1
|
Zago AM, Carvalho FB, Rahmeier FL, Santin M, Guimarães GR, Gutierres JM, da C Fernandes M. Exendin-4 Prevents Memory Loss and Neuronal Death in Rats with Sporadic Alzheimer-Like Disease. Mol Neurobiol 2024; 61:2631-2652. [PMID: 37919602 DOI: 10.1007/s12035-023-03698-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 10/06/2023] [Indexed: 11/04/2023]
Abstract
This study investigated the neuroprotective effects of exendin-4 (EXE-4), an analog of the glucagon-like peptide 1 receptor (GLP-1R) on memory and on the neuronal populations that constitute the hippocampus of rats submitted to a sporadic dementia of Alzheimer's type (SDAT). Male Wistar rats received streptozotocin (STZ icv, 3 mg/kg diluted in aCFS, 5 µl/ventricle) and were treated for 21 days with EXE-4 (10 µg/kg, ip; saline as the vehicle). Four groups were formed: vehicle, EXE-4, STZ, and STZ + EXE-4. The groups were submitted to Y-Maze (YM), object recognition (ORT), and object displacement tasks (ODT) to assess learning and memory. The brains were used for immunohistochemical and immunofluorescent techniques with antibodies to NeuN, cleaved caspase-3 (CC3), PCNA, doublecortin (DCX), synaptophysin (SYP), and insulin receptor (IR). STZ worsened spatial memory in the YMT, as well as short-term (STM) and long-term (LTM) memories in the ORT and ODT, respectively. EXE-4 protected against memory impairment in STZ animals. STZ reduced mature neuron density (NeuN) and increased cell apoptosis (CC3) in the DG, CA1, and CA3. EXE-4 protected against neuronal death in all regions. EXE-4 increased PCNA+ cells in all regions of the hippocampus, and STZ attenuated this effect. STZ reduced neurogenesis in DG per se as well as synaptogenesis induced by EXE-4. EXE-4 increased immunoreactivity to IR in the CA1. From these findings, EXE-4 showed a beneficial effect on hippocampal pyramidal and granular neurons in the SDAT showing anti-apoptotic properties and promoting cell proliferation. In parallel, EXE-4 preserved the memory of SDAT rats. EXE-4 appears to enhance synapses at CA3 and DG. In conclusion, these data indicate that agonists to GLP-1R have a beneficial effect on hippocampal neurons in AD.
Collapse
Affiliation(s)
- Adriana M Zago
- Graduate Program in Pathology, Pathology Research Laboratory, Federal University of Health Sciences of Porto Alegre, Sarmento Leite, 245, Room 514, Building 3. CEP 90050-170, Porto Alegre, RS, Brazil
| | - Fabiano B Carvalho
- Graduate Program in Pathology, Pathology Research Laboratory, Federal University of Health Sciences of Porto Alegre, Sarmento Leite, 245, Room 514, Building 3. CEP 90050-170, Porto Alegre, RS, Brazil.
| | - Francine L Rahmeier
- Graduate Program in Pathology, Pathology Research Laboratory, Federal University of Health Sciences of Porto Alegre, Sarmento Leite, 245, Room 514, Building 3. CEP 90050-170, Porto Alegre, RS, Brazil
| | - Marta Santin
- Graduate Program in Pathology, Pathology Research Laboratory, Federal University of Health Sciences of Porto Alegre, Sarmento Leite, 245, Room 514, Building 3. CEP 90050-170, Porto Alegre, RS, Brazil
| | - Giuliano R Guimarães
- Graduate Program in Pathology, Pathology Research Laboratory, Federal University of Health Sciences of Porto Alegre, Sarmento Leite, 245, Room 514, Building 3. CEP 90050-170, Porto Alegre, RS, Brazil
| | - Jessié M Gutierres
- Graduate Program in Pathology, Pathology Research Laboratory, Federal University of Health Sciences of Porto Alegre, Sarmento Leite, 245, Room 514, Building 3. CEP 90050-170, Porto Alegre, RS, Brazil
| | - Marilda da C Fernandes
- Graduate Program in Pathology, Pathology Research Laboratory, Federal University of Health Sciences of Porto Alegre, Sarmento Leite, 245, Room 514, Building 3. CEP 90050-170, Porto Alegre, RS, Brazil.
| |
Collapse
|
2
|
Li RN, Chen SL. Mechanistic Insights into the N-Hydroxylations Catalyzed by the Binuclear Iron Domain of SznF Enzyme: Key Piece in the Synthesis of Streptozotocin. Chemistry 2024; 30:e202303845. [PMID: 38212866 DOI: 10.1002/chem.202303845] [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: 11/19/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 01/13/2024]
Abstract
SznF, a member of the emerging family of heme-oxygenase-like (HO-like) di-iron oxidases and oxygenases, employs two distinct domains to catalyze the conversion of Nω-methyl-L-arginine (L-NMA) into N-nitroso-containing product, which can subsequently be transformed into streptozotocin. Using unrestricted density functional theory (UDFT) with the hybrid functional B3LYP, we have mechanistically investigated the two sequential hydroxylations of L-NMA catalyzed by SznF's binuclear iron central domain. Mechanism B primarily involves the O-O bond dissociation, forming Fe(IV)=O, induced by the H+/e- introduction to the FeA side of μ-1,2-peroxo-Fe2(III/III), the substrate hydrogen abstraction by Fe(IV)=O, and the hydroxyl rebound to the substrate N radical. The stochastic addition of H+/e- to the FeB side (mechanism C) can transition to mechanism B, thereby preventing enzyme deactivation. Two other competing mechanisms, involving the direct O-O bond dissociation (mechanism A) and the addition of H2O as a co-substrate (mechanism D), have been ruled out.
Collapse
Affiliation(s)
- Rui-Ning Li
- Key Laboratory of Cluster Science of Ministry of Education, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Shi-Lu Chen
- Key Laboratory of Cluster Science of Ministry of Education, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 100081, China
| |
Collapse
|
3
|
Fronza MG, Alves D, Praticò D, Savegnago L. The neurobiology and therapeutic potential of multi-targeting β-secretase, glycogen synthase kinase 3β and acetylcholinesterase in Alzheimer's disease. Ageing Res Rev 2023; 90:102033. [PMID: 37595640 DOI: 10.1016/j.arr.2023.102033] [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: 06/16/2023] [Revised: 08/04/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023]
Abstract
Alzheimer's Disease (AD) is the most common form of dementia, affecting almost 50 million of people around the world, characterized by a complex and age-related progressive pathology with projections to duplicate its incidence by the end of 2050. AD pathology has two major hallmarks, the amyloid beta (Aβ) peptides accumulation and tau hyperphosphorylation, alongside with several sub pathologies including neuroinflammation, oxidative stress, loss of neurogenesis and synaptic dysfunction. In recent years, extensive research pointed out several therapeutic targets which have shown promising effects on modifying the course of the disease in preclinical models of AD but with substantial failure when transposed to clinic trials, suggesting that modulating just an isolated feature of the pathology might not be sufficient to improve brain function and enhance cognition. In line with this, there is a growing consensus that an ideal disease modifying drug should address more than one feature of the pathology. Considering these evidence, β-secretase (BACE1), Glycogen synthase kinase 3β (GSK-3β) and acetylcholinesterase (AChE) has emerged as interesting therapeutic targets. BACE1 is the rate-limiting step in the Aβ production, GSK-3β is considered the main kinase responsible for Tau hyperphosphorylation, and AChE play an important role in modulating memory formation and learning. However, the effects underlying the modulation of these enzymes are not limited by its primarily functions, showing interesting effects in a wide range of impaired events secondary to AD pathology. In this sense, this review will summarize the involvement of BACE1, GSK-3β and AChE on synaptic function, neuroplasticity, neuroinflammation and oxidative stress. Additionally, we will present and discuss new perspectives on the modulation of these pathways on AD pathology and future directions on the development of drugs that concomitantly target these enzymes.
Collapse
Affiliation(s)
- Mariana G Fronza
- Neurobiotechnology Research Group (GPN) - Centre for Technology Development CDTec, Federal University of Pelotas (UFPel), Pelotas, RS, Brazil
| | - Diego Alves
- Laboratory of Clean Organic Synthesis (LASOL), Center for Chemical, Pharmaceutical and Food Sciences (CCQFA), UFPel, RS, Brazil
| | - Domenico Praticò
- Alzheimer's Center at Temple - ACT, Temple University, Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Lucielli Savegnago
- Neurobiotechnology Research Group (GPN) - Centre for Technology Development CDTec, Federal University of Pelotas (UFPel), Pelotas, RS, Brazil.
| |
Collapse
|
4
|
Li Q, Qi S, Liang J, Tian Y, He S, Liao Q, Xing S, Han L, Chen X. Review of triazole scaffolds for treatment and diagnosis of Alzheimer's disease. Chem Biol Interact 2023; 382:110623. [PMID: 37451665 DOI: 10.1016/j.cbi.2023.110623] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/28/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023]
Abstract
Triazole scaffolds, a series of 5-membered heterocycles, are well known for their high efficacy, low toxicity, and superior pharmacokinetics. Alzheimer's disease (AD) is the first neurodegenerative disorder with complex pathological mechanisms. Triazole, as an aromatic group with three nitrogen atoms, forms polar and non-polar interactions with diverse key residues in the receptor-ligand binding procedure, and has been widely used in the molecular design in the development of anti-AD agents. Moreover, considering the simple synthesis approaches, triazole scaffolds are commonly used to link two pharmacodynamic groups in one chemical molecule, forming multi-target directed ligands (MTDLs). Furthermore, the click reaction between azide- and cyano-modified enzyme and ligand provides feasibility for the new modulator discovery, compound tissue distribution evaluation, enzyme localization, and pharmacological mechanism study, promoting the diagnosis of AD course.
Collapse
Affiliation(s)
- Qi Li
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China.
| | - Shulei Qi
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Jinxin Liang
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Yuqing Tian
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Siyu He
- Guizhou Medical University, Guiyang, 550025, Guizhou, PR China
| | - Qinghong Liao
- Shandong Junrong Technology Transfer Co., Ltd, Qingdao, 266071, Shandong, PR China
| | - Shuaishuai Xing
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, PR China
| | - Lingfei Han
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, PR China
| | - Xuehong Chen
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China.
| |
Collapse
|
5
|
Fronza MG, Sacramento M, Alves D, Praticò D, Savegnago L. QTC-4-MeOBnE Ameliorated Depressive-Like Behavior and Memory Impairment in 3xTg Mice. Mol Neurobiol 2023; 60:1733-1745. [PMID: 36567360 DOI: 10.1007/s12035-022-03159-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 12/01/2022] [Indexed: 12/27/2022]
Abstract
Growing evidence has associated major depressive disorder (MDD) as a risk factor or prodromal syndrome for the occurrence of Alzheimer's disease (AD). Although this dilemma remains open, it is widely shown that a lifetime history of MDD is correlated with faster progression of AD pathology. Therefore, antidepressant drugs with neuroprotective effects could be an interesting therapeutic conception to target this issue simultaneously. In this sense, 1-(7-chloroquinolin-4-yl)-N-(4-methoxybenzyl)-5-methyl-1H-1,2,3-triazole-4- carboxamide (QTC-4-MeOBnE) was initially conceived as a multi-target ligand with affinity to β-secretase (BACE), glycogen synthase kinase 3β (GSK3β), and acetylcholinesterase but has also shown secondary effects on pathways involved in neuroinflammation and neurogenesis in preclinical models of AD. Herein, we investigated the effect of QTC-4-MeOBnE (1 mg/kg) administration for 45 days on depressive-like behavior and memory impairment in 3xTg mice, before the pathology is completely established. The treatment with QTC-4-MeOBnE prevented memory impairment and depressive-like behavior assessed by the Y-Maze task and forced swimming test. This effect was associated with the modulation of plural pathways involved in the onset and progression of AD, in cerebral structures of the cortex and hippocampus. Among them, the reduction of amyloid beta (Aβ) production mediated by changes in amyloid precursor protein metabolism and hippocampal tau phosphorylation through the inhibition of kinases. Additionally, QTC-4-MeOBnE also exerted beneficial effects on neuroinflammation and synaptic integrity. Overall, our studies suggest that QTC-4-MeOBnE has a moderate effect in a transgenic model of AD, indicating that perhaps studies regarding the neuropsychiatric effects as a neuroprotective molecule are more prone to be feasible.
Collapse
Affiliation(s)
- Mariana G Fronza
- Neurobiotechnology Research Group (GPN) - Postgraduate Program of Biotechnology, Federal University of Pelotas (UFPel), Pelotas, RS, Brazil
| | - Manoela Sacramento
- Laboratory of Clean Organic Synthesis (LASOL), Postgraduate Program of Chemistry, UFPel, Pelotas, RS, Brazil
| | - Diego Alves
- Laboratory of Clean Organic Synthesis (LASOL), Postgraduate Program of Chemistry, UFPel, Pelotas, RS, Brazil
| | - Domenico Praticò
- Alzheimer's Center at Temple - ACT, Temple University, Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Lucielli Savegnago
- Neurobiotechnology Research Group (GPN) - Postgraduate Program of Biotechnology, Federal University of Pelotas (UFPel), Pelotas, RS, Brazil.
| |
Collapse
|
6
|
Gulati HK, Kumar N, Sharma A, Jyoti, Khanna A, Sharma S, Salwan R, Bedi PMS. A comprehensive review on Triazole based Conjugates as Acetylcholinesterase Inhibitors: Design Strategies, Synthesis, Biological Activity, Structure Activity Relationships, Molecular Docking Studies. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2023.135354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2023]
|
7
|
Li JG, Blass BE, Praticò D. Beneficial Effect of a Small Pharmacologic Chaperone on the Established Alzheimer's Disease Phenotype. J Alzheimers Dis 2023; 91:463-469. [PMID: 36442197 DOI: 10.3233/jad-220869] [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: 11/27/2022]
Abstract
BACKGROUND The endosomal retromer complex system is a key controller for trafficking of proteins. Downregulation of its recognition core proteins, such as VPS35, is present in Alzheimer's disease (AD) brain, whereas its normalization prevents the development of AD pathology in a transgenic model with amyloid-β deposits and tau tangles. OBJECTIVE Assess the effect of targeting VPS35 after the AD pathology and memory impairments have developed. METHODS Twelve-month-old triple transgenic mice were treated with a small pharmacological chaperone, TPT-172, or vehicle for 14 weeks. At the end of this period, the effect of the drug on their phenotype was evaluated. RESULTS While control mice had a decline of learning and memory, the group receiving the chaperone did not. Moreover, when compared with controls the treated mice had significantly less amyloid-β peptides and phosphorylated tau, elevation of post-synaptic protein, and reduction in astrocytes activation. CONCLUSION Taken together, our findings demonstrate that pharmacologic stabilization of the retromer recognition core is beneficial also after the AD-like pathologic phenotype is established.
Collapse
Affiliation(s)
- Jian-Guo Li
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Benjamin E Blass
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Philadelphia, PA, USA.,Moulder Center for Drug Discovery Research, School of Pharmacy, Temple University, Philadelphia, PA, USA
| | - Domenico Praticò
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
8
|
Vieira MPS, Silva OBS, Souza GF, Cavalcante GTS, Souza FMA, Gitaí DLG, Castro OW, Nicácio DCSP, Cofré AHR, Amorós MA, Silva AV, Neto GJDS, Silva AHQ, Correia WBZGB, Junkes JA, Duarte FS, Guedes JS, Nogueira FCS, Meneghetti MR, Duzzioni M. First evaluation of the anxiolytic-like effects of a bromazepam‑palladium complex in mice. J Inorg Biochem 2022; 237:112012. [PMID: 36162209 DOI: 10.1016/j.jinorgbio.2022.112012] [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: 05/19/2022] [Revised: 09/09/2022] [Accepted: 09/16/2022] [Indexed: 01/18/2023]
Abstract
A significant fraction of patients are affected by persistent fear and anxiety. Currently, there are several anxiolytic drug options, however their clinical outcomes do not fully manage the symptoms. Here, we evaluated the effects of a bromazepam‑palladium derivative [2-{(7-bromo-2-oxo-1,3-dihydro-2H-1,4-benzodiazepin-5-il)pyridinyl-κ2-N,N}chloropalladium(II)], [(BMZ)PdCl2], on fear/anxiety and memory-related behavior in mice. For this, female Swiss mice were treated intraperitoneally (i.p.) with saline (NaCl 0.9%) or [(BMZ)PdCl2] (0.5, 5.0, or 50 μg/kg). After 30 min, different tests were performed to evaluate anxiety, locomotion, and memory. We also evaluated the acute toxicity of [(BMZ)PdCl2] using a cell viability assay (neutral red uptake assay), and whether the drugs mechanism of action involves the γ-aminobutyric acid type A (GABAA) receptor complex by pre-treating animals with flumazenil (1.0 mg/kg, i.p., a competitive antagonist of GABAA-binding site). Our results demonstrate that [(BMZ)PdCl2] induces an anxiolytic-like phenotype in the elevated plus-maze test and that this effect can be blocked by flumazenil. Furthermore, there were no behavioral alterations induced by [(BMZ)PdCl2], as evaluated in the light-dark box, open field, and step-down passive avoidance tests. In the acute toxicity assay, [(BMZ)PdCl2] presented IC50 and LD50 values of 218 ± 60 μg/mL and 780 ± 80 mg/kg, respectively, and GSH category 4. Taken together, our results show that the anxiolytic-like effect of acute treatment with [(BMZ)PdCl2] occurs through the modulation of the benzodiazepine site in the GABAA receptor complex. Moreover, we show indications that [(BMZ)PdCl2] does not promote sedation and amnesia and presents the same toxicity as the bromazepam prototype.
Collapse
Affiliation(s)
- Mirella P S Vieira
- Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil
| | - Ozileudiane B S Silva
- Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil
| | - Gabriela F Souza
- Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil
| | - Gabriela T S Cavalcante
- Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil
| | - Fernanda M A Souza
- Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil
| | - Daniel L G Gitaí
- Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil
| | - Olagide W Castro
- Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil
| | - Dannyele C S P Nicácio
- Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil
| | - Axel H R Cofré
- Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil
| | - Mariana A Amorós
- Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil
| | - Artur V Silva
- Grupo de Catálise e Reatividade Química, Instituto de Química e Biotecnologia, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil
| | - Geraldo José da Silva Neto
- Grupo de Catálise e Reatividade Química, Instituto de Química e Biotecnologia, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil
| | - Allysson H Q Silva
- Grupo de Catálise e Reatividade Química, Instituto de Química e Biotecnologia, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil
| | - Walleska B Z G B Correia
- Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil; Grupo de Catálise e Reatividade Química, Instituto de Química e Biotecnologia, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil
| | - Janaína A Junkes
- Instituto de Tecnologia e Pesquisa, Centro Universitário Tiradentes, Avenida Comendador Gustavo Paiva, 5017, Cruz das Almas, Maceió, Alagoas 57038-000, Brazil
| | - Filipe S Duarte
- Departamento de Fisiologia e Farmacologia, Universidade Federal de Pernambuco, Avenida Professor Moraes Rego, S/N, Cidade Universitária, Recife, Pernambuco 50670-420, Brazil
| | - Jéssica S Guedes
- Instituto de Química, Universidade Federal do Rio de Janeiro, Avenida Athos da Silveira Ramos, 149, Cidade Universitária, Rio de Janeiro, Rio de Janeiro 21941-909, Brazil
| | - Fábio C S Nogueira
- Instituto de Química, Universidade Federal do Rio de Janeiro, Avenida Athos da Silveira Ramos, 149, Cidade Universitária, Rio de Janeiro, Rio de Janeiro 21941-909, Brazil
| | - Mario R Meneghetti
- Grupo de Catálise e Reatividade Química, Instituto de Química e Biotecnologia, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil.
| | - Marcelo Duzzioni
- Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil.
| |
Collapse
|
9
|
Moreira NCDS, Tamarozzi ER, Lima JEBDF, Piassi LDO, Carvalho I, Passos GA, Sakamoto-Hojo ET. Novel Dual AChE and ROCK2 Inhibitor Induces Neurogenesis via PTEN/AKT Pathway in Alzheimer's Disease Model. Int J Mol Sci 2022; 23:ijms232314788. [PMID: 36499116 PMCID: PMC9737254 DOI: 10.3390/ijms232314788] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/07/2022] [Accepted: 11/15/2022] [Indexed: 11/29/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive and complex neurodegenerative disease. Acetylcholinesterase inhibitors (AChEIs) are a major class of drugs used in AD therapy. ROCK2, another promising target for AD, has been associated with the induction of neurogenesis via PTEN/AKT. This study aimed to characterize the therapeutic potential of a novel donepezil-tacrine hybrid compound (TA8Amino) to inhibit AChE and ROCK2 protein, leading to the induction of neurogenesis in SH-SY5Y cells. Experiments were carried out with undifferentiated and neuron-differentiated SH-SY5Y cells submitted to treatments with AChEIs (TA8Amino, donepezil, and tacrine) for 24 h or 7 days. TA8Amino was capable of inhibiting AChE at non-cytotoxic concentrations after 24 h. Following neuronal differentiation for 7 days, TA8Amino and donepezil increased the percentage of neurodifferentiated cells and the length of neurites, as confirmed by β-III-tubulin and MAP2 protein expression. TA8Amino was found to participate in the activation of PTEN/AKT signaling. In silico analysis showed that TA8Amino can stably bind to the active site of ROCK2, and in vitro experiments in SH-SY5Y cells demonstrate that TA8Amino significantly reduced the expression of ROCK2 protein, contrasting with donepezil and tacrine. Therefore, these results provide important information on the mechanism underlying the action of TA8Amino with regard to multi-target activities.
Collapse
Affiliation(s)
| | - Elvira Regina Tamarozzi
- Department of Biotechnology, School of Arts, Sciences and Humanities—USP, São Paulo 03828-000, Brazil
| | | | - Larissa de Oliveira Piassi
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo—USP, Ribeirão Preto 14049-900, Brazil
| | - Ivone Carvalho
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo—USP, Ribeirão Preto 14040-900, Brazil
| | - Geraldo Aleixo Passos
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo—USP, Ribeirão Preto 14049-900, Brazil
- Laboratory of Genetics and Molecular Biology, Department of Basic and Oral Biology, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14049-900, Brazil
| | - Elza Tiemi Sakamoto-Hojo
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo—USP, Ribeirão Preto 14049-900, Brazil
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo—USP, Ribeirão Preto 14040-901, Brazil
- Correspondence: ; Tel.: +55-16-3315-3827
| |
Collapse
|
10
|
Anti-Inflammatory Activity of 4-(4-(Heptyloxy)phenyl)-2,4-dihydro-3 H-1,2,4-triazol-3-one via Repression of MAPK/NF-κB Signaling Pathways in β-Amyloid-Induced Alzheimer's Disease Models. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27155035. [PMID: 35956985 PMCID: PMC9370156 DOI: 10.3390/molecules27155035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/31/2022] [Accepted: 08/05/2022] [Indexed: 01/03/2023]
Abstract
Alzheimer’s disease (AD) is a major neurodegenerative disease, but so far, it can only be treated symptomatically rather than changing the process of the disease. Recently, triazoles and their derivatives have been shown to have potential for the treatment of AD. In this study, the neuroprotective effects of 4-(4-(heptyloxy)phenyl)-2,4-dihydro-3H-1,2,4-triazol-3-one (W112) against β-amyloid (Aβ)-induced AD pathology and its possible mechanism were explored both in vitro and in vivo. The results showed that W112 exhibits a neuroprotective role against Aβ-induced cytotoxicity in PC12 cells and improves the learning and memory abilities of Aβ-induced AD-like rats. In addition, the assays of the protein expression revealed that W112 reversed tau hyperphosphorylation and reduced the production of proinflammatory cytokines, tumor necrosis factor-α and interleukin-6, both in vitro and in vivo studies. Further study indicated that the regulation of mitogen-activated protein kinase/nuclear factor-κB pathways played a key role in mediating the neuroprotective effects of W112 against AD-like pathology. W112 may become a potential drug for AD intervention.
Collapse
|
11
|
1-(7-Chloroquinolin-4-yl)-N-(4-Methoxybenzyl)-5-Methyl-1H-1,2, 3-Triazole-4- carboxamide Reduces Aβ Formation and Tau Phosphorylation in Cellular Models of Alzheimer's Disease. Neurochem Res 2022; 47:1110-1122. [PMID: 35165799 DOI: 10.1007/s11064-021-03514-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/17/2021] [Accepted: 12/18/2021] [Indexed: 10/19/2022]
Abstract
1-(7-Chloroquinolin-4-yl)-N-(4-methoxybenzyl)-5-methyl-1H-1,2,3-triazole-4- carboxamide (QTC-4-MeOBnE) is a new multi-target directed ligand (MTDL) rationally designed to have affinity with β-secretase (BACE), Glycogen Synthase Kinase 3β (GSK3β) and acetylcholinesterase, which are considered promising targets on the development of disease-modifying therapies against Alzheimer's Disease (AD). Previously, QTC-4-MeOBnE treatment showed beneficial effects in preclinical AD-like models by influencing in vivo neurogenesis, oxidative and inflammatory pathways. However, the biological effect and mechanism of action exerted by QTC-4-MeOBnE in AD cellular models have not been elucidated yet. Hereby we investigate the acute effect of QTC-4-MeOBnE on neuronal cells overexpressing Amyloid Protein Precursor (APP) or human tau protein, the two main features of the AD pathophysiology. When compared to the control group, QTC-4-MeOBnE treatment prevented amyloid beta (Aβ) formation through the downregulation of APP and BACE levels in APPswe-expressing cells. Furthermore, in N2a cells overexpressing human tau, QTC-4-MeOBnE reduced the levels of phosphorylated forms of tau via the modulation of the GSK3β pathway. Taken together, our findings provide new insights into the mechanism of action exerted by QTC-4-MeOBnE in AD cellular models, and further support its potential as an interesting therapeutic strategy against AD.
Collapse
|
12
|
Xiang X, Wang X, Jin S, Hu J, Wu Y, Li Y, Wu X. Activation of GPR55 attenuates cognitive impairment and neurotoxicity in a mouse model of Alzheimer's disease induced by Aβ 1-42 through inhibiting RhoA/ROCK2 pathway. Prog Neuropsychopharmacol Biol Psychiatry 2022; 112:110423. [PMID: 34363866 DOI: 10.1016/j.pnpbp.2021.110423] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 06/11/2021] [Accepted: 07/31/2021] [Indexed: 12/17/2022]
Abstract
The accumulation of amyloid-β (Aβ) peptides in the brain is considered to be the initial event in the Alzheimer's disease (AD). Neurotoxicity mediated by Aβ has been demonstrated to damage the cognitive function. In the present study, we sought to determine the effects of O-1602, a specific G-protein coupled receptor 55 (GPR55) agonist, on the impairment of learning and memory induced by intracerebroventricular (i.c.v.) of Aβ1-42 (400 pmol/mouse) in mice. Our results showed that i.c.v. injection of aggregated Aβ1-42 into the brain of mice resulted in cognitive impairment and neurotoxicity. In contrast, O-1602 (2.0 or 4.0 μg/mouse, i.c.v.) can improve memory impairment induced by Aβ1-42 in the Morris water maze (MWM), and novel object recognition (NOR) tests. Besides, we found that O-1602 reduced the activity of β-secretase 1 (BACE1) and the level of soluble Aβ1-42 in the hippocampus and frontal cortex. Importantly, O-1602 treatment reversed Aβ1-42-induced GPR55 down-regulation, decreased pro-inflammatory cytokines, and the level of malondialdehyde (MDA), increased the levels of glutathione (GSH), superoxide dismutase (SOD), and catalase (CAT), as well as suppressed apoptosis as indicated by decreased TUNEL-positive cells, and increased the ratio of Bcl-2/Bax. O-1602 treatment also pronouncedly ameliorated synaptic dysfunction by promoting the upregulation of PSD-95 and synaptophysin (SYN) proteins. Moreover, O-1602 concurrently down regulated the protein levels of RhoA, and ROCK2, the critical proteins in the RhoA/ROCK2 pathway. This study indicates that O-1602 may reverse Aβ1-42-induced cognitive impairment and neurotoxicity in mice by inhibiting RhoA/ROCK2 pathway. Taken together, these findings suggest that GPR55 could be a novel and promising target for the treatment of AD.
Collapse
Affiliation(s)
- XiaoTong Xiang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei 230032, China
| | - Xin Wang
- West Anhui Health Vocational College, Luan 237000, China
| | - ShiYu Jin
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei 230032, China
| | - Jie Hu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei 230032, China
| | - YuMei Wu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei 230032, China
| | - YueYue Li
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei 230032, China
| | - Xian Wu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei 230032, China.
| |
Collapse
|
13
|
Fronza MG, Baldinotti R, Fetter J, Rosa SG, Sacramento M, Nogueira CW, Alves D, Praticò D, Savegnago L. Beneficial effects of QTC-4-MeOBnE in an LPS-induced mouse model of depression and cognitive impairments: The role of blood-brain barrier permeability, NF-κB signaling, and microglial activation. Brain Behav Immun 2022; 99:177-191. [PMID: 34624485 DOI: 10.1016/j.bbi.2021.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 12/11/2022] Open
Abstract
Clinical and preclinical investigations have suggested a possible biological link betweenmajor depressive disorder (MDD) and Alzheimer's disease (AD). Therefore, a pharmacologic approach to treating MDD could be envisioned as a preventative therapy for some AD cases. In line with this, 1-(7-chloroquinolin-4-yl)-N-(4-methoxybenzyl)-5-methyl-1H-1,2,3-triazole-4 carboxamide (QTC-4-MeOBnE) is characterized as an inhibitor of β-secretase, glycogen synthase kinase 3β, and acetylcholinesterase and has also shown secondary effects underlying the modulation of neurogenesis and synaptic plasticity pathways. Therefore, we investigated the effects of QTC-4-MeOBnE treatment (0.1 or 1 mg/kg) on depressive-like behavior and cognitive impairments elicited by repeated injections of lipopolysaccharide (LPS; 250 μg/kg) in mice. Injections of LPS for seven days led to memory impairments and depressive-like behavior, as evidenced in the Y-maze/object recognition test and forced swimming/splash tests, respectively. However, these impairments were prevented in mice that, after the last LPS injection, were also treated with QTC-4-MeOBnE (1 mg/kg). This effect was associated with restoring blood-brain barrier permeability, reducing oxidative/nitrosative biomarkers, and decreasing neuroinflammation mediated NF-κB signaling in the hippocampus and cortex of the mice. To further investigate the involvement with NF-κB signaling, we evaluated the effects of QTC-4-MeOBnE on microglial cell activation through canonical and non-canonical pathways and the modulation of the involved components. Together, our findings highlight the pharmacological benefits of QTC-4-MeOBnE in a mouse model of sickness behavior and memory impairments, supporting the novel concept that since this molecule produces anti-depressant activity, it could also be beneficial for preventing AD onset and related dementias in subjects suffering from MDD through inflammatory pathway modulation.
Collapse
Affiliation(s)
- Mariana G Fronza
- Neurobiotechnology Research Group (GPN) - Centre for Technology Development CDTec, Federal University of Pelotas (UFPel), Pelotas, RS, Brazil
| | - Rodolfo Baldinotti
- Neurobiotechnology Research Group (GPN) - Centre for Technology Development CDTec, Federal University of Pelotas (UFPel), Pelotas, RS, Brazil
| | - Jenifer Fetter
- Neurobiotechnology Research Group (GPN) - Centre for Technology Development CDTec, Federal University of Pelotas (UFPel), Pelotas, RS, Brazil
| | - Suzan Gonçalves Rosa
- Laboratory of Synthesis, Reactivity and Pharmacological and Toxicological Evaluation of Organocalcogens, Center for Natural and Exact Sciences, Federal University of Santa Maria, UFSM, Santa Maria, RS, Brazil
| | - Manoela Sacramento
- Laboratory of Clean Organic Synthesis (LASOL), Center for Chemical, Pharmaceutical and Food Sciences (CCQFA), UFPel, RS, Brazil
| | - Cristina Wayne Nogueira
- Laboratory of Synthesis, Reactivity and Pharmacological and Toxicological Evaluation of Organocalcogens, Center for Natural and Exact Sciences, Federal University of Santa Maria, UFSM, Santa Maria, RS, Brazil
| | - Diego Alves
- Laboratory of Clean Organic Synthesis (LASOL), Center for Chemical, Pharmaceutical and Food Sciences (CCQFA), UFPel, RS, Brazil
| | - Domenico Praticò
- Alzheimer's Center at Temple - ACT, Temple University, Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Lucielli Savegnago
- Neurobiotechnology Research Group (GPN) - Centre for Technology Development CDTec, Federal University of Pelotas (UFPel), Pelotas, RS, Brazil.
| |
Collapse
|
14
|
Selective Synthesis of 2-(1,2,3-Triazoyl) Quinazolinones through Copper-Catalyzed Multicomponent Reaction. Catalysts 2021. [DOI: 10.3390/catal11101170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
We describe here our results from the copper-catalyzed three component reaction of 2-azidobenzaldehyde, anthranilamide and terminal alkynes, using Et3N as base, and DMSO as solvent. Depending on the temperature and amount of Et3N used in the reactions, 1,2,3-triazolyl-quinazolinones or 1,2,3-triazolyl-dihydroquinazolinone could be obtained. When the reactions were performed at 100 °C using 2 equivalents of Et3N, 1,2,3-triazolyl-dihydroquinazolinone was formed in 82% yield, whereas reactions carried out at 120 °C using 1 equivalent of Et3N provided 1,2,3-triazolyl-quinazolinones in moderate-to-good yields.
Collapse
|
15
|
Chen MH, Wang TJ, Chen LJ, Jiang MY, Wang YJ, Tseng GF, Chen JR. The effects of astaxanthin treatment on a rat model of Alzheimer's disease. Brain Res Bull 2021; 172:151-163. [PMID: 33932491 DOI: 10.1016/j.brainresbull.2021.04.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 04/03/2021] [Accepted: 04/25/2021] [Indexed: 01/16/2023]
Abstract
Alzheimer's disease (AD), a progressive neurodegenerative disorder characterized by memory loss and dementia, could be a consequence of the abnormalities of cortical milieu, such as oxidative stress, inflammation, and/or accompanied with the aggregation of β-amyloid. The majority of AD patients are sporadic, late-onset AD, which predominantly occurs over 65 years of age. Our results revealed that the ferrous amyloid buthionine (FAB)-infused sporadic AD-like model showed deficits in spatial learning and memory and with apparent loss of choline acetyltransferase (ChAT) expression in medial septal (MS) nucleus. In hippocampal CA1 region, the loss of pyramidal neurons was accompanied with cholinergic fiber loss and neuroinflammatory responses including glial reaction and enhanced expression of inducible nitric oxide synthase (iNOS). Surviving hippocampal CA1 pyramidal neurons showed the reduction of dendritic spines as well. Astaxanthin (ATX), a potent antioxidant, reported to improve the outcome of oxidative-stress-related diseases. The ATX treatment in FAB-infused rats decreased neuroinflammation and restored the ChAT + fibers in hippocampal CA1 region and the ChAT expression in MS nucleus. It also partly recovered the spine loss on hippocampal CA1 pyramidal neurons and ameliorated the behavioral deficits in AD-like rats. From these data, we believed that the ATX can be a potential option for slowing the progression of Alzheimer's disease.
Collapse
Affiliation(s)
- Mu-Hsuan Chen
- Ph.D. Program in Tissue Engineering and Regenerative Medicine, National Chung-Hsing University, Taichung, Taiwan
| | - Tsyr-Jiuan Wang
- Department of Nursing, National Taichung University of Science and Technology, Taichung, Taiwan
| | - Li-Jin Chen
- Department of Anatomy, College of Medicine, Tzu-Chi University, Hualien, Taiwan
| | - Ming-Ying Jiang
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung-Hsing University, Taichung, Taiwan
| | - Yueh-Jan Wang
- Department of Anatomy, College of Medicine, Tzu-Chi University, Hualien, Taiwan
| | - Guo-Fang Tseng
- Department of Anatomy, College of Medicine, Tzu-Chi University, Hualien, Taiwan.
| | - Jeng-Rung Chen
- Ph.D. Program in Tissue Engineering and Regenerative Medicine, National Chung-Hsing University, Taichung, Taiwan; Department of Veterinary Medicine, College of Veterinary Medicine, National Chung-Hsing University, Taichung, Taiwan.
| |
Collapse
|