1
|
Hussain F, Tahir A, Rehman HM, Wu Y, Shah M, Rashid U. Promising thiazolidinedione-thiazole based multi-target and neuroprotective hybrids for Alzheimer's disease: Design, synthesis, in-vitro, in-vivo and in-silico studies. Eur J Med Chem 2025; 287:117327. [PMID: 39914143 DOI: 10.1016/j.ejmech.2025.117327] [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: 11/19/2024] [Revised: 01/21/2025] [Accepted: 01/23/2025] [Indexed: 02/24/2025]
Abstract
Alzheimer's disease (AD) is marked by low neurotransmitter levels, inflammation, increased oxidative stress, and the aggregation of amyloid-β and tau proteins. The development of hybrid compounds acting as multi-target-directed ligands (MTDLs) is a novel and contemporary approach in Alzheimer's disease therapeutics. The objective of our current research focuses on identifying compounds with balanced, even moderate inhibition potential against multiple targets associated with cholinergic deficit and neuroinflammation. Inspired by our previous study, the thiazolidinedione-thiazole-based framework has been employed to design and synthesize a series of new hybrids. The inhibitory effects of the synthesized compounds on selected enzymes were investigated by employing in-vitro methods. The synergistic inhibition of acetylcholinesterase (AChE), monoamine oxidase-B (MAO-B), β-secretase (BACE-1), cyclooxygenase-2 (COX-2) and 5-lipoxygenase (5-LOX) by compound 49 is believed to have a more potent effect in treating Alzheimer's disease. Enzyme kinetic studies and their effect on substrate-enzyme interactions of the compounds with significant inhibitory potency for AChE and MAO-B were also investigated. Central nervous system (CNS) penetration was determined using in-vitro PAMPA assay. A neurotoxicity test on neuroblastoma cell lines (SH-SY5Y) showed that the compounds were non-toxic. Compound 49 showed an excellent neuroprotective effect by significantly reducing H2O2-induced oxidative stress. Antioxidant enzymes were studied in an in-vivo experiment in the brains of male BALB/c mice. Compound 49 showed its ability to reduce the oxidative stress. Furthermore, molecular docking studies and 100 ns MD simulations performed on the AChE, MAO-B, and COX-2 ligand-protein complexes revealed stable conformations of the ligand-protein interactions throughout the simulations.
Collapse
Affiliation(s)
- Fahad Hussain
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, 22060, Abbottabad, Pakistan
| | - Ayesha Tahir
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, 22060, Abbottabad, Pakistan
| | - Hafiz Muzzammel Rehman
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore, Punjab, Pakistan
| | - Yurong Wu
- Department of Chemistry, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Muhammad Shah
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, 22060, Abbottabad, Pakistan
| | - Umer Rashid
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, 22060, Abbottabad, Pakistan.
| |
Collapse
|
2
|
Vahid ZF, Eskandani M, Dadashi H, Vandghanooni S, Rashidi MR. Recent advances in potential enzymes and their therapeutic inhibitors for the treatment of Alzheimer's disease. Heliyon 2024; 10:e40756. [PMID: 39717593 PMCID: PMC11664286 DOI: 10.1016/j.heliyon.2024.e40756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 11/23/2024] [Accepted: 11/26/2024] [Indexed: 12/25/2024] Open
Abstract
Alzheimer's disease (AD), a chronic neurodegenerative disease, is clinically characterized by loss of memory and learning ability among other neurological deficits. Amyloid plaques, hyperphosphorylated tau protein, and neurofibrillary tangles involve in AD etiology. Meanwhile, enzymes and their inhibitors have become the focus of research in AD treatment. In this review, the molecular mechanisms involved in the pathogenesis of AD were overviewed and various enzymes such as acetylcholinesterase (AChE), butyrylcholinesterase (BuChE), β-secretase, γ-secretase, monoamine oxidase (MAO), and receptor of advanced glycation end products (RAGE) were highlighted as potential targets for AD treatment. Several hybrid molecules with essential substructures derived from various chemotypes have demonstrated desired pharmacological activity. It is envisioned that the development of new drugs that inhibit enzymes involved in AD is a future trend in the management of the disease.
Collapse
Affiliation(s)
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Dadashi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Somayeh Vandghanooni
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad-Reza Rashidi
- Medicinal Chemistry Department, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
3
|
Herrera-Arozamena C, Estrada-Valencia M, García-Díez G, Pérez C, León R, Infantes L, Morales-García JA, Pérez-Castillo A, Del Sastre E, López MG, Rodríguez-Franco MI. Discovery of a potent melatonin-based inhibitor of quinone reductase-2 with neuroprotective and neurogenic properties. Eur J Med Chem 2024; 277:116763. [PMID: 39146834 DOI: 10.1016/j.ejmech.2024.116763] [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: 06/12/2024] [Revised: 07/31/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024]
Abstract
5-Methoxy-3-(5-methoxyindolin-2-yl)-1H-indole (3), whose structure was unambiguously elucidated by X-ray analysis, was identified as a multi-target compound with potential application in neurodegenerative diseases. It is a low nanomolar inhibitor of QR2 (IC50 = 7.7 nM), with greater potency than melatonin and comparable efficacy to the most potent QR2 inhibitors described to date. Molecular docking studies revealed the potential binding mode of 3 to QR2, which explains its superior potency compared to melatonin. Furthermore, compound 3 inhibits hMAO-A, hMAO-B and hLOX-5 in the low micromolar range and is an excellent ROS scavenger. In phenotypic assays, compound 3 showed neuroprotective activity in a cellular model of oxidative stress damage, it was non-toxic, and was able to activate neurogenesis from neural stem-cell niches of adult mice. These excellent biological properties, together with its both good in silico and in vitro drug-like profile, highlight compound 3 as a promising drug candidate for neurodegenerative diseases.
Collapse
Affiliation(s)
- Clara Herrera-Arozamena
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), C/ Juan de la Cierva 3, E-28006, Madrid, Spain
| | - Martín Estrada-Valencia
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), C/ Juan de la Cierva 3, E-28006, Madrid, Spain
| | - Guillermo García-Díez
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), C/ Juan de la Cierva 3, E-28006, Madrid, Spain
| | - Concepción Pérez
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), C/ Juan de la Cierva 3, E-28006, Madrid, Spain
| | - Rafael León
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), C/ Juan de la Cierva 3, E-28006, Madrid, Spain
| | - Lourdes Infantes
- Instituto de Química Física Blas Cabrera, Consejo Superior de Investigaciones Científicas (IQF-CSIC), C/ Serrano 119, E-28006, Madrid, Spain
| | - José A Morales-García
- Departamento de Biología Celular, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Avda. Complutense s/n, E-28040, Madrid, Spain
| | - Ana Pérez-Castillo
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), C/ Arturo Duperier 4, E-28029, Madrid, Spain
| | - Eric Del Sastre
- Instituto Teófilo Hernando de I+D del Medicamento, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), C/ Arzobispo Morcillo 4, E-28029, Madrid, Spain
| | - Manuela G López
- Instituto Teófilo Hernando de I+D del Medicamento, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), C/ Arzobispo Morcillo 4, E-28029, Madrid, Spain
| | - María Isabel Rodríguez-Franco
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), C/ Juan de la Cierva 3, E-28006, Madrid, Spain.
| |
Collapse
|
4
|
Papagiouvannis G, Theodosis-Nobelos P, Rekka EA. Trolox, Ferulic, Sinapic, and Cinnamic Acid Derivatives of Proline and GABA with Antioxidant and/or Anti-Inflammatory Properties. Molecules 2024; 29:3763. [PMID: 39202843 PMCID: PMC11356873 DOI: 10.3390/molecules29163763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/06/2024] [Accepted: 08/06/2024] [Indexed: 09/03/2024] Open
Abstract
Degenerative conditions, such as neurodegenerative disorders (Alzheimer's disease (AD), Parkinson's disease (PD)) and cardiovascular diseases, are complex, multifactorial disorders whose pathophysiology has not been fully elucidated yet. As a result, the available treatment options cannot eliminate these diseases radically, but only alleviate the symptoms. Both inflammatory processes and oxidation are key factors in the development and evolution of neurodegeneration, while acetylcholinesterase inhibitors are the most used therapeutic options against AD. In this work, following the multi-targeting compound approach, we designed and synthesized a series of proline and gamma-aminobutyric acid (GABA) amides with various acidic moieties that possess an antioxidant and/or anti-inflammatory potency. Proline is the pharmacophore of nootropic drugs (e.g., piracetam) used for memory improvement, while GABA is the main inhibitory neurotransmitter in the central nervous system. The designed molecules were subjected to a preliminary screening of their bioactivity in antioxidant and anti-inflammatory assays, as well as against acetylcholinesterase. Most of the synthesized compounds could inhibit lipid peroxidation (IC50 as low as 8 μΜ) and oxidative protein glycation (inhibition of up to 48%) and reduce the 2,2-diphenyl-1-picrylhydrazyl free radical (DPPH). In addition, all of the compounds were moderate inhibitors of lipoxygenase (LOX) (up to 46% at 100 μΜ) and could decrease carrageenan-induced paw edema in rats by up to 55%. Finally, some of the compounds were moderate acetylcholinesterase inhibitors (IC50 as low as 219 μΜ). The results confirmed the design rationale, indicating that the compounds could be further optimized as multi-targeting molecules directed against degenerative conditions.
Collapse
Affiliation(s)
- Georgios Papagiouvannis
- Department of Pharmacy, School of Health Sciences, Frederick University, Nicosia 1036, Cyprus; (G.P.); (P.T.-N.)
| | - Panagiotis Theodosis-Nobelos
- Department of Pharmacy, School of Health Sciences, Frederick University, Nicosia 1036, Cyprus; (G.P.); (P.T.-N.)
| | - Eleni A. Rekka
- Department of Pharmaceutical Chemistry, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
5
|
Lohitaksha K, Kumari D, Shukla M, Byagari L, Ashireddygari VR, Tammineni P, Reddanna P, Gorla M. Eicosanoid signaling in neuroinflammation associated with Alzheimer's disease. Eur J Pharmacol 2024; 976:176694. [PMID: 38821162 DOI: 10.1016/j.ejphar.2024.176694] [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] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative condition affecting a substantial portion of the global population. It is marked by a complex interplay of factors, including the accumulation of amyloid plaques and tau tangles within the brain, leading to neuroinflammation and neuronal damage. Recent studies have underscored the role of free lipids and their derivatives in the initiation and progression of AD. Eicosanoids, metabolites of polyunsaturated fatty acids like arachidonic acid (AA), emerge as key players in this scenario. Remarkably, eicosanoids can either promote or inhibit the development of AD, and this multifaceted role is determined by how eicosanoid signaling influences the immune responses within the brain. However, the precise molecular mechanisms dictating the dual role of eicosanoids in AD remain elusive. In this comprehensive review, we explore the intricate involvement of eicosanoids in neuronal function and dysfunction. Furthermore, we assess the therapeutic potential of targeting eicosanoid signaling pathways as a viable strategy for mitigating or halting the progression of AD.
Collapse
Affiliation(s)
| | - Deepika Kumari
- Department of Biochemistry, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan, India
| | - Manas Shukla
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Lavanya Byagari
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | | | - Prasad Tammineni
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Pallu Reddanna
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, India; Brane Enterprises Private Limited, Hyderabad, India.
| | - Madhavi Gorla
- National Institute of Animal Biotechnology, Hyderabad, India.
| |
Collapse
|
6
|
Hans S, Stanton JE, Sauer AK, Shiels K, Saha SK, Lordan R, Tsoupras A, Zabetakis I, Grabrucker AM. Polar lipids modify Alzheimer's Disease pathology by reducing astrocyte pro-inflammatory signaling through platelet-activating factor receptor (PTAFR) modulation. Lipids Health Dis 2024; 23:113. [PMID: 38643113 PMCID: PMC11031880 DOI: 10.1186/s12944-024-02106-z] [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: 11/22/2023] [Accepted: 04/11/2024] [Indexed: 04/22/2024] Open
Abstract
BACKGROUND Pro-inflammatory processes triggered by the accumulation of extracellular amyloid beta (Aβ) peptides are a well-described pathology in Alzheimer's disease (AD). Activated astrocytes surrounding Aβ plaques contribute to inflammation by secreting proinflammatory factors. While astrocytes may phagocytize Aβ and contribute to Aβ clearance, reactive astrocytes may also increase Aβ production. Therefore, identifying factors that can attenuate astrocyte activation and neuroinflammation and how these factors influence pro-inflammatory pathways is important for developing therapeutic and preventive strategies in AD. Here, we identify the platelet-activating factor receptor (PTAFR) pathway as a key mediator of astrocyte activation. Intriguingly, several polar lipids (PLs) have exhibited anti-inflammatory protective properties outside the central nervous system through their inhibitory effect on the PTAFR pathway. Thus, we additionally investigated whether different PLs also exert inhibitory effects on the PAF pathway in astrocytes and whether their presence influences astrocytic pro-inflammatory signaling and known AD pathologies in vitro. METHODS PLs from salmon and yogurt were extracted using novel food-grade techniques and their fatty acid profile was determined using LC/MS. The effect of PLs on parameters such as astrocyte activation and generation of oxygen species (ROS) was assessed. Additionally, effects of the secretome of astrocytes treated with these polar lipids on aged neurons was measured. RESULTS We show that PLs obtained from salmon and yogurt lower astrocyte activation, the generation of reactive oxygen species (ROS), and extracellular Aβ accumulation. Cell health of neurons exposed to the secretome of astrocytes treated with salmon-derived PLs and Aβ was less affected than those treated with astrocytes exposed to Aβ only. CONCLUSION Our results highlight a novel underlying mechanism, why consuming PL-rich foods such as fish and dairy may reduce the risk of developing dementia and associated disorders.
Collapse
Affiliation(s)
- Sakshi Hans
- Department of Biological Sciences, University of Limerick, Limerick, V94PH61, Ireland
- Bernal Institute, University of Limerick, Analog Devices Building AD3-018, Castletroy, Limerick, V94PH61, Ireland
| | - Janelle E Stanton
- Department of Biological Sciences, University of Limerick, Limerick, V94PH61, Ireland
- Bernal Institute, University of Limerick, Analog Devices Building AD3-018, Castletroy, Limerick, V94PH61, Ireland
| | - Ann Katrin Sauer
- Department of Biological Sciences, University of Limerick, Limerick, V94PH61, Ireland
- Bernal Institute, University of Limerick, Analog Devices Building AD3-018, Castletroy, Limerick, V94PH61, Ireland
- Health Research Institute (HRI), University of Limerick, Limerick, V94PH61, Ireland
| | - Katie Shiels
- Shannon Applied Biotechnology Centre, Technological University of the Shannon, Moylish Park, Limerick, V94E8YF, Ireland
| | - Sushanta Kumar Saha
- Shannon Applied Biotechnology Centre, Technological University of the Shannon, Moylish Park, Limerick, V94E8YF, Ireland
| | - Ronan Lordan
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Systems Pharmacology and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexandros Tsoupras
- Hephaestus Laboratory, Department of Chemistry, School of Science, Democritus University of Thrace, Kavala University Campus, Kavala, GR65404, Greece
| | - Ioannis Zabetakis
- Department of Biological Sciences, University of Limerick, Limerick, V94PH61, Ireland
- Bernal Institute, University of Limerick, Analog Devices Building AD3-018, Castletroy, Limerick, V94PH61, Ireland
- Health Research Institute (HRI), University of Limerick, Limerick, V94PH61, Ireland
| | - Andreas M Grabrucker
- Department of Biological Sciences, University of Limerick, Limerick, V94PH61, Ireland.
- Bernal Institute, University of Limerick, Analog Devices Building AD3-018, Castletroy, Limerick, V94PH61, Ireland.
- Health Research Institute (HRI), University of Limerick, Limerick, V94PH61, Ireland.
| |
Collapse
|
7
|
Kazemi S, Safari S, Komaki S, Karimi SA, Golipoor Z, Komaki A. The effects of carvacrol and p-cymene on Aβ 1-42 -induced long-term potentiation deficit in male rats. CNS Neurosci Ther 2024; 30:e14459. [PMID: 37727020 PMCID: PMC10916422 DOI: 10.1111/cns.14459] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 08/04/2023] [Accepted: 08/25/2023] [Indexed: 09/21/2023] Open
Abstract
AIMS Alzheimer's disease (AD) is the most common type of dementia in which oxidative stress plays an important role. In this disease, learning and memory and the cellular mechanism associated with it, long-term potentiation (LTP), are impaired. Considering the beneficial effects of carvacrol (CAR) and p-cymene against AD, their effect was assessed on in vivo hippocampal LTP in the perforant pathway (PP)-dentate gyrus (DG) pathway in an Aβ1-42 -induced rat model of AD. METHODS Male Wistar rats were randomly assigned to five groups: sham: intracerebroventricular (ICV) injection of phosphate-buffered saline, Aβ: ICV Aβ1-42 injections, Aβ + CAR (50 mg/kg), Aβ + p-cymene (50 mg/kg), and Aβ + CAR + p-cymene. Administration of CAR and p-cymene was done by gavage daily 4 weeks before and 4 weeks after the Aβ injection. The population spike (PS) amplitude and field excitatory postsynaptic potentials (fEPSP) slope were determined in DG against the applied stimulation to the PP. RESULTS Aβ-treated rats exhibited impaired LTP induction in the PP-DG synapses, resulting in significant reduction in both fEPSP slope and PS amplitude compared to the sham animals. Aβ-treated rats consumed either CAR or p-cymene separately (but not their combination), and showed an enhancement in fEPSP slope and PS amplitude of the DG granular cells. CONCLUSIONS These data indicate that CAR or p-cymene can ameliorate Aβ-associated changes in synaptic plasticity. Surprisingly, the combination of CAR and p-cymene did not yield the same effect, suggesting a potential interaction between the two substances.
Collapse
Affiliation(s)
- Sahifeh Kazemi
- Department of Neuroscience, School of Science and Advanced Technologies in MedicineHamadan University of Medical SciencesHamadanIran
| | - Samaneh Safari
- Department of Neuroscience, School of Science and Advanced Technologies in MedicineHamadan University of Medical SciencesHamadanIran
- Student Research CommitteeHamadan University of Medical SciencesHamadanIran
| | - Somayeh Komaki
- Department of Physiology, School of MedicineHamadan University of Medical SciencesHamadanIran
| | - Seyed Asaad Karimi
- Department of Neuroscience, School of Science and Advanced Technologies in MedicineHamadan University of Medical SciencesHamadanIran
- Department of Physiology, School of MedicineHamadan University of Medical SciencesHamadanIran
| | - Zoleikha Golipoor
- Cellular and Molecular Research Center, Faculty of MedicineGuilan University of Medical SciencesRashtIran
| | - Alireza Komaki
- Department of Neuroscience, School of Science and Advanced Technologies in MedicineHamadan University of Medical SciencesHamadanIran
- Department of Physiology, School of MedicineHamadan University of Medical SciencesHamadanIran
| |
Collapse
|
8
|
Bartolo ND, Mortimer N, Manter MA, Sanchez N, Riley M, O'Malley TT, Hooker JM. Identification and Prioritization of PET Neuroimaging Targets for Microglial Phenotypes Associated with Microglial Activity in Alzheimer's Disease. ACS Chem Neurosci 2022; 13:3641-3660. [PMID: 36473177 DOI: 10.1021/acschemneuro.2c00607] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Activation of microglial cells accompanies the progression of many neurodegenerative disorders, including Alzheimer's disease (AD). Development of molecular imaging tools specific to microglia can help elucidate the mechanism through which microglia contribute to the pathogenesis and progression of neurodegenerative disorders. Through analysis of published genetic, transcriptomic, and proteomic data sets, we identified 19 genes with microglia-specific expression that we then ranked based on association with the AD characteristics, change in expression, and potential druggability of the target. We believe that the process we used to identify and rank microglia-specific genes is broadly applicable to the identification and evaluation of targets in other disease areas and for applications beyond molecular imaging.
Collapse
Affiliation(s)
- Nicole D Bartolo
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Charlestown, Massachusetts 02129, United States
| | - Niall Mortimer
- Human Biology and Data Science, Eisai Center for Genetics Guided Dementia Discovery, 35 Cambridgepark Drive, Cambridge, Massachusetts 02140, United States
| | - Mariah A Manter
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Charlestown, Massachusetts 02129, United States
| | - Nicholas Sanchez
- Human Biology and Data Science, Eisai Center for Genetics Guided Dementia Discovery, 35 Cambridgepark Drive, Cambridge, Massachusetts 02140, United States
| | - Misha Riley
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Charlestown, Massachusetts 02129, United States
| | - Tiernan T O'Malley
- Human Biology and Data Science, Eisai Center for Genetics Guided Dementia Discovery, 35 Cambridgepark Drive, Cambridge, Massachusetts 02140, United States
| | - Jacob M Hooker
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Charlestown, Massachusetts 02129, United States
| |
Collapse
|
9
|
Pap R, Pandur E, Jánosa G, Sipos K, Nagy T, Agócs A, Deli J. Lutein Decreases Inflammation and Oxidative Stress and Prevents Iron Accumulation and Lipid Peroxidation at Glutamate-Induced Neurotoxicity. Antioxidants (Basel) 2022; 11:2269. [PMID: 36421455 PMCID: PMC9687421 DOI: 10.3390/antiox11112269] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/11/2022] [Accepted: 11/15/2022] [Indexed: 07/30/2023] Open
Abstract
The xanthophyll carotenoid lutein has been widely used as supplementation due to its protective effects in light-induced oxidative stress. Its antioxidant and anti-inflammatory features suggest that it has a neuroprotective role as well. Glutamate is a major excitatory neurotransmitter in the central nervous system (CNS), which plays a key role in regulating brain function. Excess accumulation of intracellular glutamate accelerates an increase in the concentration of reactive oxygen species (ROS) in neurons leading to glutamate neurotoxicity. In this study, we focused on the effects of glutamate on SH-SY5Y neuroblastoma cells to identify the possible alterations in oxidative stress, inflammation, and iron metabolism that affect the neurological function itself and in the presence of antioxidant lutein. First, ROS measurements were performed, and then catalase (CAT) and Superoxide Dismutase (SOD) enzyme activity were determined by enzyme activity assay kits. The ELISA technique was used to detect proinflammatory TNFα, IL-6, and IL-8 cytokine secretions. Alterations in iron uptake, storage, and release were followed by gene expression measurements and Western blotting. Total iron level detections were performed by a ferrozine-based iron detection method, and a heme assay kit was used for heme measurements. The gene expression toward lipid-peroxidation was determined by RT-PCR. Our results show glutamate changes ROS, inflammation, and antioxidant enzyme activity, modulate iron accumulation, and may initiate lipid peroxidation in SH-SY5Y cells. Meanwhile, lutein attenuates the glutamate-induced effects on ROS, inflammation, iron metabolism, and lipid peroxidation. According to our findings, lutein could be a beneficial, supportive treatment in neurodegenerative disorders.
Collapse
Affiliation(s)
- Ramóna Pap
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary
| | - Edina Pandur
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary
| | - Gergely Jánosa
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary
| | - Katalin Sipos
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary
| | - Tamás Nagy
- Department of Laboratory Medicine, Faculty of Medical Sciences, University of Pécs, Ifjúság út 13, H-7624 Pécs, Hungary
| | - Attila Agócs
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - József Deli
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
- Department of Pharmacognosy, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary
| |
Collapse
|
10
|
Ağagündüz D, Gençer Bingöl F, Çelik E, Cemali Ö, Özenir Ç, Özoğul F, Capasso R. Recent developments in the probiotics as live biotherapeutic products (LBPs) as modulators of gut brain axis related neurological conditions. Lab Invest 2022; 20:460. [PMID: 36209124 PMCID: PMC9548122 DOI: 10.1186/s12967-022-03609-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 08/24/2022] [Indexed: 11/10/2022]
Abstract
Probiotics have been defined as “living microorganisms that create health benefits in the host when taken in sufficient amounts. Recent developments in the understanding of the relationship between the microbiom and its host have shown evidence about the promising potential of probiotics to improve certain health problems. However, today, there are some confusions about traditional and new generation foods containing probiotics, naming and classifications of them in scientific studies and also their marketing. To clarify this confusion, the Food and Drug Administration (FDA) declared that it has made a new category definition called "live biotherapeutic products" (LBPs). Accordingly, the FDA has designated LBPs as “a biological product that: i)contains live organisms, such as bacteria; ii)is applicable to the prevention, treatment, or cure of a disease/condition of human beings; and iii) is not a vaccine”. The accumulated literature focused on LBPs to determine effective strains in health and disease, and often focused on obesity, diabetes, and certain diseases like inflammatory bowel disease (IBD).However, microbiome also play an important role in the pathogenesis of diseases that age day by day in the modern world via gut-brain axis. Herein, we discuss the novel roles of LBPs in some gut-brain axis related conditions in the light of recent studies. This article may be of interest to a broad readership including those interested in probiotics as LBPs, their health effects and safety, also gut-brain axis.
Collapse
Affiliation(s)
- Duygu Ağagündüz
- Department of Nutrition and Dietetics, Gazi University, Faculty of Health Sciences, 06490, Ankara, Emek, Turkey.
| | - Feray Gençer Bingöl
- Department of Nutrition and Dietetics, Burdur Mehmet Akif Ersoy University, İstiklal Yerleşkesi, 15030, Burdur, Turkey
| | - Elif Çelik
- Department of Nutrition and Dietetics, Gazi University, Faculty of Health Sciences, 06490, Ankara, Emek, Turkey
| | - Özge Cemali
- Department of Nutrition and Dietetics, Gazi University, Faculty of Health Sciences, 06490, Ankara, Emek, Turkey
| | - Çiler Özenir
- Department of Nutrition and Dietetics, Kırıkkale University, 71100, Kırıkkale, Merkez, Turkey
| | - Fatih Özoğul
- Department of Seafood Processing Technology, Faculty of Fisheries, Cukurova University, 01330, Balcali, Adana, Turkey
| | - Raffaele Capasso
- Department of Agricultural Sciences, University of Naples Federico II, 80055, Portici, NA, Italy.
| |
Collapse
|
11
|
Basheer AS, Abas F, Othman I, Naidu R. Role of Inflammatory Mediators, Macrophages, and Neutrophils in Glioma Maintenance and Progression: Mechanistic Understanding and Potential Therapeutic Applications. Cancers (Basel) 2021; 13:4226. [PMID: 34439380 PMCID: PMC8393628 DOI: 10.3390/cancers13164226] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 02/07/2023] Open
Abstract
Gliomas are the most common, highly malignant, and deadliest forms of brain tumors. These intra-cranial solid tumors are comprised of both cancerous and non-cancerous cells, which contribute to tumor development, progression, and resistance to the therapeutic regimen. A variety of soluble inflammatory mediators (e.g., cytokines, chemokines, and chemotactic factors) are secreted by these cells, which help in creating an inflammatory microenvironment and contribute to the various stages of cancer development, maintenance, and progression. The major tumor infiltrating immune cells of the tumor microenvironment include TAMs and TANs, which are either recruited peripherally or present as brain-resident macrophages (microglia) and support stroma for cancer cell expansion and invasion. These cells are highly plastic in nature and can be polarized into different phenotypes depending upon different types of stimuli. During neuroinflammation, glioma cells interact with TAMs and TANs, facilitating tumor cell proliferation, survival, and migration. Targeting inflammatory mediators along with the reprogramming of TAMs and TANs could be of great importance in glioma treatment and may delay disease progression. In addition, an inhibition of the key signaling pathways such as NF-κB, JAK/STAT, MAPK, PI3K/Akt/mTOR, and TLRs, which are activated during neuroinflammation and have an oncogenic role in glioblastoma (GBM), can exert more pronounced anti-glioma effects.
Collapse
Affiliation(s)
- Abdul Samad Basheer
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Malaysia; (A.S.B.); (I.O.)
| | - Faridah Abas
- Laboratory of Natural Products, Faculty of Science, University Putra Malaysia (UPM), Serdang 43400, Malaysia;
- Department of Food Science, Faculty of Food Science and Technology, University Putra Malaysia (UPM), Serdang 434000, Malaysia
| | - Iekhsan Othman
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Malaysia; (A.S.B.); (I.O.)
| | - Rakesh Naidu
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Malaysia; (A.S.B.); (I.O.)
| |
Collapse
|
12
|
Dattilo MA, Benzo Y, Herrera LM, Prada JG, Lopez PF, Caruso CM, Lasaga M, García CI, Paz C, Maloberti PM. Regulation and role of Acyl-CoA synthetase 4 in glial cells. J Steroid Biochem Mol Biol 2021; 208:105792. [PMID: 33246155 DOI: 10.1016/j.jsbmb.2020.105792] [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: 08/05/2020] [Revised: 10/23/2020] [Accepted: 11/14/2020] [Indexed: 10/22/2022]
Abstract
Acyl-CoA synthetase 4 (Acsl4), an enzyme involved in arachidonic acid (AA) metabolism, participates in physiological and pathological processes such as steroidogenesis and cancer. The role of Acsl4 in neurons and in nervous system development has also been documented but little is known regarding its functionality in glial cells. In turn, several processes in glial cells, including neurosteroidogenesis, stellation and AA uptake, are regulated by cyclic adenosine monophosphate (cAMP) signal. In this context, the aim of this work was to analyze the expression and functional role of Acsl4 in primary rat astrocyte cultures and in the C6 glioma cell line by chemical inhibition and stable silencing, respectively. Results show that Acsl4 expression was regulated by cAMP in both models and that cAMP stimulation of steroidogenic acute regulatory protein mRNA levels was reduced by Acsl4 inhibition or silencing. Also, Acsl4 inhibition reduced progesterone synthesis stimulated by cAMP and also affected cAMP-induced astrocyte stellation, decreasing process elongation and increasing branching complexity. Similar effects were observed for Acsl4 silencing on cAMP-induced C6 cell morphological shift. Moreover, Acsl4 inhibition and silencing reduced proliferation and migration of both cell types. Acsl4 silencing in C6 cells reduced the capacity for colony proliferation and neurosphere formation, the latter ability also being abolished by Acsl4 inhibition. In sum, this work presents novel evidence of Acsl4 involvement in neurosteroidogenesis and the morphological changes of glial cells promoted by cAMP. Furthermore, Acsl4 participates in migration and proliferation, also affecting cell self-renewal. Altogether, these findings provide insights into Acsl4 functions in glial cells.
Collapse
Affiliation(s)
- Melina A Dattilo
- Universidad de Buenos Aires-CONICET, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina
| | - Yanina Benzo
- Universidad de Buenos Aires-CONICET, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina
| | - Lucia M Herrera
- Universidad de Buenos Aires-CONICET, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - Jesica G Prada
- Universidad de Buenos Aires-CONICET, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - Paula F Lopez
- Universidad de Buenos Aires-CONICET, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - Carla M Caruso
- Universidad de Buenos Aires-CONICET, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Biología Celular e Histología, Buenos Aires, Argentina
| | - Mercedes Lasaga
- Universidad de Buenos Aires-CONICET, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Biología Celular e Histología, Buenos Aires, Argentina
| | - Corina I García
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina; Fundación Instituto Leloir and Instituto de Investigaciones Bioquímicas de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Cristina Paz
- Universidad de Buenos Aires-CONICET, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina
| | - Paula M Maloberti
- Universidad de Buenos Aires-CONICET, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina.
| |
Collapse
|