Kadokawa H. Alpha-synuclein expression in GnRH neurons of young and old bovine hypothalami. Reprod Fertil Dev 2024; 36:RD24033. [PMID: 39283977 DOI: 10.1071/rd24033] [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: 02/29/2024] [Accepted: 08/15/2024] [Indexed: 11/13/2024] Open
Abstract
Context Understanding of central nervous system mechanisms related to age-related infertility remains limited. Fibril α-synuclein, distinct from its monomer form, is implicated in age-related diseases and propagates among neurons akin to prions. Aims We compared α-synuclein expression in gonadotropin-releasing hormone-expressing neurons (GnRH neurons) in the pre-optic area, arcuate nucleus, and median eminence of healthy heifers and aged cows to determine its role in age-related infertility. Methods We analysed mRNA and protein expression, along with fluorescent immunohistochemistry for GnRH and α-synuclein, followed by Congo red staining to detect amyloid deposits, and confocal microscopy. Key results Both mRNA and protein expressions of α-synuclein were confirmed by reverse transcription-polymerase chain reaction (RT-PCR) and western blots in bovine cortex, hippocampus, and anterior and posterior hypothalamus tissues. Significant differences in α-synuclein mRNA expression were observed in the cortex and hippocampus between young and old cows. Western blots showed five bands of α-synuclein, probably reflecting monomer, dimer, and oligomers, in the cortex, hippocampus, hypothalamus tissues, and there were significant differences in some bands between young and old cows. Bright-field and polarised light microscopy did not detect obvious amyloid deposition in aged hypothalami; however, higher-sensitive confocal microscopy unveiled strong positive signal of Congo red and α-synuclein in GnRH neurons in aged hypothalami. Additionally, α-synuclein expression was detected in immortalised GnRH neurons, GT1-7 cells. Conclusion Alpha-synuclein was expressed in GnRH neurons, and some differences were observed between young and old hypothalami. Implications Alpha-synuclein may play an important role in aging-related infertility.
Collapse
Affiliation(s)
- Yvan Bienvenu Niyonzima
- Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi-shi, Yamaguchi-ken 1677-1, Japan
| | - Yuuki Asato
- Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi-shi, Yamaguchi-ken 1677-1, Japan
| | - Tomoaki Murakami
- Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Hiroya Kadokawa
- Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi-shi, Yamaguchi-ken 1677-1, Japan
| |
Collapse
|
6
|
Carrazoni GS, Garces NB, Cadore CR, Sosa PM, Cattaneo R, Mello-Carpes PB. Supplementation with Manihot esculenta Crantz (Cassava) leaves' extract prevents recognition memory deficits and hippocampal antioxidant dysfunction induced by Amyloid-β. Nutr Neurosci 2024; 27:942-950. [PMID: 37948133 DOI: 10.1080/1028415x.2023.2280815] [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] [Indexed: 11/12/2023]
Abstract
OBJECTIVE The Manihot esculenta Crantz (Cassava) is a typical South American plant rich in nutrients and energetic compounds. Lately, our group has shown that non-pharmacological interventions with natural antioxidants present different neuroprotective effects on oxidative balance and memory deficits in AD-like animal models. Here, our objective was to evaluate the neuroprotective effects of Cassava leaves' extract (CAS) in an AD-like model induced by amyloid-beta (Aβ) 25-35 peptide. METHODS Male Wistar rats (n = 40; 60 days old) were subjected to 10 days of CAS supplementation; then, we injected 2 μL Aβ 25-35 in the hippocampus by stereotaxic surgery. Ten days later, we evaluated object recognition (OR) memory. Cassavas' total polyphenols, flavonoids, and condensed tannins content were measured, as well as hippocampal lipid peroxidation and total antioxidant capacity. RESULTS CAS protected against Aβ-induced OR memory deficits. In addition, Aβ promoted antioxidant capacity decrease, while CAS was able to prevent it, in addition to diminishing lipoperoxidation compared to Aβ. DISCUSSION We show that treatment with Cassava leaves' extract before AD induction prevents recognition memory deficits related to Aβ hippocampal injection. At least part of these effects can be related to the Cassava leaves' extract supplementation effects on diminishing lipid peroxidation and preventing a decrease in the hippocampal total antioxidant capacity in the hippocampus of AD-like animals without adverse effects. Once cassavais a plant of warm and dry ground that can adapt to growon various soil types and seems to resist several insects, our results enable Cassava to be considered asa potential preventive intervention to avoid or minimizeAD-induced memory deficits worldwide.
Collapse
Affiliation(s)
- Guilherme Salgado Carrazoni
- Physiology Research Group, Stress, Memory and Behavior Lab, Universidade Federal do Pampa, Uruguaiana, Brazil
| | | | - Caroline Ramires Cadore
- Physiology Research Group, Stress, Memory and Behavior Lab, Universidade Federal do Pampa, Uruguaiana, Brazil
| | - Priscila Marques Sosa
- Physiology Research Group, Stress, Memory and Behavior Lab, Universidade Federal do Pampa, Uruguaiana, Brazil
| | | | - Pâmela Billig Mello-Carpes
- Physiology Research Group, Stress, Memory and Behavior Lab, Universidade Federal do Pampa, Uruguaiana, Brazil
| |
Collapse
|
7
|
Niyonzima YB, Asato Y, Kadokawa H. Alpha-synuclein expression in oxytocin neurons of young and old bovine brains. J Reprod Dev 2024; 70:213-222. [PMID: 38684411 PMCID: PMC11310384 DOI: 10.1262/jrd.2024-020] [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: 02/29/2024] [Accepted: 04/10/2024] [Indexed: 05/02/2024] Open
Abstract
Understanding of central nervous system mechanisms underlying age-related infertility remains limited. Fibril α-synuclein, distinct from its monomeric form, is implicated in age-related diseases. Notably, fibril α-synuclein spreads among neurons, similar to prions, from damaged old neurons in cortex and hippocampus to healthy neurons. However, less is known whether α-synuclein propagates into oxytocin neurons, which play crucial roles in reproduction. We compared α-synuclein expression in the oxytocin neurons in suprachiasmatic nucleus (SCN), supraoptic nucleus (SON), paraventricular hypothalamic nucleus (PVN), and posterior pituitary (PP) gland of healthy heifers and aged cows to determine its role in age-related infertility. We analyzed mRNA and protein expression, along with Congo red histochemistry and fluorescent immunohistochemistry for oxytocin and α-synuclein, followed by confocal microscopy with Congo red staining. Both mRNA and protein expressions of α-synuclein were confirmed in the bovine cortex, hippocampus, SCN, SON, PVN, and PP tissues. Significant differences in α-synuclein mRNA expressions were observed in the cortex and hippocampus between young heifers and old cows. Western blots showed five bands of α-synuclein, probably reflecting monomers, dimers, and oligomers, in the cortex, hippocampus, SCN, SON, PVN, and PP tissues, and there were significant differences in some bands between the young heifers and old cows. Bright-field and polarized light microscopy did not detect obvious amyloid deposition in the aged hypothalami; however, higher-sensitive confocal microscopy unveiled strong positive signals for Congo red and α-synuclein in oxytocin neurons in the aged hypothalami. α-synuclein was expressed in oxytocin neurons, and some differences were observed between young and old hypothalami.
Collapse
Affiliation(s)
| | - Yuuki Asato
- Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi 753-8515, Japan
| | - Hiroya Kadokawa
- Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi 753-8515, Japan
| |
Collapse
|
8
|
Ramírez Hernández E, Hernández Zimbrón LF, Segura Pérez E, Sánchez Salgado JL, Pereyra Morales MA, Zenteno E. Galectin-9 and Tim-3 are upregulated in response to microglial activation induced by the peptide Amyloid-β (25-35). Neuropeptides 2024; 105:102426. [PMID: 38527407 DOI: 10.1016/j.npep.2024.102426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/17/2024] [Accepted: 03/19/2024] [Indexed: 03/27/2024]
Abstract
Galectins are a group of β-galactoside-binding lectins associated with regulating immunological response. In the brains of AD patients and 5xFAD (familial AD) mice, galectin-3 (Gal-3) was highly upregulated and found to be expressed in microglia associated with Aβ plaques. However, the participation of other galectins, specifically galectin-9 (Gal-9) and T-cell immunoglobulin and mucin domain 3 (Tim-3) receptors, are unknown in the inflammatory response. The experimental model of the Aβ25-35 peptide will allow us to study the mechanisms of neuroinflammation and describe the changes in the expression of the Gal-9 and Tim-3 receptor. This study aimed to evaluate whether Aβ25-35 peptide administration into the lateral ventricles of rats upregulated Gal-9 and Tim-3 implicated in the modulation of neuroinflammation. The vehicle or Aβ25-35 peptide (1 μg/μL) was bilaterally administered into the lateral ventricles of the rat, and control group. After the administration of the Aβ25-35 peptide, animals were tested for learning (day 29) and spatial memory (day 30) in the novel object recognition test (NOR). On day 31, hippocampus was examined for morphological changes by Nilss stain, biochemical changes by NO2 and MDA, immunohistochemical analysis by astrocytes (GFAP), microglia (Iba1), Gal-9 and Tim-3, and western blot. Our results show the administration of the Aβ25-35 peptide into the lateral ventricles of rats induce memory impairment in the NOR by increases the oxidative stress and inflammatory response. This result is associated with an upregulation of Gal-9 and Tim-3 predominantly detected in the microglia cells of Aβ25-35-treated rats with respect to the control group. Gal-9 and Tim-3 are upregulated in activated microglia that could modulate the inflammatory response and damage in neurodegenerative processes induced by the Aβ25-35 peptide. Therefore, we suggest that Gal-9 and Tim-3 participate in the inflammatory process induced by the administration of the Aβ25-35 peptide.
Collapse
Affiliation(s)
- Eleazar Ramírez Hernández
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico.
| | | | - Emmanuel Segura Pérez
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - José Luis Sánchez Salgado
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Mohamed Ali Pereyra Morales
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Edgar Zenteno
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
9
|
Halder T, Saha B, Dhas N, Acharya S, Acharya N. Development and evaluation of multi-functionalized sialic acid conjugated asiatic acid nanoconstruct to mitigate cognitive deficits in Alzheimer's disease. Drug Dev Res 2024; 85:e22146. [PMID: 38349270 DOI: 10.1002/ddr.22146] [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: 09/18/2023] [Revised: 12/06/2023] [Accepted: 01/03/2024] [Indexed: 02/15/2024]
Abstract
Sialic acid (SA) serves a critical role in neuronal repair and cognitive functions. SA is a nine-carbon carboxylated sugar with a glycoconjugate cap that acts as a ligand and surface decoration with SA facilitates delivery to the target site. The present research aimed to develop SA surface modified AA nanostructured lipid carrier (NLCs) with carbodiimide conjugation method. Sterylamine, poloxamer 188 and tween 80 were used as surfactants and several characterization studies including, differential scanning calorimetry, fourier transform infrared spectroscopy and x-ray photon spectroscopy were analyzed. Further, in vitro, neuroprotective efficiency was evaluated in SH-SY5Y cells and hCMEC/D3 cells and found significant potential effects with the treatments of developed NLCs. Pharmacodynamics studies were also assessed in beta-amyloid-injected rats following quantification of Alzheimer's disease (AD) hallmarks like, Aβ(1-42), tau-protein, glycogen synthase kinase-3β levels, interleukin-6 and tumor necrosis factor-α for neuroinflammatory responses. Characterization studies revealed the conjugation on developed NLCs. The in vitro and in vivo results showed significant effects of SA decorated NLCs in reversing the damage by toxicant which was further characterized by the levels of neurotransmitters like acetylcholinesterase, butyrylcholinesterase. The results revealed significant (p < .05) refurbishment of cholinergic functions after 28 days of treatment of developed NLCs. These preclinical findings support the use of SA as a ligand to deliver the AA at targeted site as well as to mitigate the cognitive deficits in AD.
Collapse
Affiliation(s)
- Tripti Halder
- Department of Pharmacognosy, Nirma University, Ahmedabad, Gujarat, India
- Faculty of Pharmacy, School of Pharmaceutical and Population Health Informatics, DIT University, Dehradun, Uttarakhand, India
| | - Bijit Saha
- Department of Research and Development, Jodas Expoim Pvt Ltd, Kukatpally, Hyderabad, Telangana, India
| | - Namdev Dhas
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Sanjeev Acharya
- Faculty of Pharmacy, Institute of Pharmacy, Ganpat University, Kherva, Gujarat, India
| | - Niyati Acharya
- Department of Pharmacognosy, Nirma University, Ahmedabad, Gujarat, India
| |
Collapse
|
10
|
Grychowska K, López-Sánchez U, Vitalis M, Canet G, Satała G, Olejarz-Maciej A, Gołębiowska J, Kurczab R, Pietruś W, Kubacka M, Moreau C, Walczak M, Blicharz-Futera K, Bento O, Bantreil X, Subra G, Bojarski AJ, Lamaty F, Becamel C, Zussy C, Chaumont-Dubel S, Popik P, Nury H, Marin P, Givalois L, Zajdel P. Superiority of the Triple-Acting 5-HT 6R/5-HT 3R Antagonist and MAO-B Reversible Inhibitor PZ-1922 over 5-HT 6R Antagonist Intepirdine in Alleviation of Cognitive Deficits in Rats. J Med Chem 2023; 66:14928-14947. [PMID: 37797083 PMCID: PMC10641814 DOI: 10.1021/acs.jmedchem.3c01482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Indexed: 10/07/2023]
Abstract
The multifactorial origin and neurochemistry of Alzheimer's disease (AD) call for the development of multitarget treatment strategies. We report a first-in-class triple acting compound that targets serotonin type 6 and 3 receptors (5-HT-Rs) and monoamine oxidase type B (MAO-B) as an approach for treating AD. The key structural features required for MAO-B inhibition and 5-HT6R antagonism and interaction with 5-HT3R were determined using molecular dynamic simulations and cryo-electron microscopy, respectively. Bioavailable PZ-1922 reversed scopolamine-induced cognitive deficits in the novel object recognition test. Furthermore, it displayed superior pro-cognitive properties compared to intepirdine (a 5-HT6R antagonist) in the AD model, which involved intracerebroventricular injection of an oligomeric solution of amyloid-β peptide (oAβ) in the T-maze test in rats. PZ-1922, but not intepirdine, restored levels of biomarkers characteristic of the debilitating effects of oAβ. These data support the potential of a multitarget approach involving the joint modulation of 5-HT6R/5-HT3R/MAO-B in AD.
Collapse
Affiliation(s)
- Katarzyna Grychowska
- Faculty
of Pharmacy, Jagiellonian University Medical
College, 9 Medyczna Str., 30-688 Kraków, Poland
| | | | - Mathieu Vitalis
- Molecular
Mechanisms in Neurodegenerative Dementia (MMDN) Laboratory, University of Montpellier, EPHE-PSL, INSERM U1198, 34-095 Montpellier, France
| | - Geoffrey Canet
- Faculty
of Medicine, Laval University, CR-CHUQ, G1 V 4G2 Québec
City (QC), Canada
| | - Grzegorz Satała
- Maj
Institute of Pharmacology, Polish Academy
of Sciences, 12 Smętna Str., 31-324 Kraków, Poland
| | - Agnieszka Olejarz-Maciej
- Faculty
of Pharmacy, Jagiellonian University Medical
College, 9 Medyczna Str., 30-688 Kraków, Poland
| | - Joanna Gołębiowska
- Maj
Institute of Pharmacology, Polish Academy
of Sciences, 12 Smętna Str., 31-324 Kraków, Poland
| | - Rafał Kurczab
- Maj
Institute of Pharmacology, Polish Academy
of Sciences, 12 Smętna Str., 31-324 Kraków, Poland
| | - Wojciech Pietruś
- Maj
Institute of Pharmacology, Polish Academy
of Sciences, 12 Smętna Str., 31-324 Kraków, Poland
| | - Monika Kubacka
- Faculty
of Pharmacy, Jagiellonian University Medical
College, 9 Medyczna Str., 30-688 Kraków, Poland
| | | | - Maria Walczak
- Faculty
of Pharmacy, Jagiellonian University Medical
College, 9 Medyczna Str., 30-688 Kraków, Poland
| | - Klaudia Blicharz-Futera
- Faculty
of Pharmacy, Jagiellonian University Medical
College, 9 Medyczna Str., 30-688 Kraków, Poland
| | - Ophélie Bento
- IBMM,
Université
de Montpellier, CNRS, ENSCM, 34-293 Montpellier, France
- Institut
de Génomique Fonctionnelle, Université
de Montpellier, CNRS, INSERM, 34-094 Montpellier, France
| | - Xavier Bantreil
- IBMM,
Université
de Montpellier, CNRS, ENSCM, 34-293 Montpellier, France
| | - Gilles Subra
- IBMM,
Université
de Montpellier, CNRS, ENSCM, 34-293 Montpellier, France
| | - Andrzej J. Bojarski
- Maj
Institute of Pharmacology, Polish Academy
of Sciences, 12 Smętna Str., 31-324 Kraków, Poland
| | - Frédéric Lamaty
- IBMM,
Université
de Montpellier, CNRS, ENSCM, 34-293 Montpellier, France
| | - Carine Becamel
- Institut
de Génomique Fonctionnelle, Université
de Montpellier, CNRS, INSERM, 34-094 Montpellier, France
| | - Charleine Zussy
- Molecular
Mechanisms in Neurodegenerative Dementia (MMDN) Laboratory, University of Montpellier, EPHE-PSL, INSERM U1198, 34-095 Montpellier, France
| | - Séverine Chaumont-Dubel
- Institut
de Génomique Fonctionnelle, Université
de Montpellier, CNRS, INSERM, 34-094 Montpellier, France
| | - Piotr Popik
- Maj
Institute of Pharmacology, Polish Academy
of Sciences, 12 Smętna Str., 31-324 Kraków, Poland
| | - Hugues Nury
- Univ.
Grenoble Alpes, CNRS, CEA, IBS, F-38000 Grenoble, France
| | - Philippe Marin
- Institut
de Génomique Fonctionnelle, Université
de Montpellier, CNRS, INSERM, 34-094 Montpellier, France
| | - Laurent Givalois
- Molecular
Mechanisms in Neurodegenerative Dementia (MMDN) Laboratory, University of Montpellier, EPHE-PSL, INSERM U1198, 34-095 Montpellier, France
- Faculty
of Medicine, Laval University, CR-CHUQ, G1 V 4G2 Québec
City (QC), Canada
- CNRS, 75-016 Paris, France
| | - Paweł Zajdel
- Faculty
of Pharmacy, Jagiellonian University Medical
College, 9 Medyczna Str., 30-688 Kraków, Poland
| |
Collapse
|
11
|
Hector A, Provost C, Delignat-Lavaud B, Bouamira K, Menaouar CA, Mongrain V, Brouillette J. Hippocampal injections of soluble amyloid-beta oligomers alter electroencephalographic activity during wake and slow-wave sleep in rats. Alzheimers Res Ther 2023; 15:174. [PMID: 37833786 PMCID: PMC10571363 DOI: 10.1186/s13195-023-01316-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023]
Abstract
BACKGROUND Soluble amyloid-beta oligomers (Aβo) begin to accumulate in the human brain one to two decades before a clinical diagnosis of Alzheimer's disease (AD). The literature supports that soluble Aβo are implicated in synapse and neuronal losses in the brain regions such as the hippocampus. This region importantly contributes to explicit memory, the first type of memory affected in AD. During AD preclinical and prodromal stages, people are also experiencing wake/sleep alterations such as insomnia (e.g., difficulty initiating sleep, decreased sleep duration), excessive daytime sleepiness, and sleep schedule modifications. In addition, changes in electroencephalographic (EEG) activity during wake and sleep have been reported in AD patients and animal models. However, the specific contribution of Aβo to wake/sleep alterations is poorly understood and was investigated in the present study. METHODS Chronic hippocampal injections of soluble Aβo were conducted in male rats and combined with EEG recording to determine the progressive impact of Aβ pathology specifically on wake/sleep architecture and EEG activity. Bilateral injections were conducted for 6 consecutive days, and EEG acquisition was done before, during, and after Aβo injections. Immunohistochemistry was used to assess neuron numbers in the hippocampal dentate gyrus (DG). RESULTS Aβo injections did not affect the time spent in wakefulness, slow wave sleep (SWS), and paradoxical sleep but altered EEG activity during wake and SWS. More precisely, Aβo increased slow-wave activity (SWA; 0.5-5 Hz) and low-beta activity (16-20 Hz) during wake and decreased theta (5-9 Hz) and alpha (9-12 Hz) activities during SWS. Moreover, the theta activity/SWA ratio during wake and SWS was decreased by Aβo. These effects were significant only after 6 days of Aβo injections and were found with alterations in neuron counts in the DG. CONCLUSIONS We found multiple modifications of the wake and SWS EEG following Aβo delivery to the hippocampus. These findings expose a specific EEG signature of Aβ pathology and can serve the development of non-invasive and cost-effective markers for the early diagnosis of AD or other amyloid-related diseases.
Collapse
Affiliation(s)
- Audrey Hector
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Québec, Canada
- Center for Advanced Research in Sleep Medicine, CIUSSS-NIM, Montréal, Québec, Canada
- Centre de Recherche, Centre Hospitalier de l'Université de Montréal, Montréal, Québec, Canada
| | - Chloé Provost
- Center for Advanced Research in Sleep Medicine, CIUSSS-NIM, Montréal, Québec, Canada
| | - Benoît Delignat-Lavaud
- Center for Advanced Research in Sleep Medicine, CIUSSS-NIM, Montréal, Québec, Canada
- Centre de Recherche, Centre Hospitalier de l'Université de Montréal, Montréal, Québec, Canada
| | - Khadija Bouamira
- Center for Advanced Research in Sleep Medicine, CIUSSS-NIM, Montréal, Québec, Canada
| | | | - Valérie Mongrain
- Center for Advanced Research in Sleep Medicine, CIUSSS-NIM, Montréal, Québec, Canada.
- Centre de Recherche, Centre Hospitalier de l'Université de Montréal, Montréal, Québec, Canada.
- Department of Neuroscience, Université de Montréal, Montréal, Québec, Canada.
| | - Jonathan Brouillette
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Québec, Canada.
- Center for Advanced Research in Sleep Medicine, CIUSSS-NIM, Montréal, Québec, Canada.
| |
Collapse
|
12
|
Robles-Gómez ÁA, Ordaz B, Lorea-Hernández JJ, Peña-Ortega F. Deleterious and protective effects of epothilone-D alone and in the context of amyloid β- and tau-induced alterations. Front Mol Neurosci 2023; 16:1198299. [PMID: 37900942 PMCID: PMC10603193 DOI: 10.3389/fnmol.2023.1198299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 09/20/2023] [Indexed: 10/31/2023] Open
Abstract
Amyloid-β (Aβ) and hyperphosphorylated tau (P-tau) are Alzheimer's disease (AD) biomarkers that interact in a complex manner to induce most of the cognitive and brain alterations observed in this disease. Since the neuronal cytoskeleton is a common downstream pathological target of tau and Aβ, which mostly lead to augmented microtubule instability, the administration of microtubule stabilizing agents (MSAs) can protect against their pathological actions. However, the effectiveness of MSAs is still uncertain due to their state-dependent negative effects; thus, evaluating their specific actions in different pathological or physiological conditions is required. We evaluated whether epothilone-D (Epo-D), a clinically used MSA, rescues from the functional and behavioral alterations produced by intracerebroventricular injection of Aβ, the presence of P-tau, or their combination in rTg4510 mice. We also explored the side effects of Epo-D. To do so, we evaluated hippocampal-dependent spatial memory with the Hebb-Williams maze, hippocampal CA1 integrity and the intrinsic and synaptic properties of CA1 pyramidal neurons with the patch-clamp technique. Aβ and P-tau mildly impaired memory retrieval, but produced contrasting effects on intrinsic excitability. When Aβ and P-tau were combined, the alterations in excitability and spatial reversal learning (i.e., cognitive flexibility) were exacerbated. Interestingly, Epo-D prevented most of the impairments induced Aβ and P-tau alone and combined. However, Epo-D also exhibited some side effects depending on the prevailing pathological or physiological condition, which should be considered in future preclinical and translational studies. Although we did not perform extensive histopathological evaluations or measured microtubule stability, our findings show that MSAs can rescue the consequences of AD-like conditions but otherwise be harmful if administered at a prodromal stage of the disease.
Collapse
Affiliation(s)
- Ángel Abdiel Robles-Gómez
- Instituto de Neurobiología, UNAM Campus Juriquilla, Querétaro, Mexico
- Posgrado en Ciencias Biológicas, UNAM, Ciudad Universitaria, México City, Mexico
| | - Benito Ordaz
- Instituto de Neurobiología, UNAM Campus Juriquilla, Querétaro, Mexico
| | | | | |
Collapse
|
13
|
Choi GY, Kim HB, Hwang ES, Park HS, Cho JM, Ham YK, Kim JH, Mun MK, Maeng S, Park JH. Naringin enhances long-term potentiation and recovers learning and memory deficits of amyloid-beta induced Alzheimer's disease-like behavioral rat model. Neurotoxicology 2023; 95:35-45. [PMID: 36549596 DOI: 10.1016/j.neuro.2022.12.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/15/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022]
Abstract
Alzheimer's disease (AD), as the most typical type of dementia, is a chronic neurodegenerative disorder characterized by progressive learning and memory impairment. It is known that the main causes of AD are the accumulation of β-amyloid (Aβ) plaques and neurofibrillary tangles (NFT) containing hyperphosphorylated tau protein. Naringin is a flavonoid from citrus fruits, especially in grapefruit, which has anti-inflammatory, antioxidant, anti-apoptotic, and neuroprotective activities. However, the effect of naringin in AD caused by Aβ has not been clearly studied, and there are few studies on the electrophysiological aspect. Thus, we investigated the ex vivo neuroprotective effect of naringin through the long-term potentiation (LTP) on organotypic hippocampal slice cultures. We evaluated the in vivo effects of naringin (100 mg/kg/day) orally treated for 20 days on learning, memory, and cognition which was impaired by bilateral CA1 subregion injection of Aβ. Cognitive behaviors were measured 2 weeks after Aβ injection using behavioral tests and the hippocampal expression of apoptotic and neurotrophic regulators were measured by immunoblotting. In hippocampal tissue slices, naringin dose-dependently increased the field excitatory postsynaptic potential (fEPSP) after theta burst stimulation and attenuated Aβ-induced blockade of fEPSP in the hippocampal CA1 area. In Aβ injected rats, naringin improved object recognition memory in the novel object test, avoidance memory in the passive avoidance test and spatial recognition memory in the Morris water maze test. In the hippocampus, naringin attenuated the Aβ-induced cyclooxygenase-2, Bax activation and Bcl-2, CREB, BDNF and TrkB inhibition. These results suggest that naringin has therapeutic potential to reduce neuronal inflammation and apoptosis induced by Aβ related with the BDNF/TrkB/CREB signaling.
Collapse
Affiliation(s)
- Ga-Young Choi
- Department of East-West Medicine, Graduate School of East-West Medical Science, Kyung Hee University, Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Hyun-Bum Kim
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Eun-Sang Hwang
- Department of Gerontology, Graduate School of East-West Medical Science, Kyung Hee University, Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Ho-Sub Park
- Department of Gerontology, Graduate School of East-West Medical Science, Kyung Hee University, Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Jae-Min Cho
- Graduate School of Biotechnology, Kyung Hee University, Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Young-Ki Ham
- Department of East-West Medicine, Graduate School of East-West Medical Science, Kyung Hee University, Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Jin-Hee Kim
- Department of East-West Medicine, Graduate School of East-West Medical Science, Kyung Hee University, Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Mi-Kyung Mun
- Department of East-West Medicine, Graduate School of East-West Medical Science, Kyung Hee University, Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Sungho Maeng
- Department of Gerontology, Graduate School of East-West Medical Science, Kyung Hee University, Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do 17104, Republic of Korea.
| | - Ji-Ho Park
- Department of Gerontology, Graduate School of East-West Medical Science, Kyung Hee University, Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do 17104, Republic of Korea.
| |
Collapse
|
14
|
Canet G, Zussy C, Hernandez C, Maurice T, Desrumaux C, Givalois L. The pathomimetic oAβ25–35 model of Alzheimer's disease: Potential for screening of new therapeutic agents. Pharmacol Ther 2023; 245:108398. [PMID: 37001735 DOI: 10.1016/j.pharmthera.2023.108398] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia in the elderly, currently affecting more than 40 million people worldwide. The two main histopathological hallmarks of AD were identified in the 1980s: senile plaques (composed of aggregated amyloid-β (Aβ) peptides) and neurofibrillary tangles (composed of hyperphosphorylated tau protein). In the human brain, both Aβ and tau show aggregation into soluble and insoluble oligomers. Soluble oligomers of Aβ include their most predominant forms - Aβ1-40 and Aβ1-42 - as well as shorter peptides such as Aβ25-35 or Aβ25-35/40. Most animal models of AD have been developed using transgenesis, based on identified human mutations. However, these familial forms of AD represent less than 1% of AD cases. In this context, the idea emerged in the 1990s to directly inject the Aβ25-35 fragment into the rodent brain to develop an acute model of AD that could mimic the disease's sporadic forms (99% of all cases). This review aims to: (1) summarize the biological activity of Aβ25-35, focusing on its impact on the main structural and functional alterations observed in AD (cognitive deficits, APP misprocessing, tau system dysfunction, neuroinflammation, oxidative stress, cholinergic and glutamatergic alterations, HPA axis dysregulation, synaptic deficits and cell death); and (2) confirm the interest of this pathomimetic model in AD research, as it has helped identify and characterize many molecules (marketed, in clinical development, and in preclinical testing), and to the development of alternative approaches for AD prevention and therapy. Today, the Aβ25-35 model appears as a first-intent choice model to rapidly screen the symptomatic or neuroprotective potencies of new compounds, chemical series, or innovative therapeutic strategies.
Collapse
|
15
|
Ramírez-Hernández E, Sánchez-Maldonado C, Patricio-Martínez A, Limón ID. Amyloid-β (25-35) induces the morphological alteration of dendritic spines and decreases NR2B and PSD-95 expression in the hippocampus. Neurosci Lett 2023; 795:137030. [PMID: 36572143 DOI: 10.1016/j.neulet.2022.137030] [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: 08/15/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022]
Abstract
Research on the memory impairment caused by the Amyloid-β 25-35 (Aβ25-35) peptide in animal models has provided an understanding of the causes that occurs in Alzheimer's disease. However, it is uncertain whether this cognitive impairment occurs due to disruption of information encoding and consolidation or impaired retrieval of stored memory. The aim of this study was to determine the effect of the Aβ25-35 peptide on the morphology of dendritic spines and the changes in the expression of NR2B and PSD-95 in the hippocampus associated with learning and memory deficit. Vehicle or Aβ25-35 peptide (0.1 µg/µL) was bilaterally administered into the CA1 subfield of the rat hippocampus, then tested for spatial learning and memory in the Morris Water Maze. On Day 39, the morphological changes in the CA1 of the hippocampus and dentate gyrus were examined via Golgi-Cox stain. It was observed that the Aβ25-35 peptide administered in the CA1 region of the rat hippocampus induced changes to the morphology of dendritic spines and the expression of the NR2B subunit of the NMDA receptor co-localized with both the spatial memory and PSD-95 protein in the hippocampus of learning rats. We conclude that, in soluble form, the Aβ25-35 peptide perturbs synaptic plasticity, specifically in the formation of new synapses, thus promoting the progression of memory impairment.
Collapse
Affiliation(s)
- Eleazar Ramírez-Hernández
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma Puebla, Puebla, Puebla, Mexico; Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Claudia Sánchez-Maldonado
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma Puebla, Puebla, Puebla, Mexico
| | - Aleidy Patricio-Martínez
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma Puebla, Puebla, Puebla, Mexico; Facultad de Ciencias Biológicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Ilhiucamina Daniel Limón
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma Puebla, Puebla, Puebla, Mexico.
| |
Collapse
|
16
|
Increment of CSF fractalkine-positive microvesicles preceded the spatial memory impairment in amyloid beta neurotoxicity. Cytokine 2022; 160:156050. [PMID: 36179535 DOI: 10.1016/j.cyto.2022.156050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 09/03/2022] [Accepted: 09/09/2022] [Indexed: 11/23/2022]
Abstract
BACKGROUND Fractalkine (CX3CL1) is a key chemokine, affects neuronal cell communication and involves in Alzheimer's disease pathogenesis. Microvesicles (MVs) participate in neuronal cells' cross-talk in physiological and pathological states. Microvesicles released in cerebrospinal fluid (CSF) may provide a valuable footprint of brain changes. Little information is available regarding the release of fractalkine-positive MVs (CX3CL1+ -MVs) in the nervous system. METHODS We induced cognitive impairment by bilateral injection of amyloid-beta (Aβ) into the cerebral ventricles. We analyzed the CSF by flow cytometry in two experiments (trained and untrained) to elucidate the presence of CX3CL1+ -MVs. The hippocampal TNF-α as an inflammatory factor was assessed by immunohistochemistry. RESULTS The Aβ induced spatial memory impairment after two weeks, verified by a decrease in the escape latency in Morris water maze test. It caused an increase in the anxiety-like behaviors demonstrated by a decrease in entries into the open arms of elevated plus maze test. The Aβ increased the percent of the positive area for TNF-α staining. Histological evaluation of the hippocampus confirmed the tissue injuries. The CSF levels of CX3CL1+ -MVs, increased 2 and 7 days after Aβ injection. The Aβ increased the TNF-α staining and provided an inflammatory context to facilitate the MVs release. The rise of CX3CL1+ -MVs was transient and subsided after two weeks. Both trained and untrained experiments showed a similar rise pattern of CX3CL1+ -MVs. CONCLUSION Increase of fractalkine-positive microvesicles preceded the cognitive impairment, more studies are required to approve the CX3CL1+ -MVs as a potential biomarker in the early diagnosis of Alzheimer's disease.
Collapse
|
17
|
Thapak P, Khare P, Bishnoi M, Sharma SS. Neuroprotective Effect of 2-Aminoethoxydiphenyl Borate (2-APB) in Amyloid β-Induced Memory Dysfunction: A Mechanistic Study. Cell Mol Neurobiol 2022; 42:1211-1223. [PMID: 33219878 PMCID: PMC11441215 DOI: 10.1007/s10571-020-01012-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 11/13/2020] [Indexed: 02/08/2023]
Abstract
β-Amyloid (Aβ) peptide is a characteristic feature of Alzheimer's disease (AD) and accumulation of Aβ is associated with loss of synaptic plasticity and neuronal cell death. Aggregation of Aβ initiates numerous molecular signalling pathways leading to oxidative stress, mitochondrial dysfunction as well as an imbalance of calcium ion influx homeostasis. Recently, it has been shown that transient receptor potential melastatin 2 (TRPM2), a non-selective calcium-permeable cation channel has been postulated to play a vital role in the neuronal death, indicating the potential of TRPM2 inhibition in CNS disease. In this study, neuroprotective potential of 2-aminoethoxydiphenyl borate (2-APB), a broad-spectrum calcium channels blocker was investigated in Aβ-induced memory deficits in rats. In addition, effect of 2-APB on TRPM2 channels gene and protein expressions and also on calcium and memory related proteins was investigated in the hippocampus. Intracerebroventricular (I.C.V.) administration of Aβ (Aβ25-35, 10 μg) markedly induced cognitive impairment and upregulation of mRNA and protein expression of TRPM2 in the hippocampus. In addition, AChE activity was also increased in the cortex of the Aβ administered animals. Three-week treatment with 2-APB led to the down-regulation of TRPM2 mRNA and protein expression in the hippocampus and also improved the cognitive functions which was evident from the behavioral parameters. Moreover, 2-APB treatment also increased the calcium and memory associated proteins namely p-CaMKII, p-GSK-3β, p-CREB and PSD-95 in the hippocampus and reduced the mRNA level of calcium buffering proteins and calcineurin A (PPP3CA) in the hippocampus. Furthermore, 2-APB treatment significantly reduced the AChE activity in the cortex. Thus, our findings suggest the neuroprotective effect of 2-APB in Aβ-induced cognitive impairment.
Collapse
Affiliation(s)
- Pavan Thapak
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Mohali, Punjab, 160062, India
| | - Pragyanshu Khare
- National Agri-Food Biotechnology Institute (NABI), S.A.S. Nagar, Mohali, Punjab, India
| | - Mahendra Bishnoi
- National Agri-Food Biotechnology Institute (NABI), S.A.S. Nagar, Mohali, Punjab, India
| | - Shyam Sunder Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Mohali, Punjab, 160062, India.
| |
Collapse
|
18
|
Zussy C, John R, Urgin T, Otaegui L, Vigor C, Acar N, Canet G, Vitalis M, Morin F, Planel E, Oger C, Durand T, Rajshree SL, Givalois L, Devarajan PV, Desrumaux C. Intranasal Administration of Nanovectorized Docosahexaenoic Acid (DHA) Improves Cognitive Function in Two Complementary Mouse Models of Alzheimer’s Disease. Antioxidants (Basel) 2022; 11:antiox11050838. [PMID: 35624701 PMCID: PMC9137520 DOI: 10.3390/antiox11050838] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/13/2022] [Accepted: 04/20/2022] [Indexed: 02/04/2023] Open
Abstract
Polyunsaturated fatty acids (PUFAs) are a class of fatty acids that are closely associated with the development and function of the brain. The most abundant PUFA is docosahexaenoic acid (DHA, 22:6 n-3). In humans, low plasmatic concentrations of DHA have been associated with impaired cognitive function, low hippocampal volumes, and increased amyloid deposition in the brain. Several studies have reported reduced brain DHA concentrations in Alzheimer’s disease (AD) patients’ brains. Although a number of epidemiological studies suggest that dietary DHA consumption may protect the elderly from developing cognitive impairment or dementia including AD, several review articles report an inconclusive association between omega-3 PUFAs intake and cognitive decline. The source of these inconsistencies might be because DHA is highly oxidizable and its accessibility to the brain is limited by the blood–brain barrier. Thus, there is a pressing need for new strategies to improve DHA brain supply. In the present study, we show for the first time that the intranasal administration of nanovectorized DHA reduces Tau phosphorylation and restores cognitive functions in two complementary murine models of AD. These results pave the way for the development of a new approach to target the brain with DHA for the prevention or treatment of this devastating disease.
Collapse
Affiliation(s)
- Charleine Zussy
- MMDN, University Montpellier, EPHE, INSERM, 34095 Montpellier, France; (C.Z.); (T.U.); (L.O.); (G.C.); (M.V.); (L.G.)
| | - Rijo John
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Deemed University, Mumbai 400019, India; (R.J.); (S.L.R.); (P.V.D.)
| | - Théo Urgin
- MMDN, University Montpellier, EPHE, INSERM, 34095 Montpellier, France; (C.Z.); (T.U.); (L.O.); (G.C.); (M.V.); (L.G.)
| | - Léa Otaegui
- MMDN, University Montpellier, EPHE, INSERM, 34095 Montpellier, France; (C.Z.); (T.U.); (L.O.); (G.C.); (M.V.); (L.G.)
| | - Claire Vigor
- IBMM, Pôle Chimie Balard Recherche, Université de Montpellier, CNRS, ENSCM, 34095 Montpellier, France; (C.V.); (C.O.); (T.D.)
| | - Niyazi Acar
- Centre des Sciences du Goût et de l’Alimentation, AgroSup Dijon, CNRS, INRAE, Université de Bourgogne Franche-Comté, 21000 Dijon, France;
| | - Geoffrey Canet
- MMDN, University Montpellier, EPHE, INSERM, 34095 Montpellier, France; (C.Z.); (T.U.); (L.O.); (G.C.); (M.V.); (L.G.)
| | - Mathieu Vitalis
- MMDN, University Montpellier, EPHE, INSERM, 34095 Montpellier, France; (C.Z.); (T.U.); (L.O.); (G.C.); (M.V.); (L.G.)
| | - Françoise Morin
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Laval University, CR-CHUQ, Québec City, QC G1V 0A6, Canada; (F.M.); (E.P.)
| | - Emmanuel Planel
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Laval University, CR-CHUQ, Québec City, QC G1V 0A6, Canada; (F.M.); (E.P.)
| | - Camille Oger
- IBMM, Pôle Chimie Balard Recherche, Université de Montpellier, CNRS, ENSCM, 34095 Montpellier, France; (C.V.); (C.O.); (T.D.)
| | - Thierry Durand
- IBMM, Pôle Chimie Balard Recherche, Université de Montpellier, CNRS, ENSCM, 34095 Montpellier, France; (C.V.); (C.O.); (T.D.)
| | - Shinde L. Rajshree
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Deemed University, Mumbai 400019, India; (R.J.); (S.L.R.); (P.V.D.)
| | - Laurent Givalois
- MMDN, University Montpellier, EPHE, INSERM, 34095 Montpellier, France; (C.Z.); (T.U.); (L.O.); (G.C.); (M.V.); (L.G.)
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Laval University, CR-CHUQ, Québec City, QC G1V 0A6, Canada; (F.M.); (E.P.)
| | - Padma V. Devarajan
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Deemed University, Mumbai 400019, India; (R.J.); (S.L.R.); (P.V.D.)
| | - Catherine Desrumaux
- MMDN, University Montpellier, EPHE, INSERM, 34095 Montpellier, France; (C.Z.); (T.U.); (L.O.); (G.C.); (M.V.); (L.G.)
- LIPSTIC LabEx, 21000 Dijon, France
- Correspondence: ; Tel.: +33-467-14-36-89; Fax: +33-467-14-33-86
| |
Collapse
|
19
|
Hugon G, Goutal S, Sarazin M, Bottlaender M, Caillé F, Droguerre M, Charvériat M, Winkeler A, Tournier N. Impact of Donepezil on Brain Glucose Metabolism Assessed Using [ 18F]2-Fluoro-2-deoxy-D-Glucose Positron Emission Tomography Imaging in a Mouse Model of Alzheimer's Disease Induced by Intracerebroventricular Injection of Amyloid-Beta Peptide. Front Neurosci 2022; 16:835577. [PMID: 35281502 PMCID: PMC8916213 DOI: 10.3389/fnins.2022.835577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/04/2022] [Indexed: 11/09/2022] Open
Abstract
Translational methods are needed to monitor the impact of the Alzheimer's disease (AD) and therapies on brain function in animal models and patients. The formation of amyloid plaques was investigated using [18F]florbetapir autoradiography in a mouse model of AD consisting in unilateral intracerebroventricular (i.c.v) injection of amyloid peptide Aβ25-35. Then, an optimized positron emission tomography (PET) imaging protocol using [18F]2-fluoro-2-deoxy-D-glucose ([18F]FDG) was performed to estimate brain glucose metabolism: [18F]FDG was injected in awake animals to allow for 40 min brain uptake in freely moving mice. Anesthesia was then induced for 30 min PET acquisition to capture the slow and poorly reversible brain uptake of [18F]FDG. Impact of donepezil (0.25 mg/kg daily, 7 days, orally) on brain function was investigated in AD mice (n = 6 mice/group). Formation of amyloid plaques could not be detected using autoradiography. Compared with sham controls (injection of scramble peptide), significant decrease in [18F]FDG uptake was observed in the AD group in the subcortical volume of the ipsilateral hemisphere. Donepezil restored normal glucose metabolism by selectively increasing glucose metabolism in the affected subcortical volume but not in other brain regions. In mice, [18F]FDG PET imaging can be optimized to monitor impaired brain function associated with i.c.v injection of Aβ25-35, even in the absence of detectable amyloid plaque. This model recapitulates the regional decrease in [18F]FDG uptake observed in AD patients. [18F]FDG PET imaging can be straightforwardly transferred to AD patients and may aid the development of certain therapies designed to restore the altered brain function in AD.
Collapse
Affiliation(s)
- Gaëlle Hugon
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Orsay, France
| | - Sébastien Goutal
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Orsay, France
| | - Marie Sarazin
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Orsay, France,Department of Neurology of Memory and Language, GHU Paris Psychiatry and Neurosciences, Paris, France,Faculté de Médicine, Université de Paris, Paris, France
| | - Michel Bottlaender
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Orsay, France,NeuroSpin, Frédéric Joliot Life Sciences Institute, CEA, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Fabien Caillé
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Orsay, France
| | | | | | - Alexandra Winkeler
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Orsay, France
| | - Nicolas Tournier
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Orsay, France,*Correspondence: Nicolas Tournier,
| |
Collapse
|
20
|
MacKenzie JL, Ivanova N, Nell HJ, Giordano CR, Terlecky SR, Agca C, Agca Y, Walton PA, Whitehead SN, Cechetto DF. Microglial inflammation and cognitive dysfunction in comorbid rat models of striatal ischemic stroke and alzheimer’s disease: effects of antioxidant catalase-SKL on behavioral and cellular pathology. Neuroscience 2022; 487:47-65. [DOI: 10.1016/j.neuroscience.2022.01.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/26/2022] [Accepted: 01/28/2022] [Indexed: 12/25/2022]
|
21
|
Assessment of lipophilic fluorescence products in β-amyloid-induced cognitive decline: A parallel track in hippocampus, CSF, plasma and erythrocytes. Exp Gerontol 2021; 157:111645. [PMID: 34843902 DOI: 10.1016/j.exger.2021.111645] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 11/17/2021] [Accepted: 11/23/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND Oxidative stress implicates in Alzheimer's disease (AD) pathophysiology, and associates with the creation of end products of free radical reactions, are known as lipophilic fluorescent products (LFPs). This study aimed to evaluate the probable parallel alterations in the spectral properties of the LFPs in the hippocampus tissues, cerebrospinal fluid (CSF), plasma, and erythrocytes during AD model induction by intra-cerebroventricular (ICV) amyloid β-protein fragment 25-35 (Aβ) injection. METHODS Male rats received an intra-ICV injection of Aβ. Hippocampus, CSF, plasma, and erythrocytes were harvested at 5, 14, and 21 days after Aβ injection. The fluorescent intensity of LFPs was assessed by spectrofluorimetry using synchronous fluorescence spectra 25 (SYN 25) and 50 (SYN 50) in the range of 250-500 nm. Hippocampal tissue malondialdehyde (MDA) and superoxide dismutase (SOD) were also measured. Cognitive alterations were evaluated using Morris water maze (MWM) test. RESULTS The parallel significant rise in the fluorescence intensity of LFPs was detected in the hippocampus, CSF, plasma, and erythrocytes, 14, and 21 days after ICV-Aβ injection. These alterations were found in both types of synchronous spectra 25, and 50, and were coincided with hippocampal cognitive decline, the MDA rise, and decrease of SOD activity. There was a positive correlation between hippocampus homogenate, and plasma or CSF rise in fluorescence intensity. CONCLUSION Data showed that the Aβ increased hippocampal MDA, and decreased SOD activity, led to a higher rate of oxidative products and subsequently resulted in an increase in LFPs fluorescence intensity during the development of cognitive decline. LFPs' alterations reflect a comprehensive view of tissue redox status. The fluorescence properties of LFPs indicate their composition, which may pave the way to trace the different pathological states.
Collapse
|
22
|
Lima KR, Schmidt HL, Daré LR, Soares CB, Lopes LF, Carpes FP, Mello-Carpes PB. Concurrent exercise does not prevent recognition memory deficits induced by beta-amyloid in rats. Physiol Behav 2021; 243:113631. [PMID: 34715093 DOI: 10.1016/j.physbeh.2021.113631] [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: 03/23/2021] [Revised: 10/07/2021] [Accepted: 10/25/2021] [Indexed: 10/20/2022]
Abstract
Alzheimer's disease affects thousands of people worldwide. Alternatives aiming to prevent the disease or reduce its symptoms include different physical exercise configurations. Here we investigate the potential of concurrent exercise to prevent recognition memory deficits in an Alzheimer's disease-like model induced by the hippocampal beta-amyloid (Aβ) injection in Wistar rats. We demonstrate that the concurrent exercise, which included running and strength exercises performed in the same exercise session, is ineffective in preventing recognition memory deficits in the Aβ rats. Besides, higher levels of reactive oxygen species were found in the concurrent exercise group's hippocampus. The running exercise administrated alone prevented recognition memory impairments.
Collapse
Affiliation(s)
- Karine Ramires Lima
- Physiology Research Group, Stress, Memory and Behavior Lab, Federal University of Pampa, Uruguaiana, RS, Brazil
| | - Helen Lidiane Schmidt
- Physiology Research Group, Stress, Memory and Behavior Lab, Federal University of Pampa, Uruguaiana, RS, Brazil; Applied Neuromechanics Group, Laboratory of Neuromechanics, Universidade Federal do Pampa, Uruguaiana, RS, Brazil
| | - Leticia Rossi Daré
- Physiology Research Group, Stress, Memory and Behavior Lab, Federal University of Pampa, Uruguaiana, RS, Brazil
| | - Caroline Bitencourt Soares
- Physiology Research Group, Stress, Memory and Behavior Lab, Federal University of Pampa, Uruguaiana, RS, Brazil
| | - Luiza Freitas Lopes
- Physiology Research Group, Stress, Memory and Behavior Lab, Federal University of Pampa, Uruguaiana, RS, Brazil
| | - Felipe P Carpes
- Applied Neuromechanics Group, Laboratory of Neuromechanics, Universidade Federal do Pampa, Uruguaiana, RS, Brazil
| | - Pâmela Billig Mello-Carpes
- Physiology Research Group, Stress, Memory and Behavior Lab, Federal University of Pampa, Uruguaiana, RS, Brazil.
| |
Collapse
|
23
|
Soares CB, Daré LR, Lima KR, Lopes LF, Santos AGD, Schimidt HL, Carpes FP, Lloret A, Viña J, Mello-Carpes PB. Multicomponent Training Prevents Memory Deficit Related to Amyloid-β Protein-Induced Neurotoxicity. J Alzheimers Dis 2021; 83:143-154. [PMID: 34275902 DOI: 10.3233/jad-210424] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by the accumulation of the amyloid-β peptide in the brain, leading to early oxidative stress and neurotoxicity. It has been suggested that physical exercise could be beneficial in preventing AD, but studies with multicomponent training are scanty. OBJECTIVE Verify the effects of multicomponent exercise training to prevent deficits in recognition memory related to Aβ neurotoxicity. METHODS We subjected Wistar rats to multicomponent training (including aerobic and anaerobic physical exercise and cognitive exercise) and then infused amyloid-β peptide into their hippocampus. RESULTS We show that long-term multicomponent training prevents the amyloid-β-associated neurotoxicity in the hippocampus. It reduces hippocampal lipid peroxidation, restores antioxidant capacity, and increases glutathione levels, finally preventing recognition memory deficits. CONCLUSION Multicomponent training avoids memory deficits related to amyloid-β neurotoxicity on an animal model.
Collapse
Affiliation(s)
| | - Leticia Rossi Daré
- Federal University of Pampa, campus Uruguaiana, Uruguaiana, RS, Brazil.,Federal University of Triangulo Mineiro, Uberaba, MG, Brazil
| | | | | | | | | | | | | | - Jose Viña
- University of Valencia, Valencia, Spain
| | | |
Collapse
|
24
|
Paulo SL, Ribeiro-Rodrigues L, Rodrigues RS, Mateus JM, Fonseca-Gomes J, Soares R, Diógenes MJ, Solá S, Sebastião AM, Ribeiro FF, Xapelli S. Sustained Hippocampal Neural Plasticity Questions the Reproducibility of an Amyloid-β-Induced Alzheimer's Disease Model. J Alzheimers Dis 2021; 82:1183-1202. [PMID: 34151790 DOI: 10.3233/jad-201567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The use of Alzheimer's disease (AD) models obtained by intracerebral infusion of amyloid-β (Aβ) has been increasingly reported in recent years. Nonetheless, these models may present important challenges. OBJECTIVE We have focused on canonical mechanisms of hippocampal-related neural plasticity to characterize a rat model obtained by an intracerebroventricular (icv) injection of soluble amyloid-β42 (Aβ42). METHODS Animal behavior was evaluated in the elevated plus maze, Y-Maze spontaneous or forced alternation, Morris water maze, and open field, starting 2 weeks post-Aβ42 infusion. Hippocampal neurogenesis was assessed 3 weeks after Aβ42 injection. Aβ deposition, tropomyosin receptor kinase B levels, and neuroinflammation were appraised at 3 and 14 days post-Aβ42 administration. RESULTS We found that immature neuronal dendritic morphology was abnormally enhanced, but proliferation and neuronal differentiation in the dentate gyrus was conserved one month after Aβ42 injection. Surprisingly, animal behavior did not reveal changes in cognitive performance nor in locomotor and anxious-related activity. Brain-derived neurotrophic factor related-signaling was also unchanged at 3 and 14 days post-Aβ icv injection. Likewise, astrocytic and microglial markers of neuroinflammation in the hippocampus were unaltered in these time points. CONCLUSION Taken together, our data emphasize a high variability and lack of behavioral reproducibility associated with these Aβ injection-based models, as well as the need for its further optimization, aiming at addressing the gap between preclinical AD models and the human disorder.
Collapse
Affiliation(s)
- Sara L Paulo
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Leonor Ribeiro-Rodrigues
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Rui S Rodrigues
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Joana M Mateus
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - João Fonseca-Gomes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Rita Soares
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Biologia Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Maria J Diógenes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Susana Solá
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisboa, Portugal
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Filipa F Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Sara Xapelli
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
25
|
Thapak P, Bishnoi M, Sharma SS. Tranilast, a Transient Receptor Potential Vanilloid 2 Channel (TRPV2) Inhibitor Attenuates Amyloid β-Induced Cognitive Impairment: Possible Mechanisms. Neuromolecular Med 2021; 24:183-194. [PMID: 34231190 DOI: 10.1007/s12017-021-08675-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 06/24/2021] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is associated with the accumulation of β-amyloid and leads to cognitive impairment. Numerous studies have established that neuronal calcium homeostasis is perturbed in AD. Recently, transient receptor potential vanilloid 2 (TRPV2) channels, a non-selective calcium-permeable channel, have been investigated in several diseases. However, the role of the TRPV2 channel has not been investigated in AD yet. In this study, intracerebroventricular administration of β-amyloid (10 μg) to Sprague Dawley rats resulted in cognitive impairment which was evident from the assessment of cognitive tests. Also, TRPV2 mRNA and protein expression were found to be upregulated, while the expression of Ca2+/calmodulin-dependent protein kinase II (p-CaMKII-Thr-286), glycogen synthase kinase 3β (p-GSK-3β-Ser-9), cAMP response element-binding protein (p-CREB-Ser-133), and postsynaptic density protein 95 (PSD-95) were downregulated in the hippocampus of β-amyloid-treated animals. Even, β-amyloid-treated animals showed upregulation of mRNA level of calcium buffering proteins (parvalbumin and calsequestrin) and calcineurin A (PPP3CA) in the hippocampus. Acetylcholinesterase activity was also increased in the cortex of β-amyloid-treated animals. Three-week treatment with tranilast showed improvement in the cognitive parameters which was associated with a decrease in TRPV2 expression and AChE activity. Additionally, an increase in the protein expression of p-CaMKII, p-GSK-3β, p-CREB and PSD-95 in the hippocampus was found. Downregulation in the mRNA level of calcium buffering proteins (parvalbumin and calsequestrin) and calcineurin A in the hippocampus was also seen. These results reveal the importance of TRPV2 channels in the β-amyloid-induced cognitive deficits and suggest TRPV2 as a potential target for AD.
Collapse
Affiliation(s)
- Pavan Thapak
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali, Punjab, 160062, India
| | - Mahendra Bishnoi
- National Agri-Food Biotechnology Institute, S.A.S. Nagar, Punjab, India
| | - Shyam Sunder Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali, Punjab, 160062, India.
| |
Collapse
|
26
|
Scheiner M, Hoffmann M, He F, Poeta E, Chatonnet A, Monti B, Maurice T, Decker M. Selective Pseudo-irreversible Butyrylcholinesterase Inhibitors Transferring Antioxidant Moieties to the Enzyme Show Pronounced Neuroprotective Efficacy In Vitro and In Vivo in an Alzheimer's Disease Mouse Model. J Med Chem 2021; 64:9302-9320. [PMID: 34152756 DOI: 10.1021/acs.jmedchem.1c00534] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A series of multitarget-directed ligands (MTDLs) was designed by functionalizing a pseudo-irreversible butyrylcholinesterase (BChE) inhibitor. The obtained hybrids were investigated in vitro regarding their hBChE and hAChE inhibition, their enzyme kinetics, and their antioxidant physicochemical properties (DPPH, ORAC, metal chelating). In addition, in vitro assays were applied to investigate antioxidant effects using murine hippocampal HT22 cells and immunomodulatory effects on the murine microglial N9 cell line. The MTDLs retained their antioxidative properties compared to the parent antioxidant-moieties in vitro and the inhibition of hBChE was maintained in the submicromolar range. Representative compounds were tested in a pharmacological Alzheimer's disease (AD) mouse model and demonstrated very high efficacy at doses as low as 0.1 mg/kg. The most promising compound was also tested in BChE-/- mice and showed reduced efficacy. In vivo neuroprotection by BChE inhibition can be effectively enhanced by incorporation of structurally diverse antioxidant moieties.
Collapse
Affiliation(s)
- Matthias Scheiner
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Matthias Hoffmann
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Feng He
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Eleonora Poeta
- Department of Pharmacy and Biotechnology, University of Bologna, Via Selmi 3, 40126 Bologna, Italy
| | - Arnaud Chatonnet
- DMEM, University of Montpellier, INRAE, 34060 Montpellier, France
| | - Barbara Monti
- Department of Pharmacy and Biotechnology, University of Bologna, Via Selmi 3, 40126 Bologna, Italy
| | - Tangui Maurice
- MMDN, University of Montpellier, INSERM, EPHE, 34095 Montpellier, France
| | - Michael Decker
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| |
Collapse
|
27
|
Marbouti L, Zahmatkesh M, Riahi E, Shafiee Sabet M. GnRH protective effects against amyloid β-induced cognitive decline: A potential role of the 17β-estradiol. Mol Cell Endocrinol 2020; 518:110985. [PMID: 32805333 DOI: 10.1016/j.mce.2020.110985] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 08/04/2020] [Accepted: 08/07/2020] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The 17β-estradiol (E2) enhances hippocampal dendritic spine synapses, facilitates learning processes, and exerts neuroprotection. Brain estrogen decline has been reported in Alzheimer's disease. The role of GnRH in modulating steroid biosynthesis convinced us to examine whether hippocampal GnRH administration could enhance the local E2 levels and overcome the development of cognition decline in amyloid β (Aβ) neurotoxicity. To explore if GnRH acts through regulating E2 synthesis, letrozole, an aromatase inhibitor, has been applied in combination with GnRH. METHODS Female rats received an intracerebroventricular injection of Aβ. The GnRH and, or letrozole were injected into the CA1 for 14 consecutive days. Working memory, novel object recognition memory, and anxiety-like behavior were evaluated. Serum and hippocampal E2 levels were measured. Hippocampal mRNA expression of GnRH (GnRH-R) and E2 (ERα and ERβ) receptors was assessed. GnRH effect on the excitability of pyramidal cells was studied by in vivo single-unit recording. RESULTS GnRH increased hippocampal E2 levels, evoked an increase in the spontaneous firing of pyramidal neurons, and caused mRNA overexpression of hippocampal GnRH receptors. GnRH prevented the adverse effects of Aβ on working memory, NOR index, and anxiogenic behavior. Letrozole did not reverse GnRH modulatory effects on hippocampal E2 levels and neuroprotection. CONCLUSION GnRH prevented the Aβ-induced memory deficit, which may be mediated through hippocampal E2 levels enhancement. The electrophysiological analysis revealed the enhanced neuronal excitability in the CA1 region. All these data suggest that GnRH might be a promising candidate that reduces anxiety and improves memory indices in the context of Aβ neurotoxicity.
Collapse
Affiliation(s)
- Ladan Marbouti
- Neuroscience and Addiction Studies Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Zahmatkesh
- Neuroscience and Addiction Studies Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran; Cognitive and Behavioral Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Esmail Riahi
- Physiology Department, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Shafiee Sabet
- Family Medicine Department, Ziaeian Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Lv J, Chen L, Zhu N, Sun Y, Pan J, Gao J, Liu J, Liu G, Tao Y. Beta amyloid-induced time-dependent learning and memory impairment: involvement of HPA axis dysfunction. Metab Brain Dis 2020; 35:1385-1394. [PMID: 32860609 DOI: 10.1007/s11011-020-00613-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 08/24/2020] [Indexed: 12/28/2022]
Abstract
Aβ aggregation is one of the pathological biomarkers of Alzheimer's disease (AD). However, the possible mechanism related to Aβ-induced pathological signaling pathway is still unknown. In the present study, Aβ1-42-induced time-dependent memory impairment and its possible relationship to hypothalamic-pituitary-adrenal (HPA) axis hyperactivity were examined. Aβ1-42-treated mice significantly impaired acquisition activity in the learning curve at 10 days, 1 and 4 months in the Morris water-maze (MWM) task. This learning activity was back to normal at 8 months after Aβ1-42 treatment. In the probe trial test, Aβ1-42-treated mice needed longer latencies to touch the precious platform location and fewer numbers of crossing from 10 days to 4 months after microinjection. This Aβ1-42 induced memory loss was consistent with the results of the step-down passive avoidance test. The HPA axis related parameters, such as corticosterone (CORT) level in the serum, glucocorticoid receptor (GR) and corticotropin-releasing factor receptor (CRF-R) expression in the frontal cortex and hippocampus increased in Aβ1-42-treated mice from 10 days to 4 months. While the downstream molecules phosphorylation of cyclic AMP response element binding (pCREB) and brain-derived neurotrophic factor (BDNF) expression decreased during this time. These effects were back to normal 8 months after treatment with Aβ1-42. Altogether, our results suggested that Aβ1-42 induced significant learning and memory impairment, which is involved in HPA axis dysfunction.
Collapse
Affiliation(s)
- Jinpeng Lv
- College of Pharmaceutical Engineering and Life Sciences, Changzhou University, Changzhou, 213000, China
| | - Ling Chen
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, People's Republic of China
| | - Naping Zhu
- Brain Institute, School of Pharmacy, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yindi Sun
- Department of Traditional Medical Orthopedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Jianchun Pan
- Brain Institute, School of Pharmacy, Wenzhou Medical University, Wenzhou, 325035, China
| | - Jinsheng Gao
- Department of Oncology, Shanxi Province Research Institute of Traditional Chinese Medicine, Taiyuan, 030000, China
| | - Jianwu Liu
- College of Pharmaceutical Engineering and Life Sciences, Changzhou University, Changzhou, 213000, China
| | - Guangjun Liu
- The Second People's Hospital of Changzhou, Affiliate Hospital of NanJing Medical University, Changzhou, 213000, China.
| | - Yuanxiang Tao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA.
| |
Collapse
|
29
|
Riban V, Meunier J, Buttigieg D, Villard V, Verleye M. In Vitro and In Vivo Neuroprotective Effects of Etifoxine in β-Amyloidinduced Toxicity Models. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2020; 19:227-240. [DOI: 10.2174/1871527319666200601151007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 04/15/2020] [Accepted: 04/15/2020] [Indexed: 11/22/2022]
Abstract
Aim:
The aim of this study is to examine the effect of etifoxine on β-amyloid-induced toxicity
models.
Background:
Etifoxine is an anxiolytic compound with a dual mechanism of action; it is a positive allosteric
modulator of GABAergic receptors as well as a ligand for the 18 kDa mitochondrial Translocator
Protein (TSPO). TSPO has recently raised interest in Alzheimer’s Disease (AD), and experimental studies
have shown that some TSPO ligands could induce neuroprotective effects in animal models.
Objective:
In this study, we examined the potential protective effect of etifoxine in an in vitro and an
in vivo model of amyloid beta (Aβ)-induced toxicity in its oligomeric form, which is a crucial factor in
AD pathologic mechanisms.
Method:
Neuronal cultures were intoxicated with Aβ1-42, and the effects of etifoxine on oxidative
stress, Tau-hyperphosphorylation and synaptic loss were quantified. In a mice model, behavioral deficits
induced by intracerebroventricular administration of Aβ25-35 were measured in a spatial memory
test, the spontaneous alternation and in a contextual memory test, the passive avoidance test.
Results:
In neuronal cultures intoxicated with Aβ1-42, etifoxine dose-dependently decreased oxidative
stress (methionine sulfoxide positive neurons), tau-hyperphosphorylation and synaptic loss (ratio
PSD95/synaptophysin). In a mice model, memory impairments were fully alleviated by etifoxine administered
at anxiolytic doses (12.5-50mg/kg). In addition, markers of oxidative stress and apoptosis
were decreased in the hippocampus of these animals.
Conclusion:
Our results have shown that in these two models, etifoxine could fully prevent neurotoxicity
and pathological changes induced by Aβ. These results confirm that TSPO ligands could offer an
interesting therapeutic approach to Alzheimer’s disease.
Collapse
Affiliation(s)
- Veronique Riban
- Pharmacology Department, Biocodex, 3 Chemin d’Armancourt, 60200 Compiegne, France
| | - Johann Meunier
- Amylgen, 2196 Boulevard de la Lironde, 34980 Montferrier sur Lez, France
| | | | - Vanessa Villard
- Amylgen, 2196 Boulevard de la Lironde, 34980 Montferrier sur Lez, France
| | - Marc Verleye
- Pharmacology Department, Biocodex, 3 Chemin d’Armancourt, 60200 Compiegne, France
| |
Collapse
|
30
|
Huang-Pu-Tong-Qiao Formula Ameliorates Tau Phosphorylation by Inhibiting the CaM-CaMKIV Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:8956071. [PMID: 34046075 PMCID: PMC8128057 DOI: 10.1155/2020/8956071] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 03/09/2020] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disease. It is a chronic, lethal disease in which brain function is severely impaired and neuronal damage is irreversible. Huang-Pu-Tong-Qiao (HPTQ), a formula from traditional Chinese medicine, has been used in the clinical treatment of AD for many years, with remarkable effects. However, the neuroprotective mechanisms of HPTQ in AD have not yet been investigated. In the present study, we used AD models in vivo and in vitro, to investigate both the neuroprotective effect of HPTQ water extracts (HPTQ-W) and the potential mechanisms of this action. For the in vivo study, after HPTQ intervention, the Morris water maze test was used to examine learning and memory in rats. Transmission electron microscopy and immunofluorescence methods were then used to investigate neuronal damage. For the in vitro experiments, rat primary hippocampal neurons were cultured and cell viability was examined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide. Additionally, mRNA levels of CaM, CaMKK, CaMKIV, and tau were examined using qRT-PCR, and protein expression of CaM, CaMKK, p-CaMKIV, and p-tau were examined using western blot. In vivo, we revealed that HPTQ significantly improved learning and memory deficits and attenuated neuronal damage in the AD rat model. Furthermore, in vitro results showed that HPTQ significantly increased cell viability in the AD cell model. We also demonstrated that HPTQ significantly decreased the mRNA levels of CaM, CaMKK, CaMKIV, and tau and significantly decreased the protein expressions of CaM, CaMKK, p-CaMKIV, and p-tau. In conclusion, our results indicated that HPTQ improved cognition and ameliorated neuronal damage in AD models and implicated a reduction in tau phosphorylation caused by inhibition of the CaM-CaMKIV pathway as a possible mechanism.
Collapse
|
31
|
Ooi TC, Ahmad Munawar M, Mohd Rosli NH, Abdul Malek SNA, Rosli H, Ibrahim FW, Azmi N, Haron H, Sharif R, Shahar S, Rajab NF. Neuroprotection of Tropical Fruit Juice Mixture via the Reduction of iNOS Expression and CRH Level in β-Amyloid-Induced Rats Model of Alzheimer's Disease. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2020; 2020:5126457. [PMID: 32382294 PMCID: PMC7180421 DOI: 10.1155/2020/5126457] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/16/2020] [Accepted: 03/21/2020] [Indexed: 01/08/2023]
Abstract
This study aimed to determine the effects of tropical fruit juice mixture (pomegranate, white guava, and Roselle) on biochemical, behavioral, and histopathological changes of β-amyloid- (Aβ-) induced rats. Formulation 8 (F8) of tropical fruit juice mixture was chosen for this present study due to its high phenolic content and antioxidant capacity. Forty Wistar male rats were divided into five groups: dPBS (sham-operated control), dAβ (Aβ control), JPBS (F8 and PBS), JAβ (F8 and Aβ), and IBFAβ (ibuprofen and Aβ). F8 (5 ml/kg BW), and ibuprofen (10 ml/kg BW) was given orally daily for four weeks before the intracerebroventricular infusion of Aβ for two weeks. Histological analysis and neuronal count of hippocampus tissue in the Cornu Ammonis (CA1) region showed that supplementation with F8 was able to prevent Aβ-induced tissue damage and neuronal shrinkage. However, no significant difference in locomotor activity and novel object recognition (NOR) percentage was detected among different groups at day 7 and day 14 following Aβ infusion. Only effect of time differences (main effect of day) was observed at day 7 as compared to day 14, where reduction in locomotor activity and NOR percentage was observed in all groups, with F (1, 7) = 6.940, p < 0.05 and F (1, 7) = 7.152, p < 0.05, respectively. Besides, the MDA level of the JAβ group was significantly lower (p < 0.01) than that of the dPBS group. However, no significant changes in SOD activity were detected among different groups. Significant reduction in plasma CRH level (p < 0.05) and iNOS expression (p < 0.01) in the brain was detected in the JAβ group as compared to the dAβ group. Hence, our current findings suggest that the tropical fruit juice mixture (F8) has the potential to protect the rats from Aβ-induced neurotoxicity in brain hippocampus tissue possibly via its antioxidant properties and the suppression of iNOS expression and CRH production.
Collapse
Affiliation(s)
- Theng Choon Ooi
- Center for Healthy Ageing & Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Munirah Ahmad Munawar
- Biomedical Sciences Programme, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Nur Hasnieza Mohd Rosli
- Biomedical Sciences Programme, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Siti Nur Aqilah Abdul Malek
- Center for Healthy Ageing & Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Hanisah Rosli
- Center for Healthy Ageing & Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Farah Wahida Ibrahim
- Biomedical Sciences Programme, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
- Center for Toxicology and Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Norazrina Azmi
- Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Hasnah Haron
- Center for Healthy Ageing & Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Razinah Sharif
- Center for Healthy Ageing & Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Suzana Shahar
- Center for Healthy Ageing & Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Nor Fadilah Rajab
- Center for Healthy Ageing & Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
- Biomedical Sciences Programme, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| |
Collapse
|
32
|
Min LJ, Iwanami J, Shudou M, Bai HY, Shan BS, Higaki A, Mogi M, Horiuchi M. Deterioration of cognitive function after transient cerebral ischemia with amyloid-β infusion-possible amelioration of cognitive function by AT 2 receptor activation. J Neuroinflammation 2020; 17:106. [PMID: 32264971 PMCID: PMC7140348 DOI: 10.1186/s12974-020-01775-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 03/17/2020] [Indexed: 12/24/2022] Open
Abstract
Background To promote understanding of the pathogenesis of cognitive impairment or dementia, we explored the potential interaction between transient cerebral ischemia and amyloid-β (Aβ) infusion in mediating cognitive decline and examined the possible ameliorative effect of angiotensin II type 2 (AT2) receptor activation in vascular smooth muscle cells (VSMC) on this cognitive deficit. Methods Adult male wild-type mice (WT) and mice with VSMC-specific AT2 receptor overexpression (smAT2) were subjected to intracerebroventricular (ICV) injection of Aβ1-40. Transient cerebral ischemia was induced by 15 min of bilateral common carotid artery occlusion (BCCAO) 24 h after Aβ injection. Results Aβ injection in WT induced a cognitive decline, whereas BCCAO did not cause a significant cognitive deficit. In contrast, WT with BCCAO following Aβ injection exhibited more marked cognitive decline compared to Aβ injection alone, in concert with increases in superoxide anion production, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity, and expression of p22phox, p40phox, monocyte chemoattractant protein (MCP)-1 and interleukin (IL)-1β in the hippocampus, and upregulation of RAGE (receptor for advanced glycation end product), an Aβ transporter. BCCAO following Aβ injection further enhanced neuronal pyknosis in the hippocampus, compared with BCCAO or Aβ injection alone. In contrast, smAT2 did not show a cognitive decline, increase in oxidative stress, inflammation, and RAGE level or neuronal pyknosis, which were induced by BCCAO with/without Aβ injection in WT. Conclusions Transient cerebral ischemia might worsen Aβ infusion-mediated cognitive decline and vice versa, with possible involvement of amplified oxidative stress and inflammation and impairment of the RAGE-mediated Aβ clearance system, contributing to exaggerated neuronal degeneration. AT2 receptor activation in VSMC could play an inhibitory role in this cognitive deficit.
Collapse
Affiliation(s)
- Li-Juan Min
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Shitsukawa, Tohon, Ehime, 791-0295, Japan.
| | - Jun Iwanami
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Shitsukawa, Tohon, Ehime, 791-0295, Japan
| | - Masachika Shudou
- Division of Analytical Bio-Medicine, Advanced Research Support Center (ADRES), Ehime University, Graduate School of Medicine, Tohon, Ehime, 791-0295, Japan
| | - Hui-Yu Bai
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Shitsukawa, Tohon, Ehime, 791-0295, Japan
| | - Bao-Shuai Shan
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Shitsukawa, Tohon, Ehime, 791-0295, Japan
| | - Akinori Higaki
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Shitsukawa, Tohon, Ehime, 791-0295, Japan
| | - Masaki Mogi
- Department of Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, 791-0295, Japan
| | - Masatsugu Horiuchi
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Shitsukawa, Tohon, Ehime, 791-0295, Japan
| |
Collapse
|
33
|
Sharma S, Saini R, Sharma P, Saini A, Nehru B. Maintenance of Amyloid-beta Homeostasis by Carbenoxolone Post Aβ-42 Oligomer Injection in Rat Brain. Neuroscience 2020; 431:86-102. [DOI: 10.1016/j.neuroscience.2020.02.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 02/03/2020] [Accepted: 02/04/2020] [Indexed: 10/25/2022]
|
34
|
Barilar JO, Knezovic A, Perhoc AB, Homolak J, Riederer P, Salkovic-Petrisic M. Shared cerebral metabolic pathology in non-transgenic animal models of Alzheimer's and Parkinson's disease. J Neural Transm (Vienna) 2020; 127:231-250. [PMID: 32030485 PMCID: PMC7035309 DOI: 10.1007/s00702-020-02152-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 01/24/2020] [Indexed: 12/25/2022]
Abstract
Parkinson's disease (PD) and Alzheimer's disease (AD) are the most common chronic neurodegenerative disorders, characterized by motoric dysfunction or cognitive decline in the early stage, respectively, but often by both symptoms in the advanced stage. Among underlying molecular pathologies that PD and AD patients have in common, more attention is recently paid to the central metabolic dysfunction presented as insulin resistant brain state (IRBS) and altered cerebral glucose metabolism, both also explored in animal models of these diseases. This review aims to compare IRBS and alterations in cerebral glucose metabolism in representative non-transgenic animal PD and AD models. The comparison is based on the selectivity of the neurotoxins which cause experimental PD and AD, towards the cellular membrane and intracellular molecular targets as well as towards the selective neurons/non-neuronal cells, and the particular brain regions. Mitochondrial damage and co-expression of insulin receptors, glucose transporter-2 and dopamine transporter on the membrane of particular neurons as well as astrocytes seem to be the key points which are further discussed in a context of alterations in insulin signalling in the brain and its interaction with dopaminergic transmission, particularly regarding the time frame of the experimental AD/PD pathology appearance and the correlation with cognitive and motor symptoms. Such a perspective provides evidence on IRBS being a common underlying metabolic pathology and a contributor to neurodegenerative processes in representative non-transgenic animal PD and AD models, instead of being a direct cause of a particular neurodegenerative disorder.
Collapse
Affiliation(s)
- Jelena Osmanovic Barilar
- Department of Pharmacology, University of Zagreb School of Medicine, Salata 11, 10 000, Zagreb, Croatia
| | - Ana Knezovic
- Department of Pharmacology, University of Zagreb School of Medicine, Salata 11, 10 000, Zagreb, Croatia
| | - Ana Babic Perhoc
- Department of Pharmacology, University of Zagreb School of Medicine, Salata 11, 10 000, Zagreb, Croatia
| | - Jan Homolak
- Department of Pharmacology, University of Zagreb School of Medicine, Salata 11, 10 000, Zagreb, Croatia
| | - Peter Riederer
- Center of Mental Health, Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital, Würzburg, Füchsleinstrasse 15, 97080, Würzburg, Germany
- Department and Research Unit of Psychiatry, Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Melita Salkovic-Petrisic
- Department of Pharmacology, University of Zagreb School of Medicine, Salata 11, 10 000, Zagreb, Croatia.
- Institute of Fundamental Clinical and Translational Neuroscience, Research Centre of Excellence, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Salata 12, 10 000, Zagreb, Croatia.
| |
Collapse
|
35
|
The selective GSK3 inhibitor, SAR502250, displays neuroprotective activity and attenuates behavioral impairments in models of neuropsychiatric symptoms of Alzheimer's disease in rodents. Sci Rep 2019; 9:18045. [PMID: 31792284 PMCID: PMC6888874 DOI: 10.1038/s41598-019-54557-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 11/15/2019] [Indexed: 12/20/2022] Open
Abstract
Glycogen synthase kinase 3 (GSK3) has been identified as a promising target for the treatment of Alzheimer’s disease (AD), where abnormal activation of this enzyme has been associated with hyperphosphorylation of tau proteins. This study describes the effects of the selective GSK3 inhibitor, SAR502250, in models of neuroprotection and neuropsychiatric symptoms (NPS) associated with AD. In P301L human tau transgenic mice, SAR502250 attenuated tau hyperphosphorylation in the cortex and spinal cord. SAR502250 prevented the increase in neuronal cell death in rat embryonic hippocampal neurons following application of the neurotoxic peptide, Aβ25–35. In behavioral studies, SAR502250 improved the cognitive deficit in aged transgenic APP(SW)/Tau(VLW) mice or in adult mice after infusion of Aβ25–35. It attenuated aggression in the mouse defense test battery and improved depressive-like state of mice in the chronic mild stress procedure after 4 weeks of treatment. Moreover, SAR502250 decreased hyperactivity produced by psychostimulants. In contrast, the drug failed to modify anxiety-related behaviors or sensorimotor gating deficit. This profile confirms the neuroprotective effects of GSK3 inhibitors and suggests an additional potential in the treatment of some NPS associated with AD.
Collapse
|
36
|
Canet G, Pineau F, Zussy C, Hernandez C, Hunt H, Chevallier N, Perrier V, Torrent J, Belanoff JK, Meijer OC, Desrumaux C, Givalois L. Glucocorticoid receptors signaling impairment potentiates amyloid-β oligomers-induced pathology in an acute model of Alzheimer's disease. FASEB J 2019; 34:1150-1168. [PMID: 31914623 DOI: 10.1096/fj.201900723rrr] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 11/05/2019] [Accepted: 11/07/2019] [Indexed: 02/01/2023]
Abstract
Dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis occurs early in Alzheimer's disease (AD), associated with elevated circulating glucocorticoids (GC) and glucocorticoid receptors (GR) signaling impairment. However, the precise role of GR in the pathophysiology of AD remains unclear. Using an acute model of AD induced by the intracerebroventricular injection of amyloid-β oligomers (oAβ), we analyzed cellular and behavioral hallmarks of AD, GR signaling pathways, processing of amyloid precursor protein, and enzymes involved in Tau phosphorylation. We focused on the prefrontal cortex (PFC), particularly rich in GR, early altered in AD and involved in HPA axis control and cognitive functions. We found that oAβ impaired cognitive and emotional behaviors, increased plasma GC levels, synaptic deficits, apoptosis and neuroinflammatory processes. Moreover, oAβ potentiated the amyloidogenic pathway and enzymes involved both in Tau hyperphosphorylation and GR activation. Treatment with a selective GR modulator (sGRm) normalized plasma GC levels and all behavioral and biochemical parameters analyzed. GR seems to occupy a central position in the pathophysiology of AD. Deregulation of the HPA axis and a feed-forward effect on PFC GR sensitivity could participate in the etiology of AD, in perturbing Aβ and Tau homeostasis. These results also reinforce the therapeutic potential of sGRm in AD.
Collapse
Affiliation(s)
- Geoffrey Canet
- Molecular Mechanisms in Neurodegenerative Dementia (MMDN) Laboratory, INSERM U1198, Team Environmental Impact in Alzheimer's Disease and Related Disorders (EiAlz), Montpellier, France.,University of Montpellier, Montpellier, France.,EPHE, Paris, France
| | - Fanny Pineau
- Molecular Mechanisms in Neurodegenerative Dementia (MMDN) Laboratory, INSERM U1198, Team Environmental Impact in Alzheimer's Disease and Related Disorders (EiAlz), Montpellier, France.,University of Montpellier, Montpellier, France.,EPHE, Paris, France
| | - Charleine Zussy
- Molecular Mechanisms in Neurodegenerative Dementia (MMDN) Laboratory, INSERM U1198, Team Environmental Impact in Alzheimer's Disease and Related Disorders (EiAlz), Montpellier, France.,University of Montpellier, Montpellier, France.,EPHE, Paris, France
| | - Célia Hernandez
- Molecular Mechanisms in Neurodegenerative Dementia (MMDN) Laboratory, INSERM U1198, Team Environmental Impact in Alzheimer's Disease and Related Disorders (EiAlz), Montpellier, France.,University of Montpellier, Montpellier, France.,EPHE, Paris, France
| | - Hazel Hunt
- Corcept Therapeutics, Menlo Park, CA, USA
| | - Nathalie Chevallier
- Molecular Mechanisms in Neurodegenerative Dementia (MMDN) Laboratory, INSERM U1198, Team Environmental Impact in Alzheimer's Disease and Related Disorders (EiAlz), Montpellier, France.,University of Montpellier, Montpellier, France.,EPHE, Paris, France
| | - Véronique Perrier
- Molecular Mechanisms in Neurodegenerative Dementia (MMDN) Laboratory, INSERM U1198, Team Environmental Impact in Alzheimer's Disease and Related Disorders (EiAlz), Montpellier, France.,University of Montpellier, Montpellier, France.,EPHE, Paris, France
| | - Joan Torrent
- Molecular Mechanisms in Neurodegenerative Dementia (MMDN) Laboratory, INSERM U1198, Team Environmental Impact in Alzheimer's Disease and Related Disorders (EiAlz), Montpellier, France.,University of Montpellier, Montpellier, France.,EPHE, Paris, France
| | | | - Onno C Meijer
- Einthoven Laboratory, Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
| | - Catherine Desrumaux
- Molecular Mechanisms in Neurodegenerative Dementia (MMDN) Laboratory, INSERM U1198, Team Environmental Impact in Alzheimer's Disease and Related Disorders (EiAlz), Montpellier, France.,University of Montpellier, Montpellier, France.,EPHE, Paris, France
| | - Laurent Givalois
- Molecular Mechanisms in Neurodegenerative Dementia (MMDN) Laboratory, INSERM U1198, Team Environmental Impact in Alzheimer's Disease and Related Disorders (EiAlz), Montpellier, France.,University of Montpellier, Montpellier, France.,EPHE, Paris, France
| |
Collapse
|
37
|
Scheiner M, Dolles D, Gunesch S, Hoffmann M, Nabissi M, Marinelli O, Naldi M, Bartolini M, Petralla S, Poeta E, Monti B, Falkeis C, Vieth M, Hübner H, Gmeiner P, Maitra R, Maurice T, Decker M. Dual-Acting Cholinesterase-Human Cannabinoid Receptor 2 Ligands Show Pronounced Neuroprotection in Vitro and Overadditive and Disease-Modifying Neuroprotective Effects in Vivo. J Med Chem 2019; 62:9078-9102. [PMID: 31609608 PMCID: PMC7640639 DOI: 10.1021/acs.jmedchem.9b00623] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We have designed and synthesized a series of 14 hybrid molecules out of the cholinesterase (ChE) inhibitor tacrine and a benzimidazole-based human cannabinoid receptor subtype 2 (hCB2R) agonist and investigated them in vitro and in vivo. The compounds are potent ChE inhibitors, and for the most promising hybrids, the mechanism of human acetylcholinesterase (hAChE) inhibition as well as their ability to interfere with AChE-induced aggregation of β-amyloid (Aβ), and Aβ self-aggregation was assessed. All hybrids were evaluated for affinity and selectivity for hCB1R and hCB2R. To ensure that the hybrids retained their agonist character, the expression of cAMP-regulated genes was quantified, and potency and efficacy were determined. Additionally, the effects of the hybrids on microglia activation and neuroprotection on HT-22 cells were investigated. The most promising in vitro hybrids showed pronounced neuroprotection in an Alzheimer's mouse model at low dosage (0.1 mg/kg, i.p.), lacking hepatotoxicity even at high dose (3 mg/kg, i.p.).
Collapse
Affiliation(s)
- Matthias Scheiner
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Dominik Dolles
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Sandra Gunesch
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Matthias Hoffmann
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Massimo Nabissi
- School of Pharmacy, University of Camerino, Via Madonna delle Carceri 9, 62032 Camerino, Italy
| | - Oliviero Marinelli
- School of Pharmacy, University of Camerino, Via Madonna delle Carceri 9, 62032 Camerino, Italy
| | - Marina Naldi
- Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Manuela Bartolini
- Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Sabrina Petralla
- Department of Pharmacy and Biotechnology, University of Bologna, Via Selmi 3, 40126 Bologna, Italy
| | - Eleonora Poeta
- Department of Pharmacy and Biotechnology, University of Bologna, Via Selmi 3, 40126 Bologna, Italy
| | - Barbara Monti
- Department of Pharmacy and Biotechnology, University of Bologna, Via Selmi 3, 40126 Bologna, Italy
| | - Christina Falkeis
- Pathology, Clinical Center Bayreuth, Preuschwitzer Straße 101, 95445 Bayreuth, Germany
| | - Michael Vieth
- Pathology, Clinical Center Bayreuth, Preuschwitzer Straße 101, 95445 Bayreuth, Germany
| | - Harald Hübner
- Medicinal Chemistry, Department of Chemistry and Pharmacy, Friedrich-Alexander University Erlangen-Nürnberg, Schuhstraße 19, 91052 Erlangen, Germany
| | - Peter Gmeiner
- Medicinal Chemistry, Department of Chemistry and Pharmacy, Friedrich-Alexander University Erlangen-Nürnberg, Schuhstraße 19, 91052 Erlangen, Germany
| | - Rangan Maitra
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina 27709, United States
| | - Tangui Maurice
- MMDN, University of Montpellier, INSERM, EPHE, UMR-S1198, 34095 Montpellier, France
| | - Michael Decker
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| |
Collapse
|
38
|
White JD, Urbano CM, Taylor JO, Peterman JL, Cooksey M, Eimerbrink M, Eriksson MD, Cooper BG, Chumley MJ, Boehm GW. Intraventricular murine Aβ infusion elicits hippocampal inflammation and disrupts the consolidation, but not retrieval, of conditioned fear in C57BL6/J mice. Behav Brain Res 2019; 378:112303. [PMID: 31622640 DOI: 10.1016/j.bbr.2019.112303] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 09/11/2019] [Accepted: 10/13/2019] [Indexed: 01/06/2023]
Abstract
Although one of the defining characteristics of Alzheimer's disease is the presence of amyloid-beta (Aβ) plaques, the early accumulation of soluble Aβ oligomers (AβOs) may disrupt synaptic function and trigger cognitive impairments long before the appearance of plaques. Furthermore, murine models aimed at understanding how AβOs alter formation and retrieval of associative memories are conducted using human Aβ species, which are more neurotoxic in the mouse brain than the native murine species. Unfortunately, there is currently a lack of attention in the literature as to what the murine version of the peptide (mAβ) does to synaptic function and how it impacts the consolidation and retrieval of associative memories. In the current study, adult mice were infused with mAβ 0, 2, 6, or 46 h after contextual-fear conditioning, and were tested 2-48 h later. Interestingly, only mAβ infusions within 2 h of training reduced freezing behavior at test, indicating that mAβ disrupted the consolidation, but not retrieval of fear memory. This consolidation deficit coincided with increased IL-1β and reduced synaptophysin mRNA levels, without disrupting other synaptic signaling-related genes here examined. Despite differences between murine and human Aβ, the deleterious functional outcomes of early-stage synaptic oligomer presence are similar. Thus, models utilizing or inducing the production of mAβ in non-transgenic animals are useful in exploring the role of dysregulated synaptic plasticity and resultant learning deficits induced by Aβ oligomers.
Collapse
Affiliation(s)
- J D White
- The Department of Psychology, Texas Christian University, Fort Worth, Texas, United States
| | - C M Urbano
- The Department of Psychology, Texas Christian University, Fort Worth, Texas, United States
| | - J O Taylor
- The Department of Psychology, Texas Christian University, Fort Worth, Texas, United States
| | - J L Peterman
- The Department of Psychology, Texas Christian University, Fort Worth, Texas, United States
| | - M Cooksey
- The Department of Psychology, Texas Christian University, Fort Worth, Texas, United States
| | - M Eimerbrink
- The Department of Psychology, Texas Christian University, Fort Worth, Texas, United States
| | - M D Eriksson
- The Department of Psychology, Texas Christian University, Fort Worth, Texas, United States
| | - B G Cooper
- The Department of Psychology, Texas Christian University, Fort Worth, Texas, United States
| | - M J Chumley
- The Department of Biology, Texas Christian University, Fort Worth, Texas, United States
| | - G W Boehm
- The Department of Psychology, Texas Christian University, Fort Worth, Texas, United States.
| |
Collapse
|
39
|
Ruan L, Du K, Tao M, Shan C, Ye R, Tang Y, Pan H, Lv J, Zhang M, Pan J. Phosphodiesterase-2 Inhibitor Bay 60-7550 Ameliorates Aβ-Induced Cognitive and Memory Impairment via Regulation of the HPA Axis. Front Cell Neurosci 2019; 13:432. [PMID: 31632240 PMCID: PMC6783519 DOI: 10.3389/fncel.2019.00432] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 09/09/2019] [Indexed: 01/01/2023] Open
Abstract
The dysfunction of the hypothalamus-pituitary-adrenal (HPA) axis is often seen in Alzheimer's disease (AD) patients with cognitive deficits. Selective inhibition of phosphodiesterase (PDE) 4 and 5 has already proven to be effective in reducing beta-amyloid 1-42 (Aβ1-42)-mediated pathology by regulating corticotropin-releasing factor (CRF) and glucocorticoid receptor (GR) expression, suggesting that PDE-dependent signaling is involved in Aβ1-42-induced HPA axis dysfunction. However, nausea and vomiting are the side effects of some PDE4 inhibitors, which turn our attention to other PDEs. PDE2 are highly expressed in the hippocampus and cortex, which associate with learning and memory, but not in the area postrema that would cause vomiting. The present study suggested that microinjection of Aβ1-42 to the intracerebroventricle induced learning and memory impairments and dysregulation of the HPA axis by increased expression of CRF and GR. However, the PDE2 inhibitor Bay 60-7550 significantly ameliorated the learning and memory impairment in the Morris water maze (MWM) and step-down passive avoidance tests. The Aβ1-42-induced increased CRF and GR levels were also reversed by the treatment with Bay 60-7550. These Bay 60-7550's effects were prevented by pretreatment with the PKG inhibitor KT5823. Moreover, the Bay 60-7550-induced downstream phosphorylation of cyclic AMP response element binding (pCREB) and brain-derived neurotrophic factor (BDNF) expression was also prevented (or partially prevented) by KT5823 or the PKA inhibitor H89. These results may lead to the discovery of novel strategies for the treatment of age-related cognitive disorders, such as AD, which affects approximately 44 million people worldwide.
Collapse
Affiliation(s)
- Lina Ruan
- Brain Institute, School of Pharmacy, Wenzhou Medical University, Wenzhou, China
| | - Kai Du
- Brain Institute, School of Pharmacy, Wenzhou Medical University, Wenzhou, China
| | - Mengjia Tao
- Brain Institute, School of Pharmacy, Wenzhou Medical University, Wenzhou, China
| | - Chunyan Shan
- Brain Institute, School of Pharmacy, Wenzhou Medical University, Wenzhou, China
| | - Ruixuan Ye
- Brain Institute, School of Pharmacy, Wenzhou Medical University, Wenzhou, China
| | - Yali Tang
- Brain Institute, School of Pharmacy, Wenzhou Medical University, Wenzhou, China
| | - Hanbo Pan
- Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Jinpeng Lv
- College of Pharmaceutical Engineering and Life Sciences, Changzhou University, Changzhou, China
| | - Meixi Zhang
- Brain Institute, School of Pharmacy, Wenzhou Medical University, Wenzhou, China.,Pingyang County Hospital of Traditional Chinese Medicine, Pingyang County, China
| | - Jianchun Pan
- Brain Institute, School of Pharmacy, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
40
|
Dmytriyeva O, Belmeguenai A, Bezin L, Soud K, Drucker Woldbye DP, Gøtzsche CR, Pankratova S. Short erythropoietin-derived peptide enhances memory, improves long-term potentiation, and counteracts amyloid beta–induced pathology. Neurobiol Aging 2019; 81:88-101. [DOI: 10.1016/j.neurobiolaging.2019.05.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 03/27/2019] [Accepted: 05/06/2019] [Indexed: 12/23/2022]
|
41
|
Patricio-Martínez A, Sánchez-Zavaleta R, Angulo-Cruz I, Gutierrez-Praxedis L, Ramírez E, Martínez-García I, Limón ID. The Acute Activation of the CB1 Receptor in the Hippocampus Decreases Neurotoxicity and Prevents Spatial Memory Impairment in Rats Lesioned with β-Amyloid 25-35. Neuroscience 2019; 416:239-254. [PMID: 31400487 DOI: 10.1016/j.neuroscience.2019.08.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 12/23/2022]
Abstract
Given their anti-inflammatory properties, cannabinoids have been shown to be neuroprotective agents and to reduce excitotoxicity, through the activation of the Cannabinoid receptor type 1 (CB1r). These properties have led to CB1r being proposed as pharmacological targets for the treatment of various neurodegenerative diseases. Amyloid-β 25-35 (Aβ25-35) induces the expression of inducible nitric oxide synthase (iNOS) and increases nitric oxide (NO●) levels. It has been observed that increased NO● concentrations trigger biochemical pathways that contribute to neuronal death and cognitive damage. This study aimed to evaluate the neuroprotective effect of an acute activation of CB1r on spatial memory and its impact on iNOS protein expression, NO● levels, gliosis and the neurodegenerative process induced by the injection of Aβ(25-35) into the CA1 subfield of the hippocampus. ACEA [1 μM/1 μL] and Aβ(25-35) [100 μM/1 μL] and their respective vehicle groups were injected into the CA1 subfield of the hippocampus. The animals were tested for spatial learning and memory in the eight-arm radial maze, with the results revealing that the administration of ACEA plus Aβ(25-35) improves learning and memory processes, in contrast with the Aβ(25-35) group. Moreover, ACEA plus Aβ(25-35) prevented both the increase in iNOS protein and NO● levels and the reactive gliosis induced by Aβ(25-35). Importantly, neurodegeneration was significantly reduced by the administration of ACEA plus Aβ(25-35) in the CA1 subfield of the hippocampus. The data obtained in the present research suggest that the acute early activation of CB1r is crucial for neuroprotection.
Collapse
Affiliation(s)
- Aleidy Patricio-Martínez
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas-Benemérita Universidad Autónoma de Puebla, Puebla, Mexico; Facultad de Ciencias Biológicas-Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Rodolfo Sánchez-Zavaleta
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas-Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Isael Angulo-Cruz
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas-Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Liliana Gutierrez-Praxedis
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas-Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Eleazar Ramírez
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas-Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Isabel Martínez-García
- Laboratorio de Neuroquímica, Facultad de Ciencias Químicas-Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Ilhuicamina Daniel Limón
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas-Benemérita Universidad Autónoma de Puebla, Puebla, Mexico.
| |
Collapse
|
42
|
Umar T, Shalini S, Raza MK, Gusain S, Kumar J, Seth P, Tiwari M, Hoda N. A multifunctional therapeutic approach: Synthesis, biological evaluation, crystal structure and molecular docking of diversified 1H-pyrazolo[3,4-b]pyridine derivatives against Alzheimer's disease. Eur J Med Chem 2019; 175:2-19. [DOI: 10.1016/j.ejmech.2019.04.038] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/03/2019] [Accepted: 04/13/2019] [Indexed: 11/26/2022]
|
43
|
Clarke JR, Ribeiro FC, Frozza RL, De Felice FG, Lourenco MV. Metabolic Dysfunction in Alzheimer's Disease: From Basic Neurobiology to Clinical Approaches. J Alzheimers Dis 2019; 64:S405-S426. [PMID: 29562518 DOI: 10.3233/jad-179911] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Clinical trials have extensively failed to find effective treatments for Alzheimer's disease (AD) so far. Even after decades of AD research, there are still limited options for treating dementia. Mounting evidence has indicated that AD patients develop central and peripheral metabolic dysfunction, and the underpinnings of such events have recently begun to emerge. Basic and preclinical studies have unveiled key pathophysiological mechanisms that include aberrant brain stress signaling, inflammation, and impaired insulin sensitivity. These findings are in accordance with clinical and neuropathological data suggesting that AD patients undergo central and peripheral metabolic deregulation. Here, we review recent basic and clinical findings indicating that metabolic defects are central to AD pathophysiology. We further propose a view for future therapeutics that incorporates metabolic defects as a core feature of AD pathogenesis. This approach could improve disease understanding and therapy development through drug repurposing and/or identification of novel metabolic targets.
Collapse
Affiliation(s)
- Julia R Clarke
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Felipe C Ribeiro
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rudimar L Frozza
- Oswaldo Cruz Institute, Oswaldo Cruz Foundation, FIOCRUZ, Rio de Janeiro, Brazil
| | - Fernanda G De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Mychael V Lourenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
44
|
Rossi Dare L, Garcia A, Alves N, Ventura Dias D, de Souza MA, Mello-Carpes PB. Physical and cognitive training are able to prevent recognition memory deficits related to amyloid beta neurotoxicity. Behav Brain Res 2019; 365:190-197. [DOI: 10.1016/j.bbr.2019.03.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 02/19/2019] [Accepted: 03/02/2019] [Indexed: 12/13/2022]
|
45
|
Ramírez E, Sánchez-Maldonado C, Mayoral MA, Mendieta L, Alatriste V, Patricio-Martínez A, Limón ID. Neuroinflammation induced by the peptide amyloid-β (25-35) increase the presence of galectin-3 in astrocytes and microglia and impairs spatial memory. Neuropeptides 2019; 74:11-23. [PMID: 30795916 DOI: 10.1016/j.npep.2019.02.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 02/10/2019] [Accepted: 02/12/2019] [Indexed: 01/05/2023]
Abstract
Galectins are animal lectins that bind to β-galactosides, such as lactose and N-acetyllactosamine, contained in glycoproteins or glycolipids. Galectin-1 (Gal-1) and Galectin-3 (Gal-3) are involved in pathologies associated with the inflammatory process, cell proliferation, adhesion, migration, and apoptosis. Recent evidence has shown that the administration of Amyloid-β 25-35 (Aβ25-35) into the hippocampus of rats increases the inflammatory response that is associated with memory impairment and neurodegeneration. Galectins could participate in the modulation of the neuroinflammation induced by the Aβ25-35. The aim of this study was to evaluate the presence of Gal-1 and Gal-3 in the neuroinflammation induced by administration of Aβ25-35 into the hippocampus and to examine spatial memory in the Morris water maze. After the administration of Aβ25-35, animals were tested for learning and spatial memory in the Morris water maze. Behavioral performance showed that Aβ25-35 didn't affect spatial learning but did impair memory, with animals taking longer to find the platform. On the day 32, hippocampus was examined for astrocytes (GFAP), microglia (Iba1), Gal-1 and Gal-3 via immunohistochemical analysis, and the cytokines IL-1β, TNF-α, IFN-γ by ELISA. This study's results showed a significant increase in the expression of Gal-3 in the microglia and astrocytes, while Gal-1 didn't increase in the dorsal hippocampus. The expression of galectins is associated with increased cytokines in the hippocampal formation of Aβ25-35 treated rats. These findings suggest that Gal-3 could participate in the inflammation induced by administration of Aβ25-35 and could be involved in the neurodegeneration progress and memory impairment.
Collapse
Affiliation(s)
- Eleazar Ramírez
- Laboratorio de Neurofarmacología, 105 C-FCQ BUAP, Puebla, Mexico
| | | | | | - Liliana Mendieta
- Laboratorio de Neurofarmacología, 105 C-FCQ BUAP, Puebla, Mexico
| | | | - Aleidy Patricio-Martínez
- Laboratorio de Neurofarmacología, 105 C-FCQ BUAP, Puebla, Mexico; Facultad de Ciencias Biológicas, BUAP, Puebla, Mexico
| | - I Daniel Limón
- Laboratorio de Neurofarmacología, 105 C-FCQ BUAP, Puebla, Mexico.
| |
Collapse
|
46
|
Eskandari-Roozbahani N, Shomali T, Taherianfard M. Neuroprotective Effect of Zataria Multiflora Essential Oil on Rats With Alzheimer Disease: A Mechanistic Study. Basic Clin Neurosci 2019; 10:85-97. [PMID: 31031896 PMCID: PMC6484186 DOI: 10.32598/bcn.9.10.270] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 02/05/2018] [Accepted: 05/26/2018] [Indexed: 11/20/2022] Open
Abstract
INTRODUCTION Finding herbs with promising effects to prevent or postpone Alzheimer Disease (AD) is highly demanded. The present study aimed at clarifying plausible effects and related mechanism(s) of Zataria Multiflora Essential Oil (ZMEO) against memory impairment in a rat model of the AD. METHODS Forty male adult rats were categorized into four groups and treated as follows: 1. The Negative Control (NC): no treatment; 2. Sham control (sham): distilled water by Intracerebroventricular (ICV) injection; 3. The AD control (AD): Aβ 1-42 by ICV injection; and 4. The ZMEO group: Aβ 1-42 by ICV injection and ZMEO at 100 μL/kg/d orally for 20 days. RESULTS After Congo red staining of the hippocampus, a relative decrease in amyloid deposits was observed in the ZMEO group. Moreover, rats showed better outcomes in Morris Water Maze (MWM) test, reduced hippocampal acetylcholinesterase (AchE) activity, and higher Brain-Derived Neurotrophic Factor (BDNF) content as compared with the AD group (P<0.05). However, no significant changes in antioxidant status was observed (P>0.05). CONCLUSION ZMEO has a protective effect against memory impairment in rats with AD at least partly via reducing hippocampal AchE activity and enhancement of BDNF levels without a change in antioxidant status. These findings can pave the way for future studies on the usefulness of this herb in AD prevention.
Collapse
Affiliation(s)
| | - Tahoora Shomali
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Mahnaz Taherianfard
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| |
Collapse
|
47
|
Canet G, Chevallier N, Zussy C, Desrumaux C, Givalois L. Central Role of Glucocorticoid Receptors in Alzheimer's Disease and Depression. Front Neurosci 2018; 12:739. [PMID: 30459541 PMCID: PMC6232776 DOI: 10.3389/fnins.2018.00739] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 09/25/2018] [Indexed: 01/21/2023] Open
Abstract
Alzheimer’s disease (AD) is the principal neurodegenerative pathology in the world displaying negative impacts on both the health and social ability of patients and inducing considerable economic costs. In the case of sporadic forms of AD (more than 95% of patients), even if mechanisms are unknown, some risk factors were identified. The principal risk is aging, but there is growing evidence that lifetime events like chronic stress or stress-related disorders may increase the probability to develop AD. This mini-review reinforces the rationale to consider major depressive disorder (MDD) as an important risk factor to develop AD and points the central role played by the hypothalamic-pituitary-adrenal (HPA) axis, glucocorticoids (GC) and their receptors (GR) in the etiology of MDD and AD. Several strategies directly targeting GR were tested to neutralize the HPA axis dysregulation and GC overproduction. Given the ubiquitous expression of GR, antagonists have many undesired side effects, limiting their therapeutic potential. However, a new class of molecules was developed, highly selective and acting as modulators. They present the advantage to selectively abrogate pathogenic GR-dependent processes, while retaining beneficial aspects of GR signaling. In fact, these “selective GR modulators” induce a receptor conformation that allows activation of only a subset of downstream signaling pathways, explaining their capacity to combine agonistic and antagonistic properties. Thus, targeting GR with selective modulators, alone or in association with current strategies, becomes particularly attractive and relevant to develop novel preventive and/or therapeutic strategies to tackle disorders associated with a dysregulation of the HPA axis.
Collapse
Affiliation(s)
- Geoffrey Canet
- Molecular Mechanisms in Neurodegenerative Dementia Laboratory, INSERM, U1198, Team Environmental Impact in Alzheimer's Disease and Related Disorders (EiAlz), Montpellier, France.,University of Montpellier, Montpellier, France.,EPHE, Paris, France
| | - Nathalie Chevallier
- Molecular Mechanisms in Neurodegenerative Dementia Laboratory, INSERM, U1198, Team Environmental Impact in Alzheimer's Disease and Related Disorders (EiAlz), Montpellier, France.,University of Montpellier, Montpellier, France.,EPHE, Paris, France
| | - Charleine Zussy
- Molecular Mechanisms in Neurodegenerative Dementia Laboratory, INSERM, U1198, Team Environmental Impact in Alzheimer's Disease and Related Disorders (EiAlz), Montpellier, France.,University of Montpellier, Montpellier, France.,EPHE, Paris, France
| | - Catherine Desrumaux
- Molecular Mechanisms in Neurodegenerative Dementia Laboratory, INSERM, U1198, Team Environmental Impact in Alzheimer's Disease and Related Disorders (EiAlz), Montpellier, France.,University of Montpellier, Montpellier, France.,EPHE, Paris, France
| | - Laurent Givalois
- Molecular Mechanisms in Neurodegenerative Dementia Laboratory, INSERM, U1198, Team Environmental Impact in Alzheimer's Disease and Related Disorders (EiAlz), Montpellier, France.,University of Montpellier, Montpellier, France.,EPHE, Paris, France
| |
Collapse
|
48
|
Morroni F, Sita G, Graziosi A, Turrini E, Fimognari C, Tarozzi A, Hrelia P. Protective Effects of 6-(Methylsulfinyl)hexyl Isothiocyanate on Aβ 1-42-Induced Cognitive Deficit, Oxidative Stress, Inflammation, and Apoptosis in Mice. Int J Mol Sci 2018; 19:E2083. [PMID: 30021941 PMCID: PMC6073905 DOI: 10.3390/ijms19072083] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/10/2018] [Accepted: 07/16/2018] [Indexed: 01/12/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia among older people. Although soluble amyloid species are recognized triggers of the disease, no therapeutic approach is able to stop it. 6-(Methylsulfinyl)hexyl isothiocyanate (6-MSITC) is a major bioactive compound in Wasabia japonica, which is a typical Japanese pungent spice. Recently, in vivo and in vitro studies demonstrated that 6-MSITC has several biological properties. The aim of the present study was to investigate the neuroprotective activity of 6-MSITC in a murine AD model, induced by intracerebroventricular injection of β-amyloid oligomers (Aβ1-42O). The treatment with 6-MSITC started 1 h after the surgery for the next 10 days. Behavioral analysis showed that 6-MSITC ameliorated Aβ1-42O-induced memory impairments. The decrease of glutathione levels and increase of reactive oxygen species in hippocampal tissues following Aβ1-42O injection were reduced by 6-MSITC. Moreover, activation of caspases, increase of inflammatory factors, and phosphorylation of ERK and GSK3 were inhibited by 6-MSITC. These results highlighted an interesting neuroprotective activity of 6-MSITC, which was able to restore a physiological oxidative status, interfere positively with Nrf2-pathway, decrease apoptosis and neuroinflammation and contribute to behavioral recovery. Taken together, these findings demonstrated that 6-MSITC could be a promising complement for AD therapy.
Collapse
Affiliation(s)
- Fabiana Morroni
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, via Irnerio 48, 40126 Bologna, Italy.
| | - Giulia Sita
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, via Irnerio 48, 40126 Bologna, Italy.
| | - Agnese Graziosi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, via Irnerio 48, 40126 Bologna, Italy.
| | - Eleonora Turrini
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, Corso d'Augusto, 237, 47900 Rimini, Italy.
| | - Carmela Fimognari
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, Corso d'Augusto, 237, 47900 Rimini, Italy.
| | - Andrea Tarozzi
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, Corso d'Augusto, 237, 47900 Rimini, Italy.
| | - Patrizia Hrelia
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, via Irnerio 48, 40126 Bologna, Italy.
| |
Collapse
|
49
|
Prevot V, Dehouck B, Sharif A, Ciofi P, Giacobini P, Clasadonte J. The Versatile Tanycyte: A Hypothalamic Integrator of Reproduction and Energy Metabolism. Endocr Rev 2018; 39:333-368. [PMID: 29351662 DOI: 10.1210/er.2017-00235] [Citation(s) in RCA: 171] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 01/12/2018] [Indexed: 12/16/2022]
Abstract
The fertility and survival of an individual rely on the ability of the periphery to promptly, effectively, and reproducibly communicate with brain neural networks that control reproduction, food intake, and energy homeostasis. Tanycytes, a specialized glial cell type lining the wall of the third ventricle in the median eminence of the hypothalamus, appear to act as the linchpin of these processes by dynamically controlling the secretion of neuropeptides into the portal vasculature by hypothalamic neurons and regulating blood-brain and blood-cerebrospinal fluid exchanges, both processes that depend on the ability of these cells to adapt their morphology to the physiological state of the individual. In addition to their barrier properties, tanycytes possess the ability to sense blood glucose levels, and play a fundamental and active role in shuttling circulating metabolic signals to hypothalamic neurons that control food intake. Moreover, accumulating data suggest that, in keeping with their putative descent from radial glial cells, tanycytes are endowed with neural stem cell properties and may respond to dietary or reproductive cues by modulating hypothalamic neurogenesis. Tanycytes could thus constitute the missing link in the loop connecting behavior, hormonal changes, signal transduction, central neuronal activation and, finally, behavior again. In this article, we will examine these recent advances in the understanding of tanycytic plasticity and function in the hypothalamus and the underlying molecular mechanisms. We will also discuss the putative involvement and therapeutic potential of hypothalamic tanycytes in metabolic and fertility disorders.
Collapse
Affiliation(s)
- Vincent Prevot
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| | - Bénédicte Dehouck
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| | - Ariane Sharif
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| | - Philippe Ciofi
- Inserm, Neurocentre Magendie, Bordeaux, France.,Université de Bordeaux, Bordeaux, France
| | - Paolo Giacobini
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| | - Jerome Clasadonte
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| |
Collapse
|
50
|
Perillyl alcohol alleviates amyloid-β peptides-induced mitochondrial dysfunction and cytotoxicity in SH-SY5Y cells. Int J Biol Macromol 2018; 109:1029-1038. [DOI: 10.1016/j.ijbiomac.2017.11.082] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/21/2017] [Accepted: 11/13/2017] [Indexed: 12/20/2022]
|